Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Thromb Res ; 238: 172-183, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38723522

RESUMO

INTRODUCTION: Cancer cells induce hypercoagulability in the tumoral microenvironment by expressing Tissue Factor (TF). We aimed to study the impact of the procoagulant signature of cancer cells on the quality and structure of fibrin network. We also studied the impact of fibrin clot shield (FCS) on the efficiency of anticancer agents and the migration of cancer cells. MATERIALS AND METHODS: Pancreatic cancer cells BXPC3 and breast cancer cells MDA-MB231 and MCF7, were cultured in the presence of normal Platelet Poor Plasma (PPP), diluted 10 % in conditioning media. Their potential to induce thrombin generation and their fibrinolytic activity were assessed. The structure of fibrin network was analyzed with Scanning Electron Microscopy (SEM). Cancer cells' mobility with fibrin clot and their interactions with fibrin were observed. Cancer cells were treated with paclitaxel (PTX) or 4-hydroxy-tamoxifen (4OHTam) in the presence or absence of FCS. RESULTS: Cancer cells, in presence of PPP, induced fibrin network formation. High TF-expressing cancer cells (BXPC3 and MDA-MB23 cells), led to dense fibrin network with fine fibers. Low TF expressing cells MCF7 led to thick fibers. Exogenous TF enhanced the density of fibrin network formed by MCF7 cells. Cancer cells through their inherent profibrinolytic potential migrated within the fiber scaffold. The BXPC3 and MCF7 cells moved in clusters whereas the MDA-MB231 cells moved individually within the fibrin network. FCS decreased the efficiency of PTX and 4OHTam on the viability of cancer cells. CONCLUSIONS: The procoagulant signature of cancer cells is determinant for the quality and structure of fibrin network in the microenvironment. Original SEM images show the architecture of "bird's nest"-like fibrin network being in touch with the cell membranes and surrounding cancer cells. Fibrin network constructed by triggering thrombin generation by cancer cells, provides a scaffold for cell migration. Fibrin clot shields protect cancer cells against PTX and 4OHTam.

2.
TH Open ; 3(4): e340-e347, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31693008

RESUMO

Introduction Hypercoagulability is a common blood alteration in newly diagnosed chemotherapy naïve patients with multiple myeloma. The identification of the procoagulant potential of cancer cells, which is principally related to tissue factor (TF) expression, attracts particular interest. The mechanisms by which myeloma plasma cells (MPCs) activate blood coagulation have been poorly investigated. Aim To identify the principal actors related with MPCs that boost thrombin generation (TG). Methods TF and annexin V expression by MPCs and MPC-derived microparticles (MPC-dMPs) was analyzed by flow cytometry. TF activity (TFa) and TF gene expression were also determined. TG in the presence of MPCs or MPC-dMPs was assessed with the calibrated automated thrombogram assay (CAT) in normal human PPP and in plasma depleted of factor VII or XII. TG was also assessed in plasma spiked with MPCs and MPC-dMPs. Results MPC-dMPs expressed approximately twofold higher levels of TF as compared with MPCs. The TFa expressed by MPC-dMPs was significantly higher compared with that expressed by MPCs. MPCs and MPC-dMPs enhanced TG of human plasma. TG was significantly higher with MPC-dMPs compared with MPCs. Conclusion MPCs indirectly induce blood-borne hypercoagulability through the release of MPC-dMPs rich in TF. Since MPCs, expressing low TFa, represent a weak procoagulant stimulus, the hypercoagulability at the microenvironment could be the resultant of MPC-dMPs rich in TF.

3.
Clin Appl Thromb Hemost ; 24(4): 684-690, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28681632

RESUMO

The beneficial effect of autologous peripheral blood stem cell transplantation (APBSCT) may be compromised by acute vascular complications related to hypercoagulability. We studied the impact of graft product on thrombin generation of normal plasma and the expression of tissue factor (TF) and procoagulant platelet-derived procoagulant microparticles (Pd-MPs) in samples of graft products. Graft products from 10 patients eligible for APBSCT were mixed with platelet-poor plasma (PPP) or platelet-rich plasma (PRP) from healthy volunteers and assessed for in vitro thrombin generation. In control experiments, thrombin generation was assessed in (1) PPP and PRP without any exogenous TF and/or procoagulant phospholipids, (2) PPP with the addition of TF (5 pM) and procoagulant phospholipids (4 µM), (3) in PRP with the addition of TF (5 pM). Graft products were assessed with Western blot assay for TF expression, with a specific clotting assay for TF activity and with flow cytometry assay for Pd-MPs. The graft product enhanced thrombin generation and its procoagulant activity was related to the presence of Pd-MPs and TF. The concentration of Pd-MPs in the graft product was characterized by a significant interindividual variability. The present study reveals the need for a thorough quality control of the graft products regarding their procoagulant potential.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Transplante de Células-Tronco de Sangue Periférico/métodos , Trombina/metabolismo , Tromboplastina/metabolismo , Humanos
4.
Clin Appl Thromb Hemost ; 23(2): 155-163, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27609342

RESUMO

BACKGROUND: A documented relationship between ovarian cancer and thrombosis does exist. Low-molecular-weight heparins (LMWHs) are cornerstone drugs in the primary prevention and treatment of venous thromboembolic events in patients with cancer. However, cancer cells may alter the efficiency of these antithrombotic agents. OBJECTIVE: We aimed to characterize the procoagulant phenotype of human epithelial ovarian adenocarcinoma cells, IGROV1, and to compare the capacity of tinzaparin and enoxaparin to inhibit thrombin generation triggered by these cells. METHODS: Thrombin generation induced by different concentrations of IGROV1 cells on platelet poor plasma (PPP) was assessed by the calibrated automated thrombogram assay. Tissue factor (TF) expression was studied using Western blot analysis. Then, the experimental model of thrombin generation was used to compare the inhibitory effect of clinically relevant concentrations of both tinzaparin and enoxaparin. The inhibitory concentration 50 (IC50) of the mean rate index and the endogenous thrombin potential and the 2-fold increase in lag time were analyzed on the basis of the anti-Xa and anti-IIa activities of the LMWHs. RESULTS: IGROV1 cells suspended into PPP resulted in a significant increase in thrombin generation in the absence of any exogenous source of TF and phospholipids. Tissue factor was expressed by IGROV1 cells. Tinzaparin was a more potent inhibitor of thrombin generation than enoxaparin. The inhibition of thrombin generation induced by IGROV1 cancer cells depended mainly on the anti-Xa activity of the LMWHs. CONCLUSION: This experimental study in ovarian cancer cells demonstrates that the antithrombotic activity of LMWHs is not completely predicted by the anti-Xa or anti-IIa activities measured in PPP.


Assuntos
Fibrinolíticos/farmacologia , Heparina de Baixo Peso Molecular/farmacologia , Neoplasias Ovarianas/patologia , Trombina/biossíntese , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Enoxaparina/farmacologia , Inibidores do Fator Xa/sangue , Feminino , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Plasma , Protrombina/antagonistas & inibidores , Trombina/efeitos dos fármacos , Tromboplastina/análise , Tromboplastina/efeitos dos fármacos , Tinzaparina
5.
Thromb Res ; 136(6): 1273-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26489727

RESUMO

INTRODUCTION: Cancer cells may alter the efficiency of the antithrombotic agents. To explore this possibility, the present study compared the capacity of the LMWH enoxaparin and the specific inhibitors of Xa (apixaban and fondaparinux) to inhibit thrombin generation triggered by pancreas adenocarcinoma cells (BXPC3) and human breast carcinoma cells (MCF7). MATERIALS AND METHODS: Samples of platelet poor (PPP) or platelet rich plasma (PRP) spiked with apixaban, fondaparinux or enoxaparin were added in micro wells carrying cancer cells and assessed for thrombin generation. In the control experiment thrombin generation was triggered with tissue factor reagent. RESULTS: The three antithrombotics inhibited thrombin generation in a concentration dependent manner. The BXPC3 and MCF7 cells reversed in a different intensity the effect of the studied agents. According to the histological type of the cancer the antithrombotic efficiency of apixaban was preserved or partially reversed. Fondaparinux, was more vulnerable to the presence of cancer cells as compared to apixaban. The effect of BXCP3 or MCF7 cells on the antithrombotic potency of enoxaparin was of similar magnitude as that on apixaban. CONCLUSIONS: The type of cancer cells is determinant for the antithrombotic efficiency of the specific factor Xa inhibitors. In contrast it does not significantly influence the potency of enoxaparin. The present study shows that the impact of the type of cancer cells on the antithrombotic activity of the specific Xa inhibitors should not be neglected. This has to be taken into consideration for the design of dose-finding studies of the direct orally active FXa inhibitors in patients with different histological types of cancer.


Assuntos
Anticoagulantes/química , Neoplasias da Mama/imunologia , Enoxaparina/química , Fibrinolíticos/química , Neoplasias Pancreáticas/imunologia , Polissacarídeos/química , Pirazóis/química , Piridonas/química , Trombina/química , Plaquetas/citologia , Neoplasias da Mama/complicações , Linhagem Celular Tumoral , Fator Xa/química , Feminino , Fondaparinux , Heparina de Baixo Peso Molecular/química , Humanos , Concentração Inibidora 50 , Células MCF-7 , Neoplasias Pancreáticas/complicações , Trombose/prevenção & controle
6.
Clin Appl Thromb Hemost ; 21(8): 697-704, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26023170

RESUMO

BACKGROUND: The patent protection of low-molecular-weight heparins (LMWHs) expired, so the definition of criteria for the biological similarity between LMWH copies and the original product is a real need. AIM: The present in vitro study compared copies and branded enoxaparin using the specific anti-Xa activity and the calibrated automated thrombogram assay. METHODS: Samples of platelet-poor plasma (PPP) and platelet-rich plasma (PRP) from 15 healthy volunteers were spiked with branded enoxaparin (Lovenox) or its copies (Cutenox, Dilutol, Enoxa, Fibrinox, Loparin, Lupenox, Novex, Noxprin, and Versa). The specific anti-Xa activity was measured in PPP, and thrombin generation was assessed in PPP and PRP in the presence of tissue factor or pancreatic cancer cells BXPC3. RESULTS: The anti-Xa activity of enoxaparin copies ranged from 0.072 to 0.088 IU/µg, being lower as compared to the branded enoxaparin (0.095 anti-Xa IU/µg). The potency of each copy to inhibit thrombin generation varied in the 3 experimental systems. The presence of platelets or pancreatic cancer cells BXPC3 in human plasma induced significant modifications in the inhibitory efficiency of enoxaparin copies on thrombin generation, which distinguished them from the branded product. CONCLUSION: Enoxaparin copies showed significant variability regarding their inhibitory potency on thrombin generation. Platelets and cancer cells significantly increased the variability of the antithrombotic efficiency of the copies as compared to the branded enoxaparin. The present study underlines the need for the elaboration of additional functional criteria to evaluate the global antithrombotic capacity of enoxaparin copies in order to evaluate their potential sameness with the branded drug.


Assuntos
Medicamentos Genéricos/farmacologia , Enoxaparina/farmacologia , Fibrinolíticos/farmacologia , Neoplasias Pancreáticas/metabolismo , Trombina/metabolismo , Linhagem Celular Tumoral , Medicamentos Genéricos/farmacocinética , Enoxaparina/farmacocinética , Fibrinolíticos/farmacocinética , Humanos
7.
Curr Vasc Pharmacol ; 12(6): 893-902, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-22724467

RESUMO

Low molecular weight heparins (LMWHs) and fondaparinux are widely used for prophylaxis and treatment of venous thromboembolic disease in cancer patients. However, the optimization of the antithrombotic treatment especially in patients with adenocarcinoma of the pancreas is a challenging issue. The understanding of the mechanism of action of the LMWHs and fondaparinux in cancer-induced hypercoagulability might help to optimize antithrombotic treatment. To this aim, we investigated the influence of BXPC3 pancreas adenocarcinoma cells on the antithrombotic activity of LMWHs and fondaparinux. Thrombin generation (TG) in normal platelet poor (PPP) and platelet rich plasma (PRP) spiked with clinically relevant concentrations of dalteparin, enoxaparin, nadroparin tinzaparin and fondaparinux was assessed with the Calibrated Automated Thrombogram assay. BXPC3 (5 cells/µl) were added to plasma. The mean rate index (MRI) of the propagation phase of TG and the endogenous thrombin potential (ETP) were analyzed. The IC50 of the studied compounds were determined and compared on the basis of anti-Xa and anti-IIa equivalent units. We demonstrate that the specific antithrombin (AT)-dependent anti-Xa activity of LMWHs and fondaparinux almost selectively inhibits the propagation phase of TG. The synergy between the anti-Xa and anti-IIa activities of LMWHs rather than the selective inhibition of FXa warrants abrogation of TG. The mean molecular weight and anti-Xa/anti-IIa ratio of the AT-dependent agents cannot predict the alteration of their capacity to inhibit TG. Tinzaparin was the most potent inhibitor of TG than the other LMWHs. Enoxaparin was more potent than nadroparin and dalteparin.


Assuntos
Anticoagulantes/uso terapêutico , Heparina de Baixo Peso Molecular/uso terapêutico , Neoplasias Pancreáticas/metabolismo , Polissacarídeos/uso terapêutico , Trombina/antagonistas & inibidores , Trombina/metabolismo , Anticoagulantes/farmacologia , Fondaparinux , Heparina de Baixo Peso Molecular/farmacologia , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Polissacarídeos/farmacologia , Resultado do Tratamento
8.
Thromb Res ; 129(6): 779-86, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21917301

RESUMO

Cancer histology influences the risk of venous thromboembolism and tissue factor (TF) is the key molecule in cancer-induced hypercoagulability. We investigated the relation between TF expression by pancreatic and breast cancer cells (BXPC3 and MCF7 respectively) and their capacity to trigger in vitro thrombin generation in normal human plasma. Flow cytometry and Western blot analysis for TF expression were performed using murine IgG1 monoclonal antibody against human TF. Real-time PCR for TFmRNA was also performed. Activity of TF expressed by cancer cells was measured with a specific chromogenic assay. Thrombin generation in PPP was assessed using calibrated automated thrombogram. Cancer cells were added to platelet poor plasma from healthy volunteers. In separate experiments cells were incubated with the anti-TF antibody at concentration that completely neutralized the activity of recombinant human TF on thrombin generation. BXPC3 cells expressed significantly higher amounts of functional TF as compared to MCF7 cells. Incubation of BXPC3 and MCF7 cells with PPP resulted in acceleration of the initiation phase of thrombin generation. BXPC3 cells manifested higher procoagulant potential than MCF7 cells. The incubation of BXPC3 or MCF7 cells with the anti-TF monoclonal antibody which resulted in reversal of their effect on thrombin generation. The present study establishes a link between the amount of TF expressed by cancer cells with their procoagulant activity. Both studied types of cancer cells trigger thrombin generation but they have different procoagulant potential. The procoagulant activity of BXPC3 and MCF7 cells is related to the amount of TF expressed. Kinetic parameters of thrombogram are the most relevant for the detection of the TF-dependent procoagulant activity of cancer cells. TF expression is one of the mechanisms by which cancer cells manifest their procoagulant potential but it is not the unique one. The present experimental model will allow the characterization the procoagulant fingerprint of cell lines from the same or different histological types of cancer.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias Pancreáticas/metabolismo , Trombina/metabolismo , Tromboplastina/biossíntese , Tromboembolia Venosa/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Células MCF-7 , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Trombina/genética , Tromboplastina/genética , Tromboplastina/metabolismo , Tromboembolia Venosa/genética , Tromboembolia Venosa/patologia
9.
Glycobiology ; 14(4): 311-23, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15033938

RESUMO

The stromal cell-derived factor-1 (SDF-1) is a CXC chemokine, which plays critical roles in migration, proliferation, and differentiation of leukocytes. SDF-1 is the only known ligand of CXCR4, the coreceptor of X4 HIV strains. We show that SDF-1 binds to high- and low-affinity sites on HeLa cells. Coimmunoprecipitation studies demonstrate that glycanated and oligomerized syndecan-4 but neither syndecan-1, syndecan-2, betaglycan, nor CD44 forms complexes with SDF-1 and CXCR4 on these cells as well as on primary lymphocytes or macrophages. Moreover, biotinylated SDF-1 directly binds in a glycosaminoglycans (GAGs)-dependent manner to electroblotted syndecan-4, and colocalization of SDF-1 with syndecan-4 was visualized by confocal microscopy. Glycosaminidases pretreatment of the HeLa cells or the macrophages decreases the binding of syndecan-4 to the complex formed by it and SDF-1. In addition, this treatment also decreases the binding of the chemokine to CXCR4 on the primary macrophages but not on the HeLa cells. Therefore GAGs-dependent binding of SDF-1 to the cells facilitates SDF-1 binding to CXCR4 on primary macrophages but not on HeLa cell line. Finally, an SDF-1-independent heteromeric complex between syndecan-4 and CXCR4 was visualized on HeLa cells by confocal microscopy as well as by electron microscopy. Moreover, syndecan-4 from lymphocytes, monocyte derived-macrophages, and HeLa cells coimmunoprecipitated with CXCR4. This syndecan-4/CXCR4 complex is likely a functional unit involved in SDF-1 binding. The role of these interactions in the pathophysiology of SDF-1 deserves further study.


Assuntos
Quimiocinas CXC/metabolismo , Linfócitos/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteoglicanas/metabolismo , Receptores CXCR4/metabolismo , Quimiocina CXCL12 , Fluorescência , Expressão Gênica , Células HeLa , Humanos , Complexos Multiproteicos/metabolismo , Ligação Proteica , Proteoglicanas/biossíntese , Sindecana-4
10.
Biochim Biophys Acta ; 1617(1-2): 80-8, 2003 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-14637022

RESUMO

Interaction of RANTES with its membrane ligands or receptors transduces multiple intracellular signals. Whether RANTES uses proteoglycans (PGs) belonging to the syndecan family to attach to primary cells expressing RANTES G-protein-coupled receptors (GPCRs) was investigated. We demonstrate that RANTES specifically binds to high and low affinity binding sites on human monocyte-derived macrophages (MDM). We show by co-immunoprecipitation experiments that RANTES is associated on these cells with syndecan-1 and syndecan-4, but neither with syndecan-2 nor with betaglycan, in addition to CD44 and its GPCRs, CCR5 and CCR1. Glycosaminidases pre-treatment of the monocyte derived-macrophages strongly decreases the binding of RANTES to syndecan-1 and syndecan-4 and also to CCR5, and abolishes RANTES binding to CD44. This suggests that glycosaminoglycans (GAGs) are involved in RANTES binding to the PGs and that such bindings facilitate the subsequent interaction of RANTES with CCR5, on the MDM, characterized by low membrane expression of CCR5. The role of these interactions in the pathophysiology of RANTES deserves further study.


Assuntos
Membrana Celular/química , Quimiocina CCL5/química , Macrófagos/química , Macrófagos/metabolismo , Glicoproteínas de Membrana/química , Proteoglicanas/química , Membrana Celular/metabolismo , Quimiocina CCL5/metabolismo , Humanos , Leucócitos Mononucleares/química , Leucócitos Mononucleares/metabolismo , Glicoproteínas de Membrana/biossíntese , Ligação Proteica , Proteoglicanas/biossíntese , Sindecana-1 , Sindecana-4 , Sindecanas
11.
Glycobiology ; 13(9): 623-34, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12773479

RESUMO

It is believed that proteoglycans influence biological properties of chemokines. We show that the CC chemokine RANTES binds not only to high-affinity binding sites on CCR5-positive HeLa cells but also to low-affinity binding sites on HeLa cells expressing or lacking RANTES G protein-coupled receptors. Coimmunoprecipitation studies demonstrate that RANTES forms complexes with glycanated syndecan (SD)-1 and -4, in addition to CCR5 on the CCR5-positive HeLa cells. Moreover, confocal microscopy analysis shows the colocalization of RANTES with SD-1 and -4. Glycosaminoglycans removal from the cells by glycosaminidases treatment prevented RANTES binding to SD-1 and -4 and decreased RANTES binding to CCR5 on the CCR5-positive HeLa cells. Removal of glycosaminoglycans by glycosaminidases treatment of the complexes, RANTES/SD-1/SD-4/+/-CCR5, immobilized on beads, reversed SD-1 and -4 bindings. Therefore, RANTES bindings to SD-1 and -4 depend on glycosaminoglycans and facilitate RANTES interaction with CCR5. Extracting plasma membrane cholesterol abolished the coimmunoprecipitation of SD-1 with RANTES, suggesting that rafts are involved in RANTES association to SD-1. Confocal microscopy analysis as well as coimmunoprecipitation experiments show a RANTES-independent heteromeric complex on the CCR5-positive HeLa cells, SD-1, SD-4, and CCR5. This complex is likely a functional unit in which proteoglycans may modulate RANTES binding to CCR5.


Assuntos
Quimiocina CCL5/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteoglicanas/metabolismo , beta-Ciclodextrinas , Membrana Celular/metabolismo , Ciclodextrinas , Imunofluorescência , Células HeLa , Humanos , Testes de Precipitina , Ligação Proteica , Receptores CCR5/metabolismo , Sindecana-1 , Sindecana-4 , Sindecanas
12.
Biochem Biophys Res Commun ; 296(3): 507-14, 2002 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-12176010

RESUMO

We have previously reported that alpha-fetoprotein (AFP) inhibits infection of human monocyte-derived macrophages (MDM) by R5-HIV-1 strains and that a peptide mimicking the clade B HIV-1 gp120 consensus V3 domain (V3Cs) binds to CCR5. We demonstrate here that AFP binds high- and low-affinity binding sites of MDM, characterized, respectively, by 5.15 and 100nM K(d) values. Heat denaturation or neuraminidase treatment of AFP inhibits this binding, suggesting the involvement of protein-protein and lectin-carbohydrate interactions. Moreover, AFP displaces V3Cs binding to MDM. In addition, MIP-1beta, the most specific CCR5 ligand, displaces AFP binding to MDM (IC(50)=4.3nM). Finally, we demonstrate that AFP binds to a ligand of HIV-gp120 V3Cs domain, CCR5, expressed by MDM and by HeLa cells expressing CCR5. Such binding is not observed in the presence of HeLa cells lacking CCR5. The present results provide strong evidence that AFP directly binds to CCR5 expressed by human primary macrophages and by transfected CCR5+ HeLa cells.


Assuntos
Macrófagos/metabolismo , alfa-Fetoproteínas/metabolismo , Sítios de Ligação , Ligação Competitiva , Células Cultivadas , Proteína gp120 do Envelope de HIV/química , Células HeLa , Humanos , Monócitos/fisiologia , Peptídeos/metabolismo , Receptores CCR5/metabolismo
13.
FEBS Lett ; 514(2-3): 209-13, 2002 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-11943153

RESUMO

CXCR4 is a coreceptor, along with CD4, for human immunodeficiency virus type 1 (HIV-1). Trimolecular complexes between HIV-1 glycoprotein (gp)120, CD4 and CXCR4 constitute a prerequisite for HIV entry. We studied whether CD4 is associated with CXCR4 on CD4+ CXCR4+ cells. Using the conformation-dependent anti-CXCR4 mAb 12G5, CD4 was coimmunoprecipitated with CXCR4 from the membrane of U937 cells which support HIV-1(LAI) efficient infection, and from that of peripheral blood lymphocytes (PBL). CD4 association with CXCR4 increased upon PBL coculture for 5 days with autologous monocytes, decreased upon treatment of the cells or the CD4-CXCR4 complex with either N-glycanase or stromal cell derived factor-1alpha (SDF-1alpha) and was abolished by incubation of the cells with both, N-glycanase and SDF-1alpha. This indicates that glycans are partly involved in CD4 association with CXCR4 and may partly explain the inhibitory effect of SDF-1alpha on HIV infection.


Assuntos
Antígenos CD4/metabolismo , Membrana Celular/metabolismo , Quimiocinas CXC/metabolismo , Polissacarídeos/metabolismo , Receptores CXCR4/metabolismo , Amidoidrolases/metabolismo , Amidoidrolases/farmacologia , Anticorpos Monoclonais/metabolismo , Membrana Celular/química , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/farmacologia , Técnicas de Cocultura , Humanos , Linfócitos/citologia , Linfócitos/metabolismo , Substâncias Macromoleculares , Monócitos/citologia , Monócitos/metabolismo , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase , Testes de Precipitina , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...