Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters

Publication year range
1.
J Immunol ; 186(7): 4347-53, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21368227

ABSTRACT

Sphingosylphosphorylcholine (SPC) is a component of high-density lipoprotein particles. We investigated the functional role of SPC in HUVECs. SPC stimulation induced production of the CCL2 chemokine in a PTX-sensitive G-protein-dependent manner. SPC treatment caused the activation of NF-κB and AP-1, which are essential for SPC-induced CCL2 production, and induced the activation of three MAPKs, ERK, p38 MAPK, and JNK. Inhibition of p38 MAPK or JNK by specific inhibitors caused a dramatic decrease in SPC-induced CCL2 production. The Jak/STAT3 pathway was also activated upon SPC stimulation of HUVECs. Pretreatment with a Jak inhibitor blocked not only SPC-induced p38 MAPK and JNK activation, but also NF-κB and AP-1 activation. Our results suggest that SPC stimulates HUVECs, resulting in Jak/STAT3-, NF-κB-, and AP-1-mediated CCL2 production. We also observed that SPC stimulated expression of the adhesion molecule ICAM-1 in HUVECs. Our results suggest that SPC may contribute to atherosclerosis; therefore, SPC and its unidentified target receptor offer a starting point for the development of a treatment for atherosclerosis.


Subject(s)
Chemokine CCL2/biosynthesis , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Phosphorylcholine/analogs & derivatives , Sphingosine/analogs & derivatives , Umbilical Veins/immunology , Umbilical Veins/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Membrane Lipids/physiology , Pertussis Toxin/physiology , Phosphorylcholine/pharmacology , Signal Transduction/immunology , Sphingosine/pharmacology , Umbilical Veins/cytology
2.
J Biol Chem ; 286(11): 9726-36, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21233208

ABSTRACT

The pathogenesis of dengue hemorrhagic fever and dengue shock syndrome (DHF/DSS), both serious complications of dengue virus (DV) infection, remains unclear. In this study, we found that anti-DV NS1 (nonstructural protein 1) polyclonal antibodies cross-reacted with human umbilical vein endothelial cells (HUVECs). We further identified a complex-specific mAb, DB16-1, which could recognize DV NS1 and cross-react with HUVECs and human blood vessels. The target protein of DB16-1 was further purified by immunoaffinity chromatography. LC-MS/MS analysis and co-immunoprecipitation revealed that the target protein of DB16-1 was human LYRIC (lysine-rich CEACAM1 co-isolated). Our newly generated anti-LYRIC mAbs bound to HUVECs in a pattern similar to that of DB16-1. The B-cell epitope of DB16-1 displayed a consensus motif, Lys-X-Trp-Gly (KXWG), which corresponded to amino acid residues 116-119 of DV NS1 and mimicked amino acid residues 334-337 in LYRIC. Moreover, the binding activity of DB16-1 in NS1 of DV-2 and in LYRIC disappeared after the KXWG epitope was deleted in each. In conclusion, DB16-1 targeted the same epitope in DV NS1 and LYRIC protein on human endothelial cells, suggesting that it might play a role in the pathogenesis of DHF/DSS. Future studies on the role of the anti-NS1 antibody in causing vascular permeability will undoubtedly be performed on sera collected from individuals before, during, and after the endothelial cell malfunction phase of a dengue illness.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Autoantibodies/immunology , Cell Adhesion Molecules/immunology , Dengue Virus/immunology , Dengue/immunology , Endothelial Cells/immunology , Molecular Mimicry/immunology , Umbilical Veins/immunology , Viral Nonstructural Proteins/immunology , Animals , Autoantigens/immunology , Capillary Permeability/immunology , Cells, Cultured , Dengue/complications , Epitope Mapping , Epitopes/immunology , Humans , Membrane Proteins , Mice , Mice, Inbred BALB C , RNA-Binding Proteins
3.
Arterioscler Thromb Vasc Biol ; 31(1): 183-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21051666

ABSTRACT

OBJECTIVE: Cell-mediated immune responses in peripheral tissues begin with T cell infiltration through endothelial cell (EC) microvessels and accumulation in the perivascular space occupied by pericytes (PC). Here, we investigate how human T cells interact with PC. METHODS AND RESULTS: We compared human placental PC with autologous umbilical vein EC. Cultured PC express lower levels of major histocompatibility complex (MHC) and positive costimulatory molecules but higher levels of negative costimulatory molecules than do EC. Unlike EC, interferon-γ-treated MHC class II-positive PC (PC(+)) cannot stimulate resting allogeneic CD4 T cell proliferation or cytokine production. Instead, coculture of resting CD4 T cells with PC(+) induces CD25 expression and renders T cells unresponsive to restimulation by EC(+) from the same donor. PC cultured across a semi-permeable membrane decrease alloreactive CD4 T cell proliferation to EC(+), an effect enhanced by pretreatment of PC with interferon-γ and partially reversed by interleukin-10 and transforming growth factor-ß neutralization, but do not induce anergy. CONCLUSIONS: Human placental PC are poorly immunogenic and negatively regulate CD4 T cell responses through contact-dependent and contact-independent mechanisms.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunity, Cellular , Lymphocyte Activation , Pericytes/immunology , Placenta/immunology , Cell Proliferation , Cells, Cultured , Clonal Anergy , Coculture Techniques , Endothelial Cells/immunology , Female , HLA Antigens/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunophenotyping , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Placenta/cytology , Pregnancy , Transforming Growth Factor beta/metabolism , Umbilical Veins/cytology , Umbilical Veins/immunology
4.
J Biol Chem ; 285(43): 32860-32868, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20724473

ABSTRACT

The activation of leukocyte function-associated antigen-1 (LFA-1) plays a critical role in regulating immune responses. The metal ion-dependent adhesion site on the I-domain of LFA-1 α(L) subunit is the key recognition site for ligand binding. Upon activation, conformation changes in the I-domain can lead LFA-1 from the low affinity state to the high affinity (HA) state. Using the purified HA I-domain locked by disulfide bonds for immunization, we developed an mAb, 2E8, that specifically binds to cells expressing the HA LFA-1. The surface plasmon resonance analysis has shown that 2E8 only binds to the HA I-domain and that the dissociation constant (K(D)) for HA I-domain is 197 nm. The binding of 2E8 to the HA I-domain is metal ion-dependent, and the affinity decreased as Mn(2+) was replaced sequentially by Mg(2+) and Ca(2+). Surface plasmon resonance analysis demonstrates that 2E8 inhibits the interaction of HA I-domain and ICAM-1. Furthermore, we found that 2E8 can detect activated LFA-1 on both JY and Jurkat cells using flow cytometry and parallel plate adhesion assay. In addition, 2E8 inhibits JY cell adhesion to human umbilical vein endothelial cells and homotypic aggregation. 2E8 treatment reduces the proliferation of both human CD4(+) and CD8(+) T cells upon OKT3 stimulation without the impairment of their cytolytic function. Taken together, these data demonstrate that 2E8 is specific for the high affinity form of LFA-1 and that 2E8 inhibits LFA-1/ICAM-1 interactions. As a novel activation-specific monoclonal antibody, 2E8 is a potentially useful reagent for blocking high affinity LFA-1 and modulating T cell activation in research and therapeutics.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Metals/immunology , Animals , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cations, Divalent/immunology , Cations, Divalent/metabolism , Cell Adhesion/drug effects , Cell Adhesion/immunology , Disulfides/immunology , Disulfides/metabolism , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Flow Cytometry , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Intercellular Adhesion Molecule-1/metabolism , Jurkat Cells , K562 Cells , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Metals/metabolism , Mice , Mice, Inbred BALB C , Muromonab-CD3/immunology , Muromonab-CD3/metabolism , Protein Structure, Tertiary , Protein Subunits/immunology , Protein Subunits/metabolism , Surface Plasmon Resonance/methods , Umbilical Veins/cytology , Umbilical Veins/immunology
5.
Arterioscler Thromb Vasc Biol ; 30(12): 2621-30, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20930173

ABSTRACT

OBJECTIVE: To investigate the role of CD4(+)CD25(+)forkhead box P 3 (Foxp3)(+) T-regulatory cells (Tregs) in protecting the activation and function of human umbilical vein endothelial cells (HUVECs) induced by proinflammatory stimulus and the mechanisms of it. METHODS AND RESULTS: ECs play a major role in atherogenic initiation, changing their quiescence into activated phenotypes to support every phase of the inflammatory process. HUVECs were incubated alone, with Tregs or CD4(+)CD25(-) T cells in the presence of anti-CD3 monoclonal antibodies for 48 hours, and then were stimulated with or without oxidized low-density lipoprotein/lipopolysaccharide for an additional 24 hours. Tregs are able to induce alternative expression of immune phenotypic markers of activated HUVECs by down modulating CD86 and to inhibit the adhesion molecule, such as vascular cell adhesion molecule-1 (VCAM-1) and proinflammatory cytokine (eg, monocyte chemoattractant protein-1 and interleukin 6), response of HUVECs to oxidized low-density lipoprotein/lipopolysaccharide. Moreover, Tregs downregulate proinflammatory factor nuclear factor-κB activation and induce resistance to suppression of anti-inflammatory factor Kruppellike factor 2 in HUVECs induced by a proinflammatory stimulus. Mechanism studies reveal that Treg-mediated suppression of HUVEC proinflammatory cytokines and adhesion molecule expression impaired by oxidized low-density lipoprotein/lipopolysaccharide require cell contact by cytotoxic T-lymphocyte antigen-4 and CD86 and by soluble factors (mainly interleukin 10 and transforming growth factor [TGF]-ß). CONCLUSIONS: Tregs may exert their protective effects against atherogenesis in part through inducing an immune-inhibitory phenotype of ECs involving cytotoxic T-lymphocyte antigen-4-dependent cell-to-cell contact and also requiring soluble factors (mainly interleukin 10 and TGF-ß).


Subject(s)
CD4 Antigens/analysis , Cell Communication , Endothelial Cells/immunology , Forkhead Transcription Factors/analysis , Inflammation/immunology , Interleukin-2 Receptor alpha Subunit/analysis , T-Lymphocytes, Regulatory/immunology , Umbilical Veins/immunology , Antigens, CD/metabolism , B7-2 Antigen/metabolism , CD3 Complex/metabolism , CTLA-4 Antigen , Cell Communication/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Coculture Techniques , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Kruppel-Like Transcription Factors/metabolism , Lipopolysaccharides/pharmacology , Lipoproteins, LDL/metabolism , NF-kappa B/metabolism , Paracrine Communication , Phenotype , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Time Factors , Transforming Growth Factor beta/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
6.
Eur J Vasc Endovasc Surg ; 41(3): 429-35, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21159528

ABSTRACT

AIMS: Homocysteine upregulates expression of adhesion molecules on endothelial cells which recruits leukocytes and initiates atherosclerosis. Endothelial cells in hyperhomocysteinemic patients are continuously exposed to high levels of homocysteine. This study exposed adult endothelial cells and endothelial cells from immune naïve foetal tissue to homocysteine chronically and studied effects on cellular adhesion molecule expression under static and flow conditions. METHODS: Human umbilical vein endothelial cells (HUVEC) and human saphenous vein endothelial cells (HSVEC) were cultured in medium containing 1 mM dl-homocysteine or l-cysteine for 5-9 days. Proliferation was assessed. Cells were subjected to flowing neutrophils and numbers of tethered, rolled fixed and transmigrated neutrophils on endothelial cells were counted and compared to controls. Immunofluorescence staining with antibodies against Intercellular adhesion molecule-1 (ICAM-1), E-selectin and P-selectin were used to quantify expression. RESULTS: Chronic treatment with 1 mM homocysteine inhibited proliferation of HUVEC and HSVEC. Homocysteine treated cells showed significantly increased expression of ICAM-1, E-selectin and to a lesser extent P-selectin. In both cell types, homocysteine significantly increased interactions between neutrophils and endothelial cells under flow conditions (p < 0.05) while cysteine had no effect. CONCLUSION: Endothelial cells from adult and immune naïve foetal tissue showed similar responses to chronic treatment with homocysteine.


Subject(s)
Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Homocysteine/metabolism , Leukocyte Rolling , Neutrophils/metabolism , Cell Proliferation , Cells, Cultured , E-Selectin/metabolism , Endothelial Cells/immunology , Fluorescent Antibody Technique , Humans , Intercellular Adhesion Molecule-1/metabolism , Neutrophils/immunology , P-Selectin/metabolism , Regional Blood Flow , Saphenous Vein/cytology , Saphenous Vein/immunology , Saphenous Vein/metabolism , Time Factors , Umbilical Veins/cytology , Umbilical Veins/immunology , Umbilical Veins/metabolism , Up-Regulation
7.
Arthritis Rheum ; 62(11): 3211-20, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20617529

ABSTRACT

OBJECTIVE: The interaction between CXCL12 and its receptor, CXCR4, in the synovium of patients with rheumatoid arthritis (RA) is important for local inflammatory cell recruitment, angiogenesis, and cytokine production. CXCR7 was recently identified as an alternative receptor for CXCL12. We undertook this study to analyze the expression of CXCR7 in RA synovium and the pathogenic role of the CXCL12/CXCR7 pathway in RA. METHODS: CXCR7 expression in RA synovial tissue was analyzed using immunohistochemistry, while expression of CXCR4 and CXCR7 on human umbilical vein endothelial cells (HUVECs) was examined using quantitative reverse transcription-polymerase chain reaction, and CXCR7 expression was also analyzed by flow cytometry. Tube formation and rat aortic ring angiogenesis assays were used to assess the effects of CCX733 (a CXCR7 antagonist) and AMD3100 (a CXCR4 antagonist) on CXCL12-induced angiogenesis. The effect of anti-CXCR4 monoclonal antibody (mAb) was also analyzed using a tube formation assay. The effects of CCX733 in a murine model of collagen-induced arthritis (CIA) were also evaluated. RESULTS: CXCR7 was expressed on endothelial cells in RA synovium and also on unstimulated HUVECs. The expression of CXCR7 on HUVECs was markedly up-regulated by interleukin-1ß (IL-1ß) stimulation, and this overexpression was further enhanced by CXCL12 treatment. Incubation with CXCL12 also promoted angiogenic activity, with addition of IL-1ß again augmenting the effect. CXCL12-induced angiogenesis was inhibited by both CXCR4 and CXCR7 antagonists and by anti-CXCR4 mAb. Furthermore, treatment with CCX733 significantly reduced the clinical arthritis scores and the numbers of vessels in the inflamed synovial tissue in mice with CIA. CONCLUSION: CXCR7 and CXCR4 are both important for angiogenesis in RA synovium, making CXCR7 another potential target molecule for novel RA angiogenesis-blocking therapies.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Endothelial Cells/metabolism , Receptors, CXCR/metabolism , Synovial Membrane/metabolism , Analysis of Variance , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Endothelial Cells/immunology , Flow Cytometry , Humans , Immunohistochemistry , Mice , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Rats , Receptors, CXCR/genetics , Receptors, CXCR/immunology , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane/immunology , Umbilical Veins/immunology , Umbilical Veins/metabolism
8.
J Exp Med ; 152(2 Pt 2): 249s-255s, 1980 Aug 01.
Article in English | MEDLINE | ID: mdl-6157767

ABSTRACT

Human endothelial cells were obtained by collagenase digestion of the umbilical vein wall, explanted into tissue culture, and grown as monolayers of cells. Endothelial cells extracted from these monolayers were specifically lysed by anti-HLA-DR alloantisera. Moreover, the stimulating capacity of these endothelial cells toward allogeneic peripheral blood mononuclear lymphocytes was specifically and significantly inhibited by the presence of relevant anti-HLA-DR antisera. Endothelial cells that expressed HLA-DR determinants were also capable of substituting for macrophages in the lymphoproliferative response of purified T cells to soluble protein antigens. Moreover, in concordance with the results previously reported in which macrophages were employed, the endothelial cell donor should share HLA-DR determinants with the T cell donor for an optimal response to occur.


Subject(s)
Endothelium/immunology , Histocompatibility Antigens Class II/immunology , T-Lymphocytes/immunology , Antibody-Dependent Cell Cytotoxicity , Endothelium/cytology , Epitopes , Humans , Lymphocyte Activation , Umbilical Veins/immunology
9.
J Exp Med ; 157(4): 1339-53, 1983 Apr 01.
Article in English | MEDLINE | ID: mdl-6403654

ABSTRACT

We have used monoclonal antibody binding, measured by radioimmunoassay, fluorescence flow cytometry, and ultrastructural immunocytochemistry, to measure expression of Ia antigens on cultured human umbilical vein endothelial (HUVE) cells. Under standard culture conditions, HUVE cells do not express Ia antigens. However, treatment of primary HUVE cultures with phytohemagglutinin induces the expression of Ia antigens. Every endothelial cell in the culture becomes Ia-positive and endothelial cells appear to synthesize Ia. HLA-A,B is concomitantly increased. The expression of Ia appears to be mediated by T cells because (a) pretreatment of primary HUVE cultures with OKT3 plus complement blocks the action of the lectins but not of medium conditioned by lectin-activated peripheral blood mononuclear cells; (b) co-culture of endothelial cells with allogeneic T cells, in the absence of lectin, also induces endothelial Ia; and (c) human immune (gamma) interferon, produced by Chinese hamster ovary cells transfected with the human gamma interferon gene, directly induces endothelial Ia. During co-culture with lymphocytes, about one-third of the endothelial cells are Ia-positive after 24 h and all of the endothelial cells are Ia-positive by 72 h. Proliferation of allogeneic T cells starts by 96 h and peaks at 144 h. Thus, endothelial Ia appears sufficiently early to be a determinant for the proliferation of allogeneic T cells. Inducible expression of Ia by endothelium may be important both for allograft rejection and for recruitment of circulating T cells into the site of an immune response.


Subject(s)
Histocompatibility Antigens Class II/analysis , Lymphocyte Activation , T-Lymphocytes/immunology , Umbilical Veins/immunology , Cells, Cultured , Endothelium/cytology , Endothelium/immunology , Humans , Interferon-gamma/physiology , Phytohemagglutinins/pharmacology
10.
J Exp Med ; 188(9): 1751-6, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9802986

ABSTRACT

Interleukin (IL)-8, a C-X-C chemokine, activates integrin-mediated adhesion of neutrophils. Presentation of IL-8 on the endothelial cell surface may promote leukocyte extravasation. We found that cultured human microvascular endothelial cells from the intestine (HIMEC) and from nasal polyps (PMEC), but not human umbilical vein endothelial cells (HUVEC), contained IL-8 in intracellular granules that coexpressed von Willebrand factor (vWf ). This observation was corroborated by the immunohistochemical observation of double-positive granules (IL-8(+)vWf+) in vessels of small and large intestine, nasal mucosa, and skin, whereas umbilical cords revealed no endothelial IL-8. After treatment of HIMEC or PMEC with histamine or thrombin, a dramatic increase in supernatant IL-8 concentration was observed within 3 min, whereas no increase in IL-8 was detected in supernatants of identically treated HUVEC cultures. Histamine or thrombin treatment also caused IL-8-containing granules to rapidly disappear from HIMEC. In HUVEC, IL-8-containing granules were inducible by treatment with recombinant human IL-1beta for 24 h; additional histamine treatment doubled IL-8 secretion from HUVEC in the same rapid manner observed for mucosal EC. These data suggested that IL-8 prestored in microvascular endothelial cells may provide a rapid pathway for specific activation of neutrophil adhesion at sites of acute inflammation.


Subject(s)
Endothelium, Vascular/immunology , Endothelium, Vascular/physiology , Interleukin-8/metabolism , Cell Adhesion/physiology , Cells, Cultured , Endothelium, Vascular/drug effects , Histamine/pharmacology , Humans , Inflammation/etiology , Inflammation/immunology , Microcirculation/drug effects , Microcirculation/immunology , Microcirculation/physiology , Microscopy, Fluorescence , Neutrophils/physiology , Organelles/immunology , Organelles/metabolism , Thrombin/pharmacology , Umbilical Veins/drug effects , Umbilical Veins/immunology , Umbilical Veins/physiology , von Willebrand Factor/metabolism
11.
J Exp Med ; 188(9): 1757-62, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9802987

ABSTRACT

The expression and secretion of interleukin (IL)-8, the prototype member of the C-X-C subfamily of chemokines, can be induced by diverse inflammatory stimuli in many cells, including endothelial cells (EC). Upon de novo synthesis, IL-8 localizes intracellularly in the Golgi apparatus, from where it is secreted. In addition to this constitutive secretory pathway, we describe a depot storage and separate regulated secretory pathway of IL-8 in EC. The prolonged stimulation of primary human EC with inflammatory mediators resulted in the accumulation of IL-8 in Weibel-Palade bodies, where it colocalized with von Willebrand factor. IL-8 was retained in these storage organelles for several days after the removal of the stimulus and could be released by EC secretagogues such as phorbol myristate acetate, the calcium ionophore A23187, and histamine. These findings suggest that storage of IL-8 in Weibel-Palade bodies may serve as the EC "memory" of a preceding inflammatory insult, which then enables the cells to secrete IL-8 immediately without de novo protein synthesis.


Subject(s)
Endothelium, Vascular/immunology , Inflammation/immunology , Interleukin-8/metabolism , Calcimycin/pharmacology , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/ultrastructure , Golgi Apparatus/immunology , Histamine/pharmacology , Humans , Interleukin-1/pharmacology , Ionophores/pharmacology , Microscopy, Fluorescence , Microscopy, Immunoelectron , Organelles/drug effects , Organelles/immunology , Organelles/ultrastructure , Tetradecanoylphorbol Acetate/pharmacology , Umbilical Veins/drug effects , Umbilical Veins/immunology , Umbilical Veins/ultrastructure
12.
Crit Care ; 14(6): R201, 2010.
Article in English | MEDLINE | ID: mdl-21059228

ABSTRACT

INTRODUCTION: This study was carried out to determine whether interactions of cell activation, shear stress and platelets at sites of endothelial injury explain the paradoxical maldistribution of activated leukocytes during sepsis away from local sites of infection towards disseminated leukocyte accumulation at remote sites. METHODS: Human umbilical venous endothelial cells (HUVEC) and polymorphonuclear neutrophils (PMN) were activated with lipopolysaccharide at 100 and 10 ng/ml to achieve adhesion molecule patterns as have been reported from the hyper- and hypo-inflammatory stage of sepsis. To examine effects of leukocyte activation on leukocyte-endothelial interactions, activated HUVEC were perfused with activated and non-activated neutrophils in a parallel plate flow chamber. Adhesion molecule expression and function were assessed by flow cytometry and blocking antibodies. In a subset of experiments the sub-endothelial matrix was exposed and covered with platelets to account for the effects of endothelial injury. To investigate interactions of these effects with flow, all experiments were done at various shear stress levels (3 to 0.25 dyne/cm(2)). Leukocyte-endothelial interactions were analyzed by videomicroscopy and analysis of covariance. RESULTS: Activation of neutrophils rendered adhesion increasingly dependent on shear stress reduction. At normal shear stress, shedding of L-selectin decreased adhesion by 56%. Increased rolling fractions of activated PMN at low shear stress revealed impaired integrin affinity despite numerical up-regulation of CD11b. On sub-maximally activated, intact HUVEC shear stress became the prevailing determinant of adhesion. Presence of a platelet-covered injury with high surface density of P-selectin was the strongest variable for adhesion. When compared to maximally activated HUVEC, platelets increased neutrophil adhesion by 2.7-fold. At sub-maximal activation a 10-fold increase was observed (P < 0.05 for all). CONCLUSIONS: L-selectin shedding and integrin dysfunction render leukocyte adhesion increasingly susceptible to shear stress and alternative adhesion receptors. In combination, these effects inhibit recruitment to normally perfused sites with intact endothelium and favor maldistribution towards sites with compromised perfusion or endothelial injury.


Subject(s)
Endothelium, Vascular/pathology , Inflammation Mediators/physiology , Leukocytes/metabolism , Leukocytes/pathology , Sepsis/metabolism , Shear Strength/physiology , Blood Flow Velocity/immunology , Cell Adhesion/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Leukocytes/immunology , Lipopolysaccharides/physiology , Sepsis/etiology , Sepsis/pathology , Umbilical Veins/immunology , Umbilical Veins/metabolism , Umbilical Veins/pathology
13.
J Thromb Thrombolysis ; 30(2): 164-71, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20155436

ABSTRACT

Human cytomegalovirus (HCMV) establishes a life-long persistent infection. HCMV infection could be associated with chronic inflammatory diseases, such as cardiovascular disease and atherosclerosis. Here we observed that in HCMV (AD-169) pre-exposed human umbilical vein endothelial cells (HUVEC), thrombin-induced expression of IL-1alpha and M-CSF is markedly enhanced compared to the un-exposed cells. Study of the expression of thrombin receptor genes in HUVEC showed that HCMV triggered a time- and concentration-dependent expression of the thrombin receptors PAR1, PAR3 and PAR4 at the mRNA level. Induction of PAR1 and PAR3 mRNA expression is due to transcriptional activation of their promoters as shown by gene reporter assay. Furthermore, the virus induced expression of PAR1 and PAR3 but not PAR4 proteins, as analyzed by Western immunoblotting. However, flow cytometric analysis revealed that only PAR3, expressed at very low level in control HUVEC, is induced at the surface during the exposure to the virus. Our data suggest that although exposure to HCMV induces a minor increase of cell-surface receptors expression, it does make endothelial cells more responsive to additional thrombin stimulation.


Subject(s)
Cytomegalovirus/pathogenicity , Endothelial Cells/virology , Receptors, Thrombin/metabolism , Thrombin/metabolism , Umbilical Veins/virology , Blotting, Western , Cells, Cultured , Endothelial Cells/immunology , Endothelial Cells/metabolism , Flow Cytometry , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptor, PAR-1/metabolism , Receptors, Thrombin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcriptional Activation , Umbilical Veins/cytology , Umbilical Veins/immunology , Umbilical Veins/metabolism , Up-Regulation
14.
Int J Oncol ; 34(1): 273-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19082498

ABSTRACT

In order to proliferate, solid tumours require the development and continuous expansion of an organised host-derived vascular network. The anti-vascular agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) emerged as derivative of the flavone-8-acetic acid (FAA) and xanthenone-4-acetic acid (XAA). Its anti-vascular activity is not based on direct cytotoxic effects, but is characterized by an immune-mediated component, through the activation of NF-kappaB pathway, and a direct anti-vascular action, involving the induction of endothelial cell apoptosis and changes in tumour vessel permeability. Despite promising pre-clinical results, DMXAA showed moderate anti-tumour activity in clinical trials. In this study, we compared to DMXAA the in vitro immune-modulating and the anti-vascular properties of two XAA analogues, AP/1649 and AP/1897. Their immune-stimulating activities were evaluated on a human monocyte cell line and their anti-vascular activities were studied by measuring the induction of HUVECs apoptosis and using DCE-MRI to determine tumour perfusion following drug treatment. Although the two molecules exerted an immune stimulation comparable to that produced by DMXAA, they showed reduced (AP/1649) or minimal (AP/1897) anti-vascular activity in vitro, and no anti-vascular effects in vivo. These results endorse the current theories concerning two independent actions exerted by DMXAA.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Immunity, Cellular/drug effects , Microcirculation/drug effects , Xanthones/pharmacology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Flow Cytometry , Humans , I-kappa B Kinase , Leukemia/immunology , Leukemia/metabolism , Leukemia/pathology , Magnetic Resonance Imaging , Mice , Mice, Inbred CBA , NF-kappa B/metabolism , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphorylation , Sarcoma, Experimental/immunology , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Tumor Necrosis Factor-alpha , Umbilical Veins/cytology , Umbilical Veins/immunology , Umbilical Veins/metabolism
15.
J Periodontal Res ; 44(1): 103-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19515021

ABSTRACT

BACKGROUND AND OBJECTIVE: The role of human leukocyte histocompatibility antigen (HLA) class II molecules on non-antigen-presenting cells has been a matter of controversy. We previously reported that HLA-II molecules on human gingival fibroblasts (GF) do not present antigens, but transduce signals into the cells, resulting in the expression of several cytokines, such as interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), regulated upon activation, normal T-cell expressed and secreted (RANTES) and IL-8. However, the exact role of these cytokines, as well as other cytokines which are potentially secreted from GF, in the pathogenesis of chronic periodontal inflammation is not fully understood. The aim of this study was to observe the effects of HLA-II-induced cytokines on the proliferation of human umbilical vein endothelial cells (HUVEC). MATERIAL AND METHODS: Antibody-based cytokine-microarray analyses were performed to detect potential cytokines associated with angiogenesis. Next, cytokine productivity was confirmed by quantitative methods. Then, cell proliferation assay was performed to see whether these cytokines promoted the proliferation of HUVEC. RESULTS: Besides IL-6, MCP-1, RANTES and IL-8, growth-related gene product (GRO) was newly identified as an HLA-II-induced cytokine released from GF. This was confirmed by a quantitative method. Cell culture supernatant from HLA-II-stimulated GF cultures promoted the growth of HUVEC. Addition of anti-IL-8 neutralizing antibody, anti-CXC receptor (CXCR)1 antibody and anti-MCP-1 antibody inhibited the growth of HUVEC in a dose-dependent manner, while addition of anti-GROalpha antibody did not. CONCLUSION: The HLA-II-induced IL-8, via CXCR1, as well as MCP-1 from GF, promotes endothelial cell proliferation, which is possibly associated with enhanced angiogenesis in chronic periodontal lesions.


Subject(s)
Chronic Periodontitis/pathology , Cytokines/immunology , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Fibroblasts/immunology , Gingiva/immunology , Histocompatibility Antigens Class II/immunology , Neovascularization, Pathologic/pathology , Umbilical Veins/pathology , Antibodies/immunology , Cell Proliferation , Cells, Cultured , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/immunology , Chemokine CCL5/immunology , Chemokine CXCL1/immunology , Chronic Periodontitis/immunology , Endothelial Cells/immunology , Endothelium, Vascular/immunology , Gingiva/pathology , HLA-DQ Antigens/immunology , HLA-DR Antigens/immunology , Humans , Interleukin-6/immunology , Interleukin-8/antagonists & inhibitors , Interleukin-8/immunology , Neovascularization, Pathologic/immunology , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8A/immunology , Umbilical Veins/immunology
16.
Vascul Pharmacol ; 50(3-4): 116-22, 2009.
Article in English | MEDLINE | ID: mdl-19063994

ABSTRACT

Vascular inflammation is an important factor which can promote diabetic complications. Preliminary investigations of several crude plant extracts including aqueous extract of Benincasa hispida Cogniaux exhibit anti-inflammatory properties. This study investigates the mechanism of anti-vascular inflammatory activity of an aqueous extract of B. hispida Cogniaux (ABH) in human umbilical vein endothelial cells (HUVECs). The study was performed on HUVECs that were pretreated with various concentrations (1-20 microg/ml) of ABH before exposure with high glucose (25 mM) for 48 h. Cell ELISA and Western blot analysis showed that ABH inhibited high glucose-induced cell adhesion molecules (CAMs) surface and protein expression, resulting in reduced adhesion of U937 monocytes. ABH also inhibited the mRNA expression level of monocyte chemoattractant protein-1 (MCP-1) and interleukin-8 (IL-8). High glucose-induced ROS production was inhibited by treatment of ABH. We observed that pretreatment with HUVECs with ABH blocks NF-kappaB activation via blocking phosphorylation and degradation of its inhibitory protein, IkappaB-alpha. ABH also reduced NF-kB promoter activity. These results suggest that ABH reduces high glucose-induced CAMs activation by inhibiting monocyte adhesion, ROS, and NF-kappaB in HUVECs.


Subject(s)
Cucurbitaceae , Endothelium, Vascular/immunology , Glucose/pharmacology , Plant Extracts/pharmacology , Umbilical Veins/drug effects , Umbilical Veins/immunology , Vasculitis/prevention & control , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cells, Cultured , Endothelium, Vascular/drug effects , Female , Glucose/administration & dosage , Humans , In Vitro Techniques , Interleukin-8/metabolism , Monocytes/drug effects , NF-kappa B/biosynthesis , Pregnancy , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Umbilical Veins/metabolism , Vasculitis/chemically induced
17.
Int J Cancer ; 122(10): 2266-73, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18224682

ABSTRACT

In the present study, we investigated the mechanisms by which anti-endoglin (EDG; CD105) monoclonal antibodies (mAbs) suppress angiogenesis and tumor growth. Antihuman EDG mAb SN6j specifically bound to murine endothelial cells and was internalized into the cells in vitro. SN6j effectively suppressed angiogenesis in mice in the Matrigel plug assay. We found that SN6j is more effective for tumor suppression in immunocompetent mice than in SCID mice. We hypothesized that T cell immunity is important for effective antitumor efficacy of SN6j in vivo. To test this hypothesis, we investigated effects of CpG oligodeoxynucleotides (ODN) and depletion of CD4(+) T cells and/or CD8(+) T cells on antitumor efficacy of SN6j in mice. Systemic (i.v.) administration of a relatively small dose (0.6 mug/g body weight/dose) of SN6j suppressed growth of established s.c. tumors of colon-26 in BALB/c mice and improved survival of the tumor-bearing mice. Addition of CpG ODN to SN6j synergistically enhanced antitumor efficacy of SN6j. In contrast, such enhancing effects of CpG ODN were not detected in SCID mice. Antitumor efficacy of SN6j in BALB/c mice was abrogated when CD4(+) T cells and/or CD8(+) T cells were depleted; effect of CD8(+) T cell depletion was stronger. Interestingly, CD4-depletion decreased tumor growth while CD8-depletion enhanced tumor growth in the absence of SN6j. SN6j induced apoptosis in human umbilical vein endothelial cells in a dose-dependent manner which indicates an additional mechanism of antiangiogenesis by SN6j. (c) 2008 Wiley-Liss, Inc.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Colonic Neoplasms/blood supply , Colonic Neoplasms/therapy , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/therapy , Receptors, Cell Surface/immunology , Animals , Antibodies, Monoclonal/immunology , Apoptosis , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Collagen/metabolism , Colonic Neoplasms/immunology , DNA/therapeutic use , Drug Combinations , Drug Synergism , Endoglin , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Laminin/metabolism , Lymphocyte Depletion , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , NIH 3T3 Cells , Neovascularization, Pathologic , Oligodeoxyribonucleotides , Proteoglycans/metabolism , Telangiectasia, Hereditary Hemorrhagic , Umbilical Veins/cytology , Umbilical Veins/immunology , Umbilical Veins/metabolism
18.
Eur J Cancer ; 44(3): 383-90, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18060766

ABSTRACT

Vaccines targeting tumour angiogenesis were recently shown to inhibit tumour growth in animal models. However, there is still a lack of information about the clinical utility of anti-angiogenic vaccination. Therefore, here, we aimed to test the clinical effects of a vaccine using glutaraldehyde-fixed human umbilical vein endothelial cells (HUVECs). Six patients with recurrent malignant brain tumours and three patients with metastatic colorectal cancer received intradermal injections of 5x10(7) HUVECs/dose (in total 230 vaccinations). ELISA and flow cytometry revealed immunoglobulin response against HUVECs' membrane antigens. ELISPOT and chromium-release cytotoxicity assay revealed a specific cellular immune response against HUVECs, which were lysed in an effectors:targets ratio-dependent manner. Gadolinium-contrasted MRI showed partial or complete tumour responses in three malignant brain tumour patients. Except for a DTH-like skin reaction at the injection site, no adverse effect of vaccination could be observed. Our results suggest that the endothelial vaccine can overcome peripheral tolerance of self-angiogenic antigens in clinical settings, and therefore should be useful for adjuvant immunotherapy of cancer.


Subject(s)
Brain Neoplasms/prevention & control , Cancer Vaccines/administration & dosage , Colorectal Neoplasms/prevention & control , Endothelium, Vascular/immunology , Neovascularization, Pathologic/prevention & control , Umbilical Veins/immunology , Adult , Aged , Brain Neoplasms/blood supply , Colorectal Neoplasms/blood supply , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunity, Cellular , Interferon-gamma/metabolism , Male , Middle Aged , Neoplasm Metastasis/prevention & control , Neoplasm Recurrence, Local/prevention & control , Pilot Projects , T-Lymphocytes, Cytotoxic/immunology , Treatment Outcome , Umbilical Veins/cytology
19.
Scand J Immunol ; 67(5): 523-31, 2008 May.
Article in English | MEDLINE | ID: mdl-18363595

ABSTRACT

Activated endothelium releases the soluble adhesion molecules vascular cell adhesion molecule-1 (sVCAM-1) and intercellular adhesion molecule-1 (sICAM-1). Measurement of fluid-phase adhesion molecules is therefore used to quantify endothelial activation, but it is unclear which is the better marker. The aims of the study were to compare the relationships between mRNA, surface and total expression and released VCAM-1 and ICAM-1 in endothelial cell cultures during activation, and to compare human umbilical vein endothelial cells (HUVEC) with the microvascular cell line HMEC-1. sVCAM-1 better represented mRNA and surface expression changes in HUVEC undergoing endotoxin stimulation than did sICAM-1. Very little VCAM-1 was released from endotoxin-stimulated HMEC-1, and sICAM-1 seemed a better activation marker for these cells. During incubation of HUVEC in media with glucose concentrations of 5.6, 10.6 or 20.6 mM, VCAM-1 was released to the media in a dose-dependent way without changes in surface expression. ICAM-1 was not influenced by the glucose concentration. There are situations when VCAM-1 concentrations in the media do not mirror the surface expression on HUVEC in culture, indicating that measurements of soluble adhesion molecules may not necessarily be representative of the conditions on the cell surface. Endothelium from different locations showed varying responses with respect to VCAM-1 and ICAM-1 liberation upon endotoxin stimulation. Thus, both sVCAM-1 and sICAM-1 should be quantified in clinical studies of endothelial activation until their characteristics are better clarified.


Subject(s)
Endothelial Cells/immunology , Intercellular Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Endotoxins/pharmacology , Humans , Intercellular Adhesion Molecule-1/genetics , RNA, Messenger/genetics , Solubility , Umbilical Veins/immunology , Vascular Cell Adhesion Molecule-1/genetics
20.
Clin Exp Pharmacol Physiol ; 35(1): 89-96, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18047634

ABSTRACT

1. Human endothelial cells express proteinase-activated receptor-2 (PAR-2), inflammatory cytokines and trypsin (EC 3.4.21.4). However, little is known about the mechanism through which trypsin induces cytokine release from endothelial cells. 2. In the present study, we investigated the effect of trypsin on cytokine release from primary cultures of human umbilical vein endothelial cells (HUVEC) using an antibody based protein microarray and ELISA. 3. The results showed that 1 microg/mL trypsin induced release of 32 different inflammatory factors, whereas 100 micromol/L Ser-Leu-Ile-Gly-Lys-Val-NH2 (SLIGKV-NH2) only stimulated secretion of 16 inflammatory factors from HUVEC, as assessed by an antibody based protein microarray. Because the release of interleukin (IL)-1a, IL-8, IL-10 and IL-12 was markedly increased following PAR-2 activation, their release was investigated further using ELISA. Increases in release of up to approximately 4.8-, 4.3-, 4.1- and 1.8-fold were observed for IL-1a, IL-10, IL-12 and IL-8, respectively, when HUVEC were challenged with trypsin for 16 h. Agonist peptides of PAR-2, namely SLIGKV-NH2 and trans-cinnamoyl-Leu-Ile-Gly-Arg-Leu-Orn-NH2 (tc-LIGRLO-NH2), also provoked significant release of IL-8. Trypsin-induced cytokine release was inhibited by its inhibitors soybean trypsin inhibitor, alpha1-antitrypsin and the inhibitor peptide of PAR-2 Phe-Ser-Leu-Leu-Arg-Tyr-NH2 (FSLLRY-NH2). 4. These data indicate the action of trypsin on HUVEC is most likely through activation of PAR-2, suggesting that PAR-2-related mechanisms are involved in the inflammatory process in humans.


Subject(s)
Cytokines/metabolism , Endothelial Cells/drug effects , Inflammation Mediators/metabolism , Oligopeptides/pharmacology , Receptor, PAR-2/agonists , Signal Transduction/drug effects , Trypsin/metabolism , Umbilical Veins/drug effects , Antigens, CD/analysis , Cells, Cultured , Dose-Response Relationship, Drug , Endoglin , Endothelial Cells/immunology , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-1alpha/metabolism , Interleukin-8/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Protein Array Analysis , Receptor, PAR-2/metabolism , Receptors, Cell Surface/analysis , Trypsin/pharmacology , Umbilical Veins/cytology , Umbilical Veins/immunology , Umbilical Veins/metabolism , alpha 1-Antitrypsin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL