Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
EMBO Mol Med ; 5(3): 471-81, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23417922

ABSTRACT

We used a novel approach of cytostatically induced leucocyte depletion and subsequent reconstitution with leucocytes deprived of classical (inflammatory/Gr1(hi) ) or non-classical (resident/Gr1(lo) ) monocytes to dissect their differential role in atheroprogression under high-fat diet (HFD). Apolipoprotein E-deficient (Apoe(-/-) ) mice lacking classical but not non-classical monocytes displayed reduced lesion size and macrophage and apoptotic cell content. Conversely, HFD induced a selective expansion of classical monocytes in blood and bone marrow. Increased CXCL1 levels accompanied by higher expression of its receptor CXCR2 on classical monocytes and inhibition of monocytosis by CXCL1-neutralization indicated a preferential role for the CXCL1/CXCR2 axis in mobilizing classical monocytes during hypercholesterolemia. Studies correlating circulating and lesional classical monocytes in gene-deficient Apoe(-/-) mice, adoptive transfer of gene-deficient cells and pharmacological modulation during intravital microscopy of the carotid artery revealed a crucial function of CCR1 and CCR5 but not CCR2 or CX3 CR1 in classical monocyte recruitment to atherosclerotic vessels. Collectively, these data establish the impact of classical monocytes on atheroprogression, identify a sequential role of CXCL1 in their mobilization and CCR1/CCR5 in their recruitment.


Subject(s)
Aortic Diseases/immunology , Atherosclerosis/immunology , Carotid Artery Diseases/immunology , Chemotaxis, Leukocyte , Monocytes/immunology , Receptors, Chemokine/metabolism , Adoptive Transfer , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/etiology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Carotid Arteries/immunology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/etiology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Chemokine CXCL1/metabolism , Diet, High-Fat , Disease Models, Animal , Disease Progression , Leukocyte Count , Leukocyte Reduction Procedures , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/classification , Monocytes/transplantation , Receptors, CCR1/metabolism , Receptors, CCR5/metabolism , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Receptors, Interleukin-8B/metabolism , Signal Transduction , Time Factors
2.
Arterioscler Thromb Vasc Biol ; 32(7): 1613-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22556330

ABSTRACT

OBJECTIVE: Inflammatory leukocyte accumulation drives atherosclerosis. Although monocytes/macrophages and polymorphonuclear neutrophilic leukocytes (PMN) contribute to lesion formation, sequelae of myeloproliferative disease remain to be elucidated. METHODS AND RESULTS: We used mice deficient in interferon regulatory factor 8 (IRF8(-/-)) in hematopoietic cells that develop a chronic myelogenous leukemia-like phenotype. Apolipoprotein E-deficient mice reconstituted with IRF8(-/-) or IRF8(-/-) apolipoprotein E-deficient bone marrow displayed an exacerbated atherosclerotic lesion formation compared with controls. The chronic myelogenous leukemia-like phenotype in mice with IRF8(-/-) bone marrow, reflected by an expansion of PMN in the circulation, was associated with an increased lesional accumulation and apoptosis of PMN, and enlarged necrotic cores. IRF8(-/-) compared with IRF8(+/+) PMN displayed unaffected reactive oxygen species formation and discharge of PMN granule components. In contrast, accumulating in equal numbers at sites of inflammation, IRF8(-/-) macrophages were defective in efferocytosis, lipid uptake, and interleukin-10 cytokine production. Importantly, depletion of PMN in low-density lipoprotein receptor or apolipoprotein E-deficient mice with IRF8(-/-) or IRF8(-/-) apolipoprotein E-deficient bone marrow abrogated increased lesion formation. CONCLUSIONS: These findings indicate that a chronic myelogenous leukemia-like phenotype contributes to accelerated atherosclerosis in mice. Among proatherosclerotic effects of other cell types, this, in part, is linked to an expansion of functionally intact PMN.


Subject(s)
Atherosclerosis/etiology , Interferon Regulatory Factors/physiology , Animals , Apolipoproteins E/physiology , Apoptosis , Bone Marrow Transplantation , Capillary Permeability , Female , Interleukin-10/biosynthesis , Macrophages/physiology , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Peroxidase/physiology , Reactive Oxygen Species/metabolism , Receptors, LDL/physiology
3.
J Immunol Methods ; 381(1-2): 9-13, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22537800

ABSTRACT

BACKGROUND: Accumulating evidence intensively advises circulating endothelial progenitor cells (EPCs) and monocyte subsets as surrogate cellular biomarkers in cardiovascular and cancer disease. However, a general standard on their quantification is still elusive, thus precluding a routine monitoring and comparative interpretation of clinical studies. OBJECTIVE: We intend to develop an advanced and express flow cytometric protocol for proper ex vivo quantification of monocyte subsets and EPCs in human blood. METHODS: We employ now lyse/no-wash procedure and bead-based determination of absolute cell counts. We use three-color antibody panels at appropriate compensation. Analysis of rare events and low antigen expression in the EPC experiment is strengthening by sequential gating with exclusion of dead cells, as well as by matching high-intensity fluorochromes to low-density markers and by implementing the fluorescence-minus-one control. RESULTS: Analysis of peripheral blood of ten healthy donors revealed median (IQR) value of 1.88 (1.35-2.85) viable CD45(dim)CD34)VEGFR2+ EPCs per microliter. Analysis of monocytes revealed 329.5 (264.5-374.8), 16.0 (8.0-22.2) and 26.5 (19.8-36.3) cells per microliter for classical CD14++(high))CD16⁻, intermediate CD14++CD16+(mid) and non-classical CD14+(low))CD16++ monocytes. CONCLUSION: Our current protocol provides quantitative information under a simple gating logic while using commonly accepted fluorochromes. This assay is therefore highly adapted for routine use.


Subject(s)
Endothelial Cells/cytology , Flow Cytometry/methods , Monocytes/cytology , Stem Cells/cytology , Adult , Antigens, CD34/blood , Blood Cell Count/instrumentation , Blood Cell Count/methods , Endothelial Cells/metabolism , Female , Humans , Leukocyte Common Antigens/blood , Male , Monocytes/metabolism , Reproducibility of Results , Stem Cells/metabolism , Vascular Endothelial Growth Factor Receptor-2/blood , Young Adult
4.
J Cardiothorac Surg ; 7: 2, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22214418

ABSTRACT

BACKGROUND: Cardiovascular disease has been linked to endothelial progenitor cell (EPC) depletion and functional impairment in atherosclerosis and aortic stenosis. EPCs may play a pivotal role in vascular grafting. However, the EPC depletion in coronary artery bypass grafting (CABG) patients has not been compared to coronary artery disease-free valvular replacement patients with aortic stenosis. METHODS: We aimed to assess the basal number of CD34+/KDR+ and CD34+/CD144+ cells in CABG patients, compared to aortic stenosis valvular replacement patients. 100 patients (51 CABG and 49 valvular surgery ones) were included in the present study. All CABG or valvular patients had angiographic demonstration of the presence or the absence of coronary artery disease, respectively. Numbers of CD34+/KDR+ and CD34+/CD144+ were assessed by flow cytometry of pre-surgical blood samples. RESULTS: We found a lower number of CD34+/CD144+ cells in CABG patients compared to valvular patients (0.21 ± 0.03% vs. 0.47 ± 0.08%), and this difference remained statistically significant after the P was adjusted for multiple comparisons (P = 0.01428). Both groups had more EPCs than healthy controls. CONCLUSIONS: Pre-surgical CD34+/CD144+ numbers are decreased in CABG patients, compared to valvular patients with absence of coronary disease.


Subject(s)
Antigens, CD34/immunology , Antigens, CD/immunology , Aortic Valve Stenosis/complications , Cadherins/immunology , Coronary Artery Disease/immunology , Coronary Vessels/pathology , Endothelium, Vascular/immunology , Aged , Antigens, CD/blood , Antigens, CD34/blood , Aortic Valve Stenosis/immunology , Aortic Valve Stenosis/surgery , Cadherins/blood , Cell Count , Coronary Angiography , Coronary Artery Bypass , Coronary Artery Disease/complications , Coronary Artery Disease/surgery , Coronary Vessels/immunology , Coronary Vessels/surgery , Endothelium, Vascular/pathology , Female , Flow Cytometry , Heart Valve Prosthesis Implantation , Humans , Male , Middle Aged , Preoperative Period
5.
J Am Coll Cardiol ; 58(23): 2415-23, 2011 Nov 29.
Article in English | MEDLINE | ID: mdl-22115649

ABSTRACT

OBJECTIVES: Here we assess the intrinsic functions of the chemokine receptor CXCR4 in remodeling after myocardial infarction (MI) using Cxcr4 heterozygous (Cxcr4(+/-)) mice. BACKGROUND: Myocardial necrosis triggers complex remodeling and inflammatory changes. The chemokine CXCL12 has been implicated in protection and therapeutic regeneration after MI through recruiting angiogenic outgrowth cells, improving neovascularization and cardiac function, but the endogenous role of its receptor CXCR4 is unknown. METHODS: MI was induced by ligation of the left descending artery. Langendoff perfusion, echocardiography, quantitative immunohistochemistry, flow cytometry, angiogenesis assays, and cardiomyocyte analysis were performed. RESULTS: After 4 weeks, infarct size was reduced in Cxcr4(+/-) mice compared with wild-type mice and in respective bone marrow chimeras compared with controls. This was associated with altered inflammatory cell recruitment, decreased neutrophil content, delayed monocyte infiltration, and a predominance of Gr1(low) over classic Gr1(high) monocytes. Basal coronary flow and its recovery after MI were impaired in Cxcr4(+/-)mice, paralleled by reduced angiogenesis, myocardial vessel density, and endothelial cell count. Notably, no differences in cardiac function were seen in Cxcr4(+/-)mice compared with wild-type mice. Despite defective angiogenesis, Cxcr4(+/-) mouse hearts showed no difference in CXCL12, vascular endothelial growth factor or apoptosis-related gene expression. Electron microscopy revealed lipofuscin-like lipid accumulation in Cxcr4(+/-) mouse hearts and analysis of lipid extracts detected high levels of phosphatidylserine, which protect cardiomyocytes from hypoxic stress in vitro. CONCLUSIONS: CXCR4 plays a crucial role in endogenous remodeling processes after MI, contributing to inflammatory/progenitor cell recruitment and neovascularization, whereas its deficiency limits infarct size and causes adaptation to hypoxic stress. This should be carefully scrutinized when devising therapeutic strategies involving the CXCL12/CXCR4 axis.


Subject(s)
Chemokine CXCL12/genetics , DNA/genetics , Gene Expression Regulation , Myocardial Infarction/genetics , Receptors, CXCR4/genetics , Animals , Apoptosis , Chemokine CXCL12/biosynthesis , Disease Models, Animal , Mice , Mice, Inbred C57BL , Microscopy, Electron , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Receptors, CXCR4/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
6.
J Clin Invest ; 121(7): 2898-910, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21633167

ABSTRACT

Immune mechanisms are known to control the pathogenesis of atherosclerosis. However, the exact role of DCs, which are essential for priming of immune responses, remains elusive. We have shown here that the DC-derived chemokine CCL17 is present in advanced human and mouse atherosclerosis and that CCL17+ DCs accumulate in atherosclerotic lesions. In atherosclerosis-prone mice, Ccl17 deficiency entailed a reduction of atherosclerosis, which was dependent on Tregs. Expression of CCL17 by DCs limited the expansion of Tregs by restricting their maintenance and precipitated atherosclerosis in a mechanism conferred by T cells. Conversely, a blocking antibody specific for CCL17 expanded Tregs and reduced atheroprogression. Our data identify DC-derived CCL17 as a central regulator of Treg homeostasis, implicate DCs and their effector functions in atherogenesis, and suggest that CCL17 might be a target for vascular therapy.


Subject(s)
Atherosclerosis/immunology , Chemokine CCL17/immunology , Dendritic Cells/immunology , Homeostasis , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow Transplantation , Cell Movement , Chemokine CCL17/genetics , Humans , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocyte Subsets/metabolism
7.
J Cell Mol Med ; 15(3): 668-78, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20178462

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a pleiotropic inflammatory cytokine that was recently identified as a non-cognate ligand of the CXC-family chemokine receptors 2 and 4 (CXCR2 and CXCR4). MIF is expressed and secreted from endothelial cells (ECs) following atherogenic stimulation, exhibits chemokine-like properties and promotes the recruitment of leucocytes to atherogenic endothelium. CXCR4 expressed on endothelial progenitor cells (EPCs) and EC-derived CXCL12, the cognate ligand of CXCR4, have been demonstrated to be critical when EPCs are recruited to ischemic tissues. Here we studied whether hypoxic stimulation triggers MIF secretion from ECs and whether the MIF/CXCR4 axis contributes to EPC recruitment. Exposure of human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAoECs) to 1% hypoxia led to the specific release of substantial amounts of MIF. Hypoxia-induced MIF release followed a biphasic behaviour. MIF secretion in the first phase peaked at 60 min. and was inhibited by glyburide, indicating that this MIF pool was secreted by a non-classical mechanism and originated from pre-formed MIF stores. Early hypoxia-triggered MIF secretion was not inhibited by cycloheximide and echinomycin, inhibitors of general and hypoxia-inducible factor (HIF)-1α-induced protein synthesis, respectively. A second phase of MIF secretion peaked around 8 hrs and was likely due to HIF-1α-induced de novo synthesis of MIF. To functionally investigate the role of hypoxia-inducible secreted MIF on the recruitment of EPCs, we subjected human AcLDL(+) KDR(+) CD31(+) EPCs to a chemotactic MIF gradient. MIF potently promoted EPC chemotaxis in a dose-dependent bell-shaped manner (peak: 10 ng/ml MIF). Importantly, EPC migration was induced by supernatants of hypoxia-conditioned HUVECs, an effect that was completely abrogated by anti-MIF- or anti-CXCR4-antibodies. Thus, hypoxia-induced MIF secretion from ECs might play an important role in the recruitment and migration of EPCs to hypoxic tissues such as after ischemia-induced myocardial damage.


Subject(s)
Endothelial Cells/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Stem Cells/metabolism , Cell Hypoxia , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Chemotaxis/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme-Linked Immunosorbent Assay , Glyburide/pharmacology , Humans , Macrophage Migration-Inhibitory Factors/genetics , Oxygen/pharmacology , Receptors, CXCR4/metabolism , Recombinant Proteins/pharmacology , Stem Cells/drug effects , Time Factors
8.
Eur Heart J ; 32(4): 476-84, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21036775

ABSTRACT

AIMS: We sought to determine whether circulating vascular progenitor cells, such as endothelial progenitor cells (EPCs) or smooth muscle progenitor cells (SPCs), were associated with the severity of cardiac allograft vasculopathy (CAV). METHODS AND RESULTS: CD34(+)CD140b(+) SPCs and CD34(+)KDR(+) EPCs were measured in the peripheral circulation of 187 adult heart transplant recipients by flow cytometry. Cardiac allograft vasculopathy was quantified by angiography using a CAV-specific scoring system. Cardiac allograft vasculopathy was present in 84 patients (44.7%) and was classified as mild in 59 and severe in 25 cases. Circulating SPCs were more frequently detectable in CAV patients than in patients without CAV. The number of CD34(+)CD140b(+) cells showed a stepwise increase in patients with moderate and severe CAV. Smooth muscle progenitor cell counts were higher in patients with coronary stent implant compared with unstented patients with CAV. In contrast, peripheral CD34(+)KDR(+) EPC counts were not changed in CAV patients. Plasma CXCL12 levels correlated with the degree of CAV and SPC counts. None of the different immunosuppressive drug regimes was related to the SPC count or the CXCL12 levels. A multivariate regression analysis revealed that the SPC count was independently associated with the presence of CAV. CONCLUSION: Circulating SPCs, but not EPCs, and plasma CXCL12 concentrations are elevated in CAV patients, indicating that they play prominent roles in transplant arteriosclerosis.


Subject(s)
Antigens, CD34/metabolism , Chemokine CXCL12/metabolism , Graft Rejection/etiology , Heart Transplantation/pathology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stem Cells/pathology , Adult , Aged , Cell Proliferation , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Female , Flow Cytometry , Graft Rejection/pathology , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Transplantation, Homologous
9.
J Immunol ; 185(9): 5326-35, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20881193

ABSTRACT

Dendritic cells (DCs) in lymphoid tissue comprise conventional DCs (cDCs) and plasmacytoid DCs (pDCs) that develop from common DC progenitors (CDPs). CDPs are Flt3(+)c-kit(int)M-CSFR(+) and reside in bone marrow. In this study, we describe a two-step culture system that recapitulates DC development from c-kit(hi)Flt3(-/lo) multipotent progenitors (MPPs) into CDPs and further into cDC and pDC subsets. MPPs and CDPs are amplified in vitro with Flt3 ligand, stem cell factor, hyper-IL-6, and insulin-like growth factor-1. The four-factor mixture readily induces self-renewal of MPPs and their progression into CDPs and has no self-renewal activity on CDPs. The amplified CDPs respond to all known DC poietins and generate all lymphoid tissue DCs in vivo and in vitro. Additionally, in vitro CDPs recapitulate the cell surface marker and gene expression profile of in vivo CDPs and possess a DC-primed transcription profile. TGF-ß1 impacts on CDPs and directs their differentiation toward cDCs. Genome-wide gene expression profiling of TGF-ß1-induced genes identified instructive transcription factors for cDC subset specification, such as IFN regulatory factor-4 and RelB. TGF-ß1 also induced the transcription factor inhibitor of differentiation/DNA binding 2 that suppresses pDC development. Thus, TGF-ß1 directs CDP differentiation into cDCs by inducing both cDC instructive factors and pDC inhibitory factors.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Hematopoietic Stem Cells/cytology , Transforming Growth Factor beta1/immunology , Animals , Cell Separation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Gene Expression , Gene Expression Profiling , Gene Knock-In Techniques , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Transforming Growth Factor beta1/metabolism
10.
Circulation ; 122(5): 495-506, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20644015

ABSTRACT

BACKGROUND: Angiogenic early outgrowth cells (EOCs) have been reported to contribute to endothelial regeneration and to limit neointima formation after vascular injury. Vascular pathologies comprise platelet activation and concomitant generation of platelet microparticles (PMPs). We hypothesized that PMPs may interact with EOCs in the context of vascular injury and modulate their regenerative potential. METHODS AND RESULTS: Using flow cytometry, confocal microscopy, and scanning electron microscopy, we demonstrated the binding of thrombin/collagen-induced PMPs to EOCs with subsequent membrane assimilation and incorporation. This interaction promoted phenotypic alterations of EOCs with increased expression of endothelial cell markers and transfer of the chemokine receptor CXCR4 to EOCs with enhanced responsiveness to its ligand CXCL12/SDF-1alpha. In addition, PMPs augmented the adhesion of EOCs to extracellular matrix components and to the injured vessel wall and accelerated cytoskeletal reorganization and migration of EOCs. PMPs induced changes in the EOC secretome toward a more proangiogenic profile and amplified the EOC-mediated induction of proliferation, migration, and capillary tube formation by mature endothelial cells. Compared with untreated EOCs, the injection of PMP-treated EOCs resulted in accelerated reendothelialization after arterial denudation injury in athymic nude mice, whereas the EOC-mediated reduction of neointima formation remained unchanged. CONCLUSIONS: Our data provide evidence that PMPs can boost the potential of EOCs to restore endothelial integrity after vascular injury. Major mechanisms involve the enhancement of EOC recruitment, migration, differentiation, and release of proangiogenic factors.


Subject(s)
Blood Platelets/physiology , Carotid Artery Injuries/physiopathology , Cell-Derived Microparticles/physiology , Endothelial Cells/physiology , Neovascularization, Physiologic/physiology , Animals , Biomarkers/metabolism , Blood Platelets/cytology , Carotid Arteries/pathology , Carotid Arteries/physiology , Carotid Artery Injuries/pathology , Cell Communication/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cell Movement/physiology , Cells, Cultured , Chemokine CXCL12/metabolism , Cytoskeleton/physiology , Disease Models, Animal , Endothelial Cells/cytology , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Receptors, CXCR4/metabolism , Regeneration/physiology , Umbilical Veins/cytology
12.
Circulation ; 121(2): 315-24, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20048211

ABSTRACT

BACKGROUND: Recent work has revealed an essential involvement of soluble CD40 ligand (sCD40L) in inflammation and atherosclerosis. We investigated whether sCD40L functionally affects peripheral blood-derived angiogenic early outgrowth cells (EOCs) and neointimal remodeling after arterial injury. METHODS AND RESULTS: Besides myeloid and endothelial markers, cultured human EOCs strongly expressed CD40 mRNA and protein. EOC adhesion to fibronectin, fibrinogen, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 under flow conditions, as well as their transmigration toward stromal cell-derived factor-1alpha, was dose-dependently reduced after preincubation with recombinant human sCD40L for 24 hours. Integrin expression was unaffected by sCD40L, implying that integrin adhesiveness was attenuated. Surface-immobilized CD40L supported much lower adhesion of EOCs than fibronectin. Treatment of EOCs with sCD40L increased superoxide anion production and decreased viability and proliferation. Notably, CD40(-/-) mice displayed reduced neointima and improved re-endothelialization after carotid wire injury compared with wild-type mice, and therapeutic infusion of control EOCs but not EOCs pretreated with sCD40L attenuated neointimal growth after wire injury in nude mice. Furthermore, neointimal growth was more markedly diminished by infusion of spleen-derived CD40(-/-) mouse EOCs than by that of wild-type EOCs. Preincubation of wild-type EOCs but not CD40(-/-) EOCs with sCD40L before their infusion markedly aggravated neointimal formation. Treatment with sCD40L attenuated luminal incorporation of EOCs and accelerated neointimal progression. CONCLUSIONS: Endothelial dysfunction due to persistently elevated plasma levels of sCD40L may be attributable to an impairment of EOC function. Hence, in the context of arterial injury, therapeutic blockade of sCD40L may provide a novel strategy for accelerating endothelial regeneration and attenuating neointimal remodeling.


Subject(s)
CD40 Ligand/pharmacology , Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Animals , CD40 Ligand/therapeutic use , Carotid Artery Injuries/therapy , Cell Adhesion , Cell Movement , Cell Proliferation , Cell Survival , Cell Transplantation , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/transplantation , Humans , Mice , Regeneration/drug effects , Superoxides
13.
Sci Signal ; 2(100): ra81, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19996457

ABSTRACT

Apoptosis is a pivotal process in embryogenesis and postnatal cell homeostasis and involves the shedding of membranous microvesicles termed apoptotic bodies. In response to tissue damage, the CXC chemokine CXCL12 and its receptor CXCR4 counteract apoptosis and recruit progenitor cells. Here, we show that endothelial cell-derived apoptotic bodies are generated during atherosclerosis and convey paracrine alarm signals to recipient vascular cells that trigger the production of CXCL12. CXCL12 production was mediated by microRNA-126 (miR-126), which was enriched in apoptotic bodies and repressed the function of regulator of G protein (heterotrimeric guanosine triphosphate-binding protein) signaling 16, an inhibitor of G protein-coupled receptor (GPCR) signaling. This enabled CXCR4, a GPCR, to trigger an autoregulatory feedback loop that increased the production of CXCL12. Administration of apoptotic bodies or miR-126 limited atherosclerosis, promoted the incorporation of Sca-1+ progenitor cells, and conferred features of plaque stability on different mouse models of atherosclerosis. This study highlights functions of microRNAs in health and disease that may extend to the recruitment of progenitor cells during other forms of tissue repair or homeostasis.


Subject(s)
Apoptosis/physiology , Chemokine CXCL12/physiology , Endothelium, Vascular/cytology , MicroRNAs/administration & dosage , Animals , Atherosclerosis/pathology , Base Sequence , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , DNA Primers , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Mice , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
J Cell Mol Med ; 13(11-12): 4293-303, 2009.
Article in English | MEDLINE | ID: mdl-19900213

ABSTRACT

Atherosclerosis is a chronic inflammatory disease wherein the infiltration of myeloid cells of the vessel wall is a hallmark event. Lymphocytes, platelets and endothelial cells stand out as prominent suspects being involved in atherosclerosis. However, recent advances suggest a crucial role for myeloid leucocytes, specifically monocyte subsets, neutrophils, dendritic cells and endothelial progenitor cells. These cell types are not just rapidly recruited or already reside in the vascular wall, but also initiate and perpetuate core mechanisms in plaque formation and destabilization. Hyperlipidaemia is an independent risk factor for atherosclerosis. Herein, hyperlipidaemia skews myeloid cell haemostasis, phenotype and transcriptional regulation of pro-inflammatory factors ultimately promoting myeloid cell extravasation and atherosclerosis. We here review the role of myeloid cells in atherosclerosis as well as the effects of hyperlipidaemia on these cells.


Subject(s)
Atherosclerosis/pathology , Atherosclerosis/physiopathology , Hyperlipidemias/pathology , Hyperlipidemias/physiopathology , Myeloid Cells/pathology , Animals , Humans , Risk Factors
15.
Cytometry A ; 75(10): 848-53, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19739088

ABSTRACT

Circulating adult CD34(+)VEGFR2(+) endothelial progenitor cells (EPCs) have been shown to differentiate into endothelial cells, thus contributing to vascular homeostasis. Furthermore, a subset of circulating CD14(+) monocytes coexpresses CD16 together with the angiopoietin receptor Tie2 and has been functionally implicated in tumor angiogenesis. However, clinically applicable protocols for flow cytometric quantification of EPCs and Tie2(+) monocytes in peripheral blood and a consensus on reference values remain elusive. The number of Tie2(+)CD14(+)CD16(mid) angiogenic monocytes and CD34(+)VEGFR2(+)CD45(low/-) EPCs was assessed in the peripheral venous blood of patients with stable coronary artery disease by three-color flow cytometry using specific monoclonal antibodies conjugated to PerCP, PE, PE-Cy7, APC, and APC-Cy7. Scatter multigating with exclusion of dead cells was performed to dissect complex mononuclear cell populations. This analysis was further refined by matching bright fluorochromes (PE-Cy7, PE, APC) with dimly expressed markers (CD34, VEGFR2, Tie2), by automatic compensation for minimizing fluorescence spillover and by using fluorescence-minus-one (FMO) controls to determine positive/negative boundaries. Presuming a Gaussian distribution, we obtained average values (mean +/- SD) of 1.45 +/- 1.29% for Tie2(+)CD14(+)CD16(mid) monocytes (n = 11, range: 0.12-3.64%) and 0.019 +/- 0.013% for CD34(+)VEGFR2(+)CD45(low/-) EPCs (n = 17, range: 0.003-0.042%). The intra- and inter-assay variability was 1.6% and 4.5%, respectively. We have optimized a fast and sensitive assay for the flow cytometric quantification of circulating angiogenic monocytes and EPCs in cardiovascular medicine. This protocol may represent a basis for standardized analysis and monitoring of these cell subsets to define their normal range and prognostic/diagnostic value in clinical use.


Subject(s)
Endothelial Cells/cytology , Flow Cytometry/methods , Monocytes/cytology , Neovascularization, Physiologic , Stem Cells/cytology , Aged , Endothelial Cells/metabolism , Female , Humans , Leukocyte Common Antigens/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Receptor, TIE-2/metabolism , Receptors, IgG/metabolism , Stem Cells/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Nephrol Dial Transplant ; 24(1): 321-4, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18840896

ABSTRACT

BACKGROUND: Altered IgA1 galactosylation is involved in the pathogenesis of IgA nephropathy (IgAN). The galactosyltransferase core-1 beta3-galactosyltransferase-1 (C1GALT1) and its chaperone cosmc are specifically required for O-galactosylation of the IgA1 hinge region. Mutations in the cosmc gene result in a secondary loss of function of C1GALT1 with subsequent undergalactosylation of glycoproteins. Mosaic mutations of cosmc have been shown to result in autoimmune disease. We hypothesized that cosmc mutations might contribute to the altered IgA1 galactosylation in IgAN patients. METHODS: We studied cosmc gene sequences in genomic DNA obtained from male patients with biopsy-proven sporadic (n = 33) and familial IgAN (n = 6 patients from different families). To account for a potential mosaicism we sequenced cosmc in 10 different peripheral blood mononuclear cell DNA clones of every patient. To specifically assess potential mosaic mutations in IgA-producing cells, cosmc mutations were also analysed in DNA isolated from CD20+ B-lymphocytes from three male IgAN patients. RESULTS: Despite our extensive genomic analysis, the data revealed no functionally relevant cosmc gene variants in sporadic or familial IgAN cases. A cosmc gene polymorphism, rs17261572, was identified in these IgAN patients in a similar frequency as previously reported in healthy adults. A functional consequence of this polymorphism has not yet been determined. CONCLUSION: Although decreased C1GALT1 activity has been implicated in the IgAN pathogenesis and cosmc chaperone mutations can cause autoimmune disease, our data provide no evidence for a relevant role of cosmc gene mutations in European patients with sporadic or familial IgAN.


Subject(s)
Glomerulonephritis, IGA/genetics , Molecular Chaperones/genetics , Mutation , Adult , Aged , Child , Chromosomes, Human, X/genetics , DNA Mutational Analysis , Europe , Gene Frequency , Humans , Male , Middle Aged , Mosaicism , Polymorphism, Single Nucleotide , Young Adult
17.
Curr Opin Lipidol ; 19(5): 491-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18769230

ABSTRACT

PURPOSE OF REVIEW: To discuss crucial cues (chemokines, adhesion molecules and pharmacological means) that guide and control the context-specific mobilization, recruitment and fate of circulating progenitor cells in arterial repair and plaque stability. RECENT FINDINGS: The mobilization and recruitment of bone marrow derived or resident progenitor cells giving rise to smooth muscle cells have been implicated in accelerated forms of primary plaque formation and neointimal hyperplasia after arterial injury. By contrast, convincing evidence has emerged that the arterial homing of endothelial progenitor cells contributes to endothelial recovery and thereby limits neointimal growth after endothelial denudation. In the chronic context of primary atherosclerosis, plaque progression and destabilization, a more complex picture has become apparent. Clinically, the number and function of endothelial progenitor cells have been linked with an improved endothelial function or regeneration and have been frequently inversely correlated with cardiovascular risk (factors). In animal models, however, the injection of bone marrow cells or endothelial progenitor cells, as well as the application of stem-cell mobilizing factors, have been associated with an exacerbation of atherosclerosis and unstable plaque phenotype, whereas the contribution of smooth muscle progenitors to primary atherosclerosis appears to be more confined to supporting plaque stability. SUMMARY: Considering the balance between distinct circulating vascular progenitor cells and identifying mechanisms for selective control of their mobilization and homing appears crucial to improve prediction and to directly modulate endogenous vascular remodeling processes.


Subject(s)
Atherosclerosis/pathology , Endothelial Cells/cytology , Stem Cells/cytology , Vascular Diseases/pathology , Animals , Atherosclerosis/physiopathology , Atherosclerosis/surgery , Cell Movement , Endothelial Cells/physiology , Endothelial Cells/transplantation , Humans , Models, Biological , Neovascularization, Physiologic , Stem Cell Transplantation/methods , Stem Cells/physiology , Vascular Diseases/physiopathology , Vascular Diseases/surgery
18.
Pharmacol Res ; 58(2): 148-51, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18722530

ABSTRACT

Postnatal bone marrow contains a subtype of unique progenitor cells that have the capacity to differentiate into functional endothelial cells. Hence, these cells have been termed endothelial progenitor cells (EPCs). In general, circulating EPCs were characterized by the expression of CD133, CD34 and the vascular endothelial growth factor receptor-2 (VEGFR2). Recent data have additionally described some CD14(+/low) myeloid subsets as functional endothelial precursors. Convincing evidence in vivo has further emerged that the vascular homing of EPCs contributes to endothelial regeneration thereby limiting neointimal hyperplasia after arterial injury. However, in the context of primary atherosclerosis, plaque progression and destabilization, injection of EPCs as well as application of stem-cell mobilizing factors have been shown to correlate with conversion to unstable plaque phenotype. Clinically, the number and function of EPCs have been positively linked with an improved endothelial function or regeneration but frequently inversely correlated with cardiovascular risk (factors). Thus, considering the dual contribution of EPCs in vascular repair and remodeling in primary atherosclerosis versus arterial injury and identifying mechanisms for selective control of their recruitment appears crucial to improve prediction and to directly modulate endogenous vascular homeostasis.


Subject(s)
Blood Vessels/physiology , Endothelial Cells/cytology , Regeneration/physiology , Stem Cells/physiology , Animals , Atherosclerosis/etiology , Atherosclerosis/physiopathology , Cell Movement , Hematopoietic Stem Cell Mobilization , Humans , Neovascularization, Physiologic
19.
Biol Chem ; 389(7): 837-44, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18627306

ABSTRACT

The mobilization and recruitment of bone marrow-derived, circulating or tissue resident progenitor cells giving rise to smooth muscle-like cells have been implicated in neointima hyperplasia after arterial injury and in accelerated forms of arterial lesion formation, e.g., transplant arteriopathy or graft vasculopathy. By contrast, convincing evidence has emerged that the vascular homing of endothelial progenitor cells (EPCs) contributes to endothelial recovery, thus limiting neointima formation after arterial injury. In the chronic context of primary atherosclerosis, plaque progression and destabilization, a more complex picture has become apparent. In patients with coronary artery disease, the number and function of EPCs have been linked with an improved endothelial function or regeneration, but have been inversely correlated with cardiovascular risk. In animal models, however, the injection of bone marrow cells or EPCs, or the application of stem-cell mobilizing factors, have been associated with an exacerbation of atherosclerosis and unstable plaque phenotypes, whereas the contribution of bone marrow-derived smooth muscle progenitors to primary atherosclerosis appears to be rather confined. Here, we discuss crucial biochemical cues, namely chemokines, adhesion molecules, growth factors and pharmacological means that guide and control the context-specific mobilization, recruitment and fate of vascular progenitor cells in arterial remodeling during atherosclerosis.


Subject(s)
Adult Stem Cells/pathology , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Blood Vessels/pathology , Blood Vessels/physiopathology , Endothelial Cells/pathology , Animals , Cell Movement , Humans , Myocytes, Smooth Muscle/pathology
20.
J Immunol Methods ; 335(1-2): 21-7, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18402976

ABSTRACT

Although determination of circulating endothelial progenitor cell (EPC) in peripheral blood by flow cytometry is an emerging marker for cardiovascular medicine, a common standardized protocol is still not available, due to the low numbers achieved in peripheral blood. In the present paper we describe a novel technique for EPC quantification as CD34+/CD144+/CD3- cells within the lymphocyte gate, which increases the percentages of EPC positivity described before and also offers high intra-assay reproducibility. These improvements are based on a gating strategy for big-sized lymphocytes, smooth fixation and cytometric clearance of CD3+ lymphocytes (T-cells). This last procedure is able to increase intra-assay Pearson's correlation from 0.8517 to 0.8908. Therefore, the technical setting described here offers a high-performance and clinically oriented EPC determination strategy in human peripheral blood.


Subject(s)
Antigens, CD34/analysis , Antigens, CD/analysis , CD3 Complex/analysis , Cadherins/analysis , Endothelial Cells/immunology , Flow Cytometry , Immunophenotyping/methods , Lymphocyte Subsets/immunology , Stem Cells/immunology , Cell Adhesion , Cell Differentiation , Cell Shape , Cells, Cultured , Female , Humans , Male , Middle Aged , Phenotype , Reproducibility of Results , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...