Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 190
Filter
1.
Int J Mol Sci ; 22(17)2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34502520

ABSTRACT

In our previous study, we introduced the platelet endothelial cell adhesion molecule 1 (PECAM-1)/thrombus ratio, which is a parameter indicating the proportion of PECAM-1 in laser-induced thrombi in mice. Because PECAM-1 is an antithrombotic molecule, the higher the PECAM-1/thrombus ratio, the less activated the platelets. In this study, we used an extracorporeal model of thrombosis (flow chamber model) to verify its usefulness in the assessment of the PECAM-1/thrombus ratio in animal and human studies. Using the lipopolysaccharide (LPS)-induced inflammation model, we also evaluated whether the PECAM-1/thrombus ratio determined in the flow chamber (without endothelium) differed from that calculated in laser-induced thrombosis (with endothelium). We observed that acetylsalicylic acid (ASA) decreased the area of the thrombus while increasing the PECAM-1/thrombus ratio in healthy mice and humans in a dose-dependent manner. In LPS-treated mice, the PECAM-1/thrombus ratio decreased as the dose of ASA increased in both thrombosis models, but the direction of change in the thrombus area was inconsistent. Our study demonstrates that the PECAM-1/thrombus ratio can more accurately describe the platelet activation status than commonly used parameters such as the thrombus area, and, hence, it can be used in both human and animal studies.


Subject(s)
Platelet Activation/physiology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Animals , Aspirin/analysis , Blood Platelets/metabolism , Blood Platelets/physiology , Cell Adhesion , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Female , Healthy Volunteers , Humans , Inflammation , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Thrombosis/metabolism
2.
J Biol Chem ; 292(48): 19639-19655, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28974577

ABSTRACT

Platelet endothelial cell adhesion molecule 1 (PECAM-1) is a cell adhesion protein involved in the regulation of cell adhesion and migration. Interestingly, several PECAM-1-deficient hematopoietic cells exhibit impaired chemotactic responses to stromal cell-derived factor 1 (SDF-1), a chemokine essential for B lymphopoiesis and bone marrow myelopoiesis. However, whether PECAM-1 is involved in SDF-1-regulated chemotaxis is unknown. We report here that SDF-1 induces tyrosine phosphorylation of PECAM-1 at its immunoreceptor tyrosine-based inhibition motifs in several hematopoietic cell lines via the Src family kinase Lyn, Bruton's tyrosine kinase, and JAK2 and that inhibition of these kinases reduced chemotaxis. Overexpression and knockdown of PECAM-1 enhanced and down-regulated, respectively, SDF-1-induced Gαi-dependent activation of the PI3K/Akt/mTORC1 pathway and small GTPase Rap1 in hematopoietic 32Dcl3 cells, and these changes in activation correlated with chemotaxis. Furthermore, pharmacological or genetic inhibition of the PI3K/Akt/mTORC1 pathway or Rap1, respectively, revealed that these pathways are independently activated and required for SDF-1-induced chemotaxis. When coexpressed in 293T cells, PECAM-1 physically associated with the SDF-1 receptor CXCR4. Moreover, PECAM-1 overexpression and knockdown reduced and enhanced SDF-1-induced endocytosis of CXCR4, respectively. Furthermore, when expressed in 32Dcl3 cells, an endocytosis-defective CXCR4 mutant, CXCR4-S324A/S325A, could activate the PI3K/Akt/mTORC1 pathway as well as Rap1 and induce chemotaxis in a manner similar to PECAM-1 overexpression. These findings suggest that PECAM-1 enhances SDF-1-induced chemotaxis by augmenting and prolonging activation of the PI3K/Akt/mTORC1 pathway and Rap1 and that PECAM-1, at least partly, exerts its activity by inhibiting SDF-1-induced internalization of CXCR4.


Subject(s)
Chemokine CXCL12/physiology , Leukocytes/physiology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Animals , Bone Marrow Cells/metabolism , Cell Line , Mice , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/chemistry , Protein-Tyrosine Kinases/metabolism , Receptors, CXCR4/metabolism , Tyrosine/metabolism
3.
Am J Pathol ; 186(5): 1387-402, 2016 05.
Article in English | MEDLINE | ID: mdl-26968345

ABSTRACT

Leukocyte transendothelial migration (TEM) requires two major events: local dissociation of adherens junctions manifested as gaps in vascular endothelial (VE)-cadherin staining at the site of TEM and targeted trafficking of the lateral border recycling compartment (LBRC) to the site of TEM. However, the association between LBRC recycling and VE-cadherin gaps remains unknown. We found that when targeting of the LBRC is selectively inhibited using established methods, such as a function blocking anti-platelet endothelial cell adhesion molecule 1 antibody, depolymerizing microtubules, or microinjection of an antibody that inhibits kinesin, VE-cadherin gaps do not form around the blocked leukocyte. This is the first time that the LBRC has been implicated in this process. We obtained similar results for neutrophils and monocytes and in studies using live cell imaging microscopy conducted under fluid shear conditions. Depolymerizing microtubules did not affect the ability of leukocytes to induce tyrosine phosphorylation of VE-cadherin. A VE-cadherin double mutant (Y658F, Y731F) expressed in endothelial cells acted as a dominant negative and inhibited VE-cadherin gap formation and TEM, yet targeting of the LBRC still occurred. These data suggest that targeting of the LBRC to the site of TEM precedes VE-cadherin clearance. Recruitment of the LBRC may play a role in clearing VE-cadherin from the site of TEM.


Subject(s)
Adherens Junctions/physiology , Leukocytes/physiology , Transendothelial and Transepithelial Migration/physiology , Antibodies, Monoclonal/pharmacology , Antigens, CD/metabolism , Biological Transport/physiology , Cadherins/antagonists & inhibitors , Cadherins/metabolism , Cells, Cultured , Endothelium, Vascular/physiology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Intercellular Adhesion Molecule-1/metabolism , Kinesins/antagonists & inhibitors , Leukocytes, Mononuclear/physiology , Microinjections , Microtubules/physiology , Neutrophils/physiology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Protein Transport/physiology
4.
Reproduction ; 154(5): 595-605, 2017 11.
Article in English | MEDLINE | ID: mdl-28780571

ABSTRACT

CD31 has been shown to play a role in endothelial cell migration and angiogenesis, which are critical to the formation and function of the endometrium and myometrium in uterine development during early pregnancy. However, the role of CD31 in uterine receptivity during blastocyst implantation is poorly understood. The pregnancy rate in CD31-/- female mice mated with CD31+/+ male mice was higher than that observed in CD31+/+ female mice mated with CD31+/+ male mice. During the receptive phase of implantation, uterine glands were more developed in CD31-/- mice than in CD31+/+ mice, and the uterine weights of CD31-/- mice were increased. Leukemia inhibitory factor (LIF) was highly expressed in the CD31-/- mice during implantation and the expression of LIF was up-regulated by estradiol-17ß (E2 ) + progesterone (P4 ) in ovariectomized CD31-/- mice, compared with CD31+/+ mice at 8 h after hormone treatment. E2 -induced protein synthesis was inhibited by P4 in the CD31+/+ uterus, but not in the uterus of CD31-/- mice. Also, STAT3, HAND2, LIF, and mTOR signals were enhanced in CD31-/- mice. Stromal DNA replication was highly activated in the uterus of CD31-/- mice, manifested by upregulated cyclin series signaling and PCNA expression after E2 + P4 treatment. Collectively, CD31 inhibits E2 -mediated epithelial proliferation via recruitment and phosphorylation of SHP-2 upon receiving P4 signal in early pregnancy.


Subject(s)
Platelet Endothelial Cell Adhesion Molecule-1/physiology , Progesterone/pharmacology , Uterus/metabolism , Animals , Embryo Implantation/drug effects , Embryo Implantation/genetics , Endometrium/drug effects , Endometrium/metabolism , Estradiol/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pregnancy , Signal Transduction/drug effects , Uterus/drug effects
5.
Arterioscler Thromb Vasc Biol ; 34(9): 1968-76, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24969778

ABSTRACT

OBJECTIVE: Platelet endothelial cell adhesion molecule-1 (PECAM-1) regulates platelet response to multiple agonists. How this immunoreceptor tyrosine-based inhibitory motif-containing receptor inhibits G protein-coupled receptor-mediated thrombin-induced activation of platelets is unknown. APPROACH AND RESULTS: Here, we show that the activation of PECAM-1 inhibits fibrinogen binding to integrin αIIbß3 and P-selectin surface expression in response to thrombin (0.1-3 U/mL) but not thrombin receptor-activating peptides SFLLRN (3×10(-7)-1×10(-5) mol/L) and GYPGQV (3×10(-6)-1×10(-4) mol/L). We hypothesized a role for PECAM-1 in reducing the tethering of thrombin to glycoprotein Ibα (GPIbα) on the platelet surface. We show that PECAM-1 signaling regulates the binding of fluorescein isothiocyanate-labeled thrombin to the platelet surface and reduces the levels of cell surface GPIbα by promoting its internalization, while concomitantly reducing the binding of platelets to von Willebrand factor under flow in vitro. PECAM-1-mediated internalization of GPIbα was reduced in the presence of both EGTA and cytochalasin D or latrunculin, but not either individually, and was reduced in mice in which tyrosines 747 and 759 of the cytoplasmic tail of ß3 integrin were mutated to phenylalanine. Furthermore, PECAM-1 cross-linking led to a significant reduction in the phosphorylation of glycogen synthase kinase-3ß Ser(9), but interestingly an increase in glycogen synthase kinase-3α pSer(21). PECAM-1-mediated internalization of GPIbα was reduced by inhibitors of dynamin (Dynasore) and glycogen synthase kinase-3 (CHIR99021), an effect that was enhanced in the presence of EGTA. CONCLUSIONS: PECAM-1 mediates internalization of GPIbα in platelets through dual AKT/protein kinase B/glycogen synthase kinase-3/dynamin-dependent and αIIbß3-dependent mechanisms. These findings expand our understanding of how PECAM-1 regulates nonimmunoreceptor signaling pathways and helps to explains how PECAM-1 regulates thrombosis.


Subject(s)
Dynamins/physiology , Glycogen Synthase Kinase 3/physiology , Platelet Activation/physiology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/drug effects , Thrombin/pharmacology , von Willebrand Factor/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Calcium/metabolism , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Humans , Mice , Mice, Knockout , Platelet Activation/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/pharmacology , Protein Structure, Tertiary , Protein Transport , Signal Transduction/physiology , Thiazolidines/pharmacology
6.
Basic Res Cardiol ; 109(6): 448, 2014.
Article in English | MEDLINE | ID: mdl-25344833

ABSTRACT

In acute coronary syndrome (ACS), T cell abnormalities are associated to a worse outcome. Loss of inhibitory activity of CD31, an Ig-like adhesion molecule, on peripheral leukocytes has been found to enhance atherosclerosis in experimental models. In this study, we examined the expression of CD31 on T cells, and its role on TCR signaling in 35 patients with non-ST elevation ACS, in 35 patients with stable angina (SA), and in 35 controls. Furthermore, 10 ACS and 10 SA patients were re-analyzed at 1-year follow-up. Flow-cytometry analysis showed that in ACS patients, CD31 expression was reduced on total CD4(+) and CD4(+)CD28(null) (P < 0.001, ACS vs. SA), on naïve (P < 0.001, ACS vs. SA) and on central-memory and effector-memory CD4(+) T cells (P < 0.05, ACS vs. SA and controls). The immunomodulatory effect of CD31 on TCR signaling of CD4(+) and CD4(+)CD28(null) T cells, was lower in ACS than SA patients (P < 0.05, for both comparisons). At 1-year follow-up, CD31 expression and function increased in ACS becoming similar to that found in SA. CD31 recruitment in the immunological synapse was lower in ACS than controls (P = 0.012). Moreover, CD31 modulated MAPK signaling and reduced the expression of T bet and Rorγ-t, necessary for Th1 and Th17 differentiation. Finally, we studied TCR signaling in CD31(+) naïve and primed T cell subsets observing a different pattern of protein phosphorylation. A CD31-mediated regulatory pathway is enhanced in SA and temporarily downregulated in ACS. As CD31 modulates both T cell activation, by increasing the threshold for TCR stimulation, and T cell differentiation, it might represent a novel molecular target to treat T cell abnormalities in ACS.


Subject(s)
Acute Coronary Syndrome/immunology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , T-Lymphocytes, Helper-Inducer/physiology , Acute Coronary Syndrome/metabolism , Aged , Female , Humans , Lymphocyte Activation , Male , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Prospective Studies
7.
J Immunol ; 189(8): 4104-11, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22966083

ABSTRACT

The role of CD31, an Ig-like molecule expressed by leukocytes and endothelial cells (ECs), in the regulation of T lymphocyte trafficking remains contentious. Using CD31-deficient mice, we show that CD31 regulates both constitutive and inflammation-induced T cell migration in vivo. Specifically, T cell:EC interactions mediated by CD31 molecules are required for efficient localization of naive T lymphocytes to secondary lymphoid tissue and constitutive recirculation of primed T cells to nonlymphoid tissues. In inflammatory conditions, T cell:EC CD31-mediated interactions facilitate T cell recruitment to Ag-rich sites. However, endothelial CD31 also provides a gate-keeping mechanism to limit the rate of Ag-driven T cell extravasation. This event contributes to the formation of Ag-specific effector T cell infiltrates and is induced by recognition of Ag on the endothelium. In this context, CD31 engagement is required for restoring endothelial continuity, which is temporarily lost upon MHC molecule ligation by migrating cognate T cells. We propose that integrated adhesive and signaling functions of CD31 molecules exert a complex regulation of T cell trafficking, a process that is differentially adapted depending on cell-specific expression, the presence of inflammatory conditions and the molecular mechanism facilitating T cell extravasation.


Subject(s)
Platelet Endothelial Cell Adhesion Molecule-1/physiology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transendothelial and Transepithelial Migration/immunology , Animals , Cell Communication/immunology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Male , Mice , Organ Culture Techniques , Platelet Endothelial Cell Adhesion Molecule-1/administration & dosage , Platelet Endothelial Cell Adhesion Molecule-1/genetics , T-Lymphocytes/metabolism , Transendothelial and Transepithelial Migration/genetics
8.
Blood ; 117(14): 3903-6, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21297004

ABSTRACT

Inhibition of platelet responsiveness is important to control pathologic thrombus formation. Platelet-endothelial cell adhesion molecule-1 (PECAM-1) and the Src family kinase Lyn inhibit platelet activation by the glycoprotein VI (GPVI) collagen receptor; however, it is not known whether PECAM-1 and Lyn function in the same or different inhibitory pathways. In these studies, we found that, relative to wild-type platelets, platelets derived from PECAM-1-deficient, Lyn-deficient, or PECAM-1/Lyn double-deficient mice were equally hyperresponsive to stimulation with a GPVI-specific agonist, indicating that PECAM-1 and Lyn participate in the same inhibitory pathway. Lyn was required for PECAM-1 tyrosine phosphorylation and subsequent binding of the Src homology 2 domain-containing phosphatase-2, SHP-2. These results support a model in which PECAM-1/SHP-2 complexes, formed in a Lyn-dependent manner, suppress GPVI signaling.


Subject(s)
Platelet Aggregation Inhibitors , Platelet Aggregation/genetics , Platelet Endothelial Cell Adhesion Molecule-1/physiology , src-Family Kinases/physiology , Animals , Cells, Cultured , Drug Synergism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/physiology , Platelet Aggregation Inhibitors/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Platelet Membrane Glycoproteins/agonists , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , src-Family Kinases/genetics , src-Family Kinases/metabolism
9.
Arterioscler Thromb Vasc Biol ; 32(8): e90-102, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22723439

ABSTRACT

OBJECTIVE: Endothelial cells provide a barrier between the blood and tissues, which is reduced during inflammation to allow selective passage of molecules and cells. Adherens junctions (AJ) play a central role in regulating this barrier. We aim to investigate the role of a distinctive 3-dimensional reticular network of AJ found in the endothelium. METHODS AND RESULTS: In endothelial AJ, vascular endothelial-cadherin recruits the cytoplasmic proteins ß-catenin and p120-catenin. ß-catenin binds to α-catenin, which links AJ to actin filaments. AJ are usually described as linear structures along the actin-rich intercellular contacts. Here, we show that these AJ components can also be organized in reticular domains that contain low levels of actin. Reticular AJ are localized in areas where neighboring cells overlap and encompass the cell adhesion receptor platelet endothelial cell adhesion molecule-1 (PECAM-1). Superresolution microscopy revealed that PECAM-1 forms discrete structures distinct from and distributed along AJ, within the voids of reticular domains. Inflammatory tumor necrosis factor-α increases permeability by mechanisms that are independent of actomyosin-mediated tension and remain incompletely understood. Reticular AJ, but not actin-rich linear AJ, were disorganized by tumor necrosis factor-α. This correlated with PECAM-1 dispersal from cell borders. PECAM-1 inhibition with blocking antibodies or small interfering RNA specifically disrupted reticular AJ, leaving linear AJ intact. This disruption recapitulated typical tumor necrosis factor-α-induced alterations of barrier function, including increased ß-catenin phosphorylation, without altering the actomyosin cytoskeleton. CONCLUSIONS: We propose that reticular AJ act coordinately with PECAM-1 to maintain endothelial barrier function in regions of low actomyosin-mediated tension. Selective disruption of reticular AJ contributes to permeability increase in response to tumor necrosis factor-α.


Subject(s)
Adherens Junctions/physiology , Endothelial Cells/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Amides/pharmacology , Cells, Cultured , Focal Adhesion Kinase 2/physiology , Humans , Permeability , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Pyridines/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , beta Catenin/metabolism
10.
Proc Natl Acad Sci U S A ; 107(43): 18616-21, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20926749

ABSTRACT

Most patients who die from cancer succumb to treatment-refractory advanced metastatic progression. Although the early stages of tumor metastasis result in the formation of clinically silent micrometastatic foci, its later stages primarily reflect the progressive, organ-destructive growth of already advanced metastases. Early-stage metastasis is regulated by multiple factors within tumor cells as well as by the tumor microenvironment (TME). In contrast, the molecular determinants that control advanced metastatic progression remain essentially uncharacterized, precluding the development of therapies targeted against it. Here we show that the TME, functioning in part through platelet endothelial cell adhesion molecule 1 (PECAM-1), drives advanced metastatic progression and is essential for progression through its preterminal end stage. PECAM-1-KO and chimeric mice revealed that its metastasis-promoting effects are mediated specifically through vascular endothelial cell (VEC) PECAM-1. Anti-PECAM-1 mAb therapy suppresses both end-stage metastatic progression and tumor-induced cachexia in tumor-bearing mice. It reduces proliferation, but not angiogenesis or apoptosis, within advanced tumor metastases. Because its antimetastatic effects are mediated by binding to VEC rather than to tumor cells, anti-PECAM-1 mAb appears to act independently of tumor type. A modified 3D coculture assay showed that anti-PECAM-1 mAb inhibits the proliferation of PECAM-1-negative tumor cells by altering the concentrations of secreted factors. Our studies indicate that a complex interplay between elements of the TME and advanced tumor metastases directs end-stage metastatic progression. They also suggest that some therapeutic interventions may target late-stage metastases specifically. mAb-based targeting of PECAM-1 represents a TME-targeted therapeutic approach that suppresses the end stages of metastatic progression, until now a refractory clinical entity.


Subject(s)
Neoplasms, Experimental/secondary , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Apoptosis , Bone Marrow Transplantation , Cachexia/therapy , Cell Line, Tumor , Cell Proliferation , Disease Progression , Endothelial Cells/physiology , Female , Humans , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, Transgenic , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neovascularization, Pathologic , Paracrine Communication , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/immunology
11.
Carcinogenesis ; 33(9): 1717-25, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22777959

ABSTRACT

Hypoxia-inducible factors, HIF-1α and HIF-2α, are expressed in the majority of clear-cell renal cell carcinoma (CC-RCC). In vitro, HIFα isoforms regulate a differential set of genes, and their effects in vivo within CC-RCC tumours may affect outcome. The role of angiogenesis and HIFα transcriptional products, including those involved in cell metabolism and morphological dedifferentiation have not been extensively investigated and might have relevance to the development of antiangiogenic or anti-HIFα trials in primary CC-RCC, either before or after radical nephrectomy. We analysed 168 consecutive clear-cell renal tumours from 1983 to 1999 within tissue microarrays and assessed expression of HIF-1α and HIF-2α together with the protein expression of seven of their target genes (BNIP3, CA9, Cyclin D1, GLUT-1, LDH5, Oct-4 and VEGF). The expression of these factors was compared with patient overall survival and CD31 angiogenesis. We found that HIFα antigenicity deteriorated with the age of the paraffin block (P < 0.0001) and in tumours from 1983 to 1992 was deemed not to be reliable. Similar findings were found in aged archival osteosarcoma samples. This might have important implications for retrospective biomarker studies that rely on archival tissue material. HIF-1α(HIGH)/HIF-2α(LOW) tumours had a worse overall survival compared with HIF-1α(LOW)/HIF-2α(LOW) tumours (P = 0.04). Surprisingly, on multivariate analysis, high levels of CD31(+) angiogenesis was shown to be an independent prognostic marker of increased overall survival (P = 0.003). We propose that better differentiation of vascular endothelium may be a reflection of a greater production of vessel stabilization factors versus pro-angiogenic factors, and therefore a less aggressive phenotype.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/physiology , Carcinoma, Renal Cell/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Kidney Neoplasms/drug therapy , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Adult , Aged , Aged, 80 and over , Basic Helix-Loop-Helix Transcription Factors/analysis , Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Cyclin D1/analysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Kidney Neoplasms/blood supply , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Prognosis
12.
Blood ; 116(7): 1172-84, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20479283

ABSTRACT

Leukocyte extravasation depends on various adhesion receptors at endothelial cell contacts. Here we have analyzed how mouse CD99 and CD99L2 cooperate with PECAM-1. We found that antibodies against mouse CD99 and PECAM-1 trap neutrophils between endothelial cells in in vitro transmigration assays. A sequential function, as has been suggested for human PECAM-1 and CD99, could not be demonstrated. In contrast to these in vitro results, blocking CD99 or CD99L2 or gene disruption of PECAM-1 trapped neutrophils in vivo between endothelial cells and the underlying basement membrane as revealed by electron microscopy and by 3-dimensional confocal fluorescence microscopy in the inflamed cremaster tissue. Leukocyte extravasation was inhibited in interleukin-1beta-inflamed peritoneum and in the cremaster by PECAM-1 gene disruption and was further attenuated by blocking antibodies against CD99 and CD99L2. In addition, CD99 and CD99L2 were required for leukocyte extravasation in the cremaster after stimulation with tumor necrosis factor-alpha, where the need for PECAM-1 is known to be bypassed. We conclude that CD99 and CD99L2 act independently of PECAM-1 in leukocyte extravasation and cooperate in an independent way to help neutrophils overcome the endothelial basement membrane.


Subject(s)
Antigens, CD/physiology , Endothelium, Vascular/metabolism , Leukocytes/immunology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , 12E7 Antigen , Animals , Basement Membrane/immunology , Basement Membrane/metabolism , Cell Adhesion , Cell Movement , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Fluorescent Antibody Technique , Humans , Inflammation , Leukocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Peritoneum/immunology
13.
Circ Res ; 107(11): 1355-63, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20930147

ABSTRACT

RATIONALE: Hemodynamic forces caused by the altered blood flow in response to an occlusion lead to the induction of collateral remodeling and arteriogenesis. Previous work showed that platelet endothelial cell adhesion molecule (PECAM)-1 is a component of a mechanosensory complex that mediates endothelial cell responses to shear stress. OBJECTIVE: We hypothesized that PECAM-1 plays an important role in arteriogenesis and collateral remodeling. METHODS AND RESULTS: PECAM-1 knockout (KO) and wild-type littermates underwent femoral artery ligation. Surprisingly, tissue perfusion and collateral-dependent blood flow were significantly increased in the KO mice immediately after surgery. Histology confirmed larger caliber of preexisting collaterals in the KO mice. Additionally, KO mice showed blunted recovery of perfusion from hindlimb ischemia and reduced collateral remodeling, because of deficits in shear stress-induced signaling, including activation of the nuclear factor κB pathway and inflammatory cell accumulation. Partial recovery was associated with normal responses to circumferential wall tension in the absence of PECAM-1, as evidenced by the upregulation of ephrin B2 and monocyte chemoattractant protein-1, which are 2 stretch-induced regulators of arteriogenesis, both in vitro and in vivo. CONCLUSIONS: Our findings suggest a novel role for PECAM-1 in arteriogenesis and collateral remodeling. Furthermore, we identify PECAM-1 as the first molecule that determines preexisting collateral diameter.


Subject(s)
Collateral Circulation/physiology , Neovascularization, Physiologic/physiology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Animals , Cells, Cultured , Femoral Artery/physiology , Hindlimb/blood supply , Hindlimb/physiology , Ischemia/metabolism , Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Vasomotor System/physiology
14.
J Immunol ; 184(6): 3157-63, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20173029

ABSTRACT

PECAM-1 is a cell adhesion and signaling receptor that is expressed on many hematopoietic cells and at endothelial cell-cell junctions. Accumulating evidence from a number of in vitro and in vivo model systems suggests that PECAM-1 suppresses cytokine production and vascular permeability induced by a wide range of inflammatory stimuli. In several of these models of inflammatory disease, endothelial, and not leukocyte or platelet, PECAM-1 conferred protection against inflammatory insult. However, the mechanism by which endothelial PECAM-1 functions as an anti-inflammatory protein is poorly understood. It was recently suggested that PECAM-1 exerts its anti-inflammatory effects in endothelial cells by inhibiting the activity of NF-kappaB, a proinflammatory transcription factor. To confirm and extend these observations, we examined the effect of engaging, cross-linking, or expressing PECAM-1 on NF-kappaB activation in a variety of human cells. PECAM-1 had no effect on the phosphorylation of the NF-kappaB inhibitory protein, IkappaBalpha; on the nuclear translocation of NF-kappaB; on the suppression of cytokine-induced transcriptional activation of an NF-kappaB luciferase reporter plasmid; or on the cytokine-stimulated upregulation of ICAM-1, an NF-kappaB target gene, in endothelial cells. Taken together, these studies strongly suggest that the anti-inflammatory actions of PECAM-1 in endothelial cells are not likely to involve its regulation of NF-kappaB.


Subject(s)
Blood Platelets/immunology , Blood Platelets/pathology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Inflammation Mediators/physiology , NF-kappa B , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Blood Platelets/metabolism , Cell Line , Cells, Cultured , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Endothelium, Vascular/metabolism , Humans , Inflammation Mediators/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , NF-kappa B/metabolism , NF-kappa B/physiology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Transcription, Genetic/immunology
15.
Blood ; 113(24): 6246-57, 2009 Jun 11.
Article in English | MEDLINE | ID: mdl-19211506

ABSTRACT

Leukocyte transmigration is mediated by endothelial cell (EC) junctional molecules, but the associated mechanisms remain unclear. Here we investigate how intercellular adhesion molecule-2 (ICAM-2), junctional adhesion molecule-A (JAM-A), and platelet endothelial cell adhesion molecule (PECAM-1) mediate neutrophil transmigration in a stimulus-dependent manner (eg, as induced by interleukin-1beta [IL-1beta] but not tumor necrosis factor-alpha [TNF-alpha]), and demonstrate their ability to act in sequence. Using a cell-transfer technique, transmigration responses of wild-type and TNF-alpha p55/p75 receptor-deficient leukocytes (TNFR(-/-)) through mouse cremasteric venules were quantified by fluorescence intravital microscopy. Whereas wild-type leukocytes showed a normal transmigration response to TNF-alpha in ICAM-2(-/-), JAM-A(-/-), and PECAM-1(-/-) recipient mice, TNFR(-/-) leukocytes exhibited a reduced transmigration response. Hence, when the ability of TNF-alpha to directly stimulate neutrophils is blocked, TNF-alpha-induced neutrophil transmigration is rendered dependent on ICAM-2, JAM-A, and PECAM-1, suggesting that the stimulus-dependent role of these molecules is governed by the target cell being activated. Furthermore, analysis of the site of arrest of neutrophils in inflamed tissues from ICAM-2(-/-), JAM-A(-/-), and PECAM-1(-/-) mice demonstrated that these molecules act sequentially to mediate transmigration. Collectively, the findings provide novel insights into the mechanisms of action of key molecules implicated in leukocyte transmigration.


Subject(s)
Antigens, CD/physiology , Cell Adhesion Molecules/physiology , Cell Movement/physiology , Endothelium, Vascular/metabolism , Neutrophils/physiology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Receptors, Cell Surface/physiology , Animals , Cell Adhesion , Cells, Cultured , Endothelium, Vascular/cytology , Fluorescent Antibody Technique , Leukocytes/cytology , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscles/cytology , Muscles/metabolism , Receptors, Tumor Necrosis Factor, Type I/physiology , Tumor Necrosis Factor-alpha/pharmacology
16.
Arterioscler Thromb Vasc Biol ; 30(11): 2196-204, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20724702

ABSTRACT

OBJECTIVE: Ferlins are known to regulate plasma membrane repair in muscle cells and are linked to muscular dystrophy and cardiomyopathy. Recently, using proteomic analysis of caveolae/lipid rafts, we reported that endothelial cells (EC) express myoferlin and that it regulates membrane expression of vascular endothelial growth factor receptor 2 (VEGFR-2). The goal of this study was to document the presence of other ferlins in EC. METHODS AND RESULTS: EC expressed another ferlin, dysferlin, and that in contrast to myoferlin, it did not regulate VEGFR-2 expression levels or downstream signaling (nitric oxide and Erk1/2 phosphorylation). Instead, loss of dysferlin in subconfluent EC resulted in deficient adhesion followed by growth arrest, an effect not observed in confluent EC. In vivo, dysferlin was also detected in intact and diseased blood vessels of rodent and human origin, and angiogenic challenge of dysferlin-null mice resulted in impaired angiogenic response compared with control mice. Mechanistically, loss of dysferlin in cultured EC caused polyubiquitination and proteasomal degradation of platelet endothelial cellular adhesion molecule-1 (PECAM-1/CD31), an adhesion molecule essential for angiogenesis. In addition, adenovirus-mediated gene transfer of PECAM-1 rescued the abnormal adhesion of EC caused by dysferlin gene silencing. CONCLUSIONS: Our data describe a novel pathway for PECAM-1 regulation and broaden the functional scope of ferlins in angiogenesis and specialized ferlin-selective protein cargo trafficking in vascular settings.


Subject(s)
Cell Adhesion/physiology , Endothelial Cells/physiology , Membrane Proteins/physiology , Muscle Proteins/physiology , Neovascularization, Pathologic/physiopathology , Animals , Cattle , Cell Proliferation , Cells, Cultured , Down-Regulation , Dysferlin , Humans , Membrane Proteins/biosynthesis , Mice , Muscle Proteins/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
17.
Nephron Exp Nephrol ; 118(3): e60-8, 2011.
Article in English | MEDLINE | ID: mdl-21228600

ABSTRACT

AIMS: We used a rat model of renal ischemia (35 min) to test the potential involvement of platelet/endothelial cell adhesion molecule 1 (PECAM-1/CD31) in the process of S3 tubule regeneration. METHODS: A monoclonal antibody specific for murine PECAM-1 was injected i.p. immediately after kidney reperfusion or 48 h post-ischemia. One day before ischemia, each animal received an i.p. injection of 80 mg/kg 5-bromo-2'-deoxyuridine (BrdU). Experimental animals were sacrificed 1, 2, 3, 7 and 14 days post-ischemia. Renal sections were processed to characterize the histopathological alterations and the distribution of BrdU-immunopositive cells. RESULTS: Our observations showed that anti-PECAM-1 administration was associated with an inhibition of S3 tubule regeneration along with a progressive cystic dilatation of renal tubules that was particularly prominent 2 weeks post-ischemia. Interestingly, injection of anti-PECAM-1 48 h post-ischemia failed to block renal regeneration and was followed by a normal re-epithelialization of S3 tubules. CONCLUSION: Our data showed that the blockade of PECAM-1 immediately after kidney reperfusion inhibits tubular regeneration. These observations suggest that transendothelial migration of extrarenal cells could be a precocious and pivotal step in kidney reparation, but also suggest that these extrarenal cells could be essential to the process of tubular regeneration.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Kidney Tubules/physiology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Reperfusion Injury/immunology , Animals , Kidney Tubular Necrosis, Acute/immunology , Kidney Tubular Necrosis, Acute/pathology , Male , Mice , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Rats , Rats, Wistar , Regeneration/drug effects , Regeneration/immunology , Reperfusion Injury/drug therapy , Transendothelial and Transepithelial Migration
18.
Curr Opin Hematol ; 17(1): 43-52, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19996887

ABSTRACT

PURPOSE OF REVIEW: Monocytes play multiple roles in immune system functions and inflammatory diseases such as atherosclerosis. These roles are coupled to diverse trafficking and cellular migration behaviors. Here, we review recent advances in our understanding of such behaviors with emphasis on broad scale trafficking patterns and the cellular and molecular mechanisms regulating diapedesis, a central aspect of trafficking. RECENT FINDINGS: Monocytes consist of 'inflammatory' and 'resident' subsets, which exhibit differential functions and trafficking properties. Notably, the spleen has recently been identified as a reservoir of inflammatory monocytes, which are readily recruited to injured myocardium and possibly other tissues. Resident monocytes have been shown to undergo long-range crawling within the lumen of the microvasculature, which facilitates immune surveillance and rapid response to infection. Monocyte diapedesis has been demonstrated to utilize both para and transcellular migration routes facilitated by endothelial 'transmigratory cups'. A significant number of new adhesion molecules and signaling pathways have recently been uncovered as functional mediators and modulators of these processes. SUMMARY: Our improving understanding of monocyte trafficking and migration mechanisms has begun to shed light on the functions of these often enigmatic cells. Continued progress in this area will be critical for elucidating roles of monocytes in disease and for developing therapeutics that target monocytes.


Subject(s)
Cell Movement , Monocytes/cytology , Animals , Cell Adhesion , Humans , Monocytes/immunology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Signal Transduction , Spleen/cytology , Spleen/immunology
19.
Pflugers Arch ; 459(6): 793-806, 2010 May.
Article in English | MEDLINE | ID: mdl-20012875

ABSTRACT

Endothelial cells situated at the interface between blood and the vessel wall play a crucial role in controlling vascular tone and homeostasis, particularly in determining the expression of pro- and anti-atherosclerotic genes. Many of these effects are mediated by changes in the generation and release of the vasodilator nitric oxide (NO) in response to hemodynamic stimuli exerted on the luminal surface of endothelial cells by the streaming blood (shear stress) and the cyclic strain of the vascular wall. The endothelial NO synthase (eNOS) is activated in response to fluid shear stress and numerous agonists via cellular events such as; increased intracellular Ca(2+), interaction with substrate and co-factors, as well as adaptor and regulatory proteins, protein phosphorylation, and through shuttling between distinct sub-cellular domains. Dysregulation of these processes leads to attenuated eNOS activity and reduced NO output which is a characteristic feature of numerous patho-physiological disorders such as diabetes and atherosclerosis. This review summarizes some of the recent findings relating to the molecular events regulating eNOS activity.


Subject(s)
Nitric Oxide Synthase Type III/metabolism , Adaptor Proteins, Signal Transducing/physiology , Amino Acid Sequence , Biopterins/analogs & derivatives , Biopterins/metabolism , Calmodulin/metabolism , Caveolin 1/metabolism , Dynamins/physiology , Enzyme Activation , Guanylate Cyclase/metabolism , HSP90 Heat-Shock Proteins/physiology , Humans , Nitric Oxide/metabolism , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Protein Kinases/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Soluble Guanylyl Cyclase
20.
J Exp Med ; 192(1): 1-10, 2000 Jul 03.
Article in English | MEDLINE | ID: mdl-10880521

ABSTRACT

Erythrocytes infected with mature forms of Plasmodium falciparum do not circulate but are withdrawn from the peripheral circulation; they are bound to the endothelial lining and to uninfected erythrocytes in the microvasculature. Blockage of the blood flow, hampered oxygen delivery, and severe malaria may follow if binding is excessive. The NH(2)-terminal head structure (Duffy binding-like domain 1 [DBL1alpha]-cysteine-rich interdomain region [CIDR1alpha]) of a single species of P. falciparum erythrocyte membrane protein 1 (PfEMP1) is here shown to mediate adherence to multiple host receptors including platelet-endothelial cell adhesion molecule 1 (PECAM-1)/CD31, the blood group A antigen, normal nonimmune immunoglobulin M, three virulence-associated receptor proteins, a heparan sulfate-like glucosaminoglycan, and CD36. DBL2delta was found to mediate additional binding to PECAM-1/CD31. The exceptional binding activity of the PfEMP1 head structure and its relatively conserved nature argues that it holds an important role in erythrocyte sequestration and therefore in the virulence of the malaria parasite.


Subject(s)
Erythrocyte Membrane/parasitology , Plasmodium falciparum/physiology , Protozoan Proteins/physiology , ABO Blood-Group System/physiology , Animals , Binding Sites , CD36 Antigens/physiology , CHO Cells , Cell Adhesion , Cricetinae , Glycosylation , Humans , Immunoglobulin G/physiology , Immunoglobulin M/physiology , Intercellular Adhesion Molecule-1/physiology , L Cells , Mice , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Protozoan Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL