Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 483
Filtrar
1.
Thromb Haemost ; 105(6): 1053-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21544310

RESUMO

The physiologic activation of the plasma kallikrein-kinin system requires the assembly of its constituents on a cell membrane. High- molecular-weight kininogen (HK) and cleaved HK (HKa) both interact with at least three endothelial cell binding proteins: urokinase plasminogen activator receptor (uPAR), globular C1q receptor (gC1qR,) and cytokeratin 1 (CK1). The affinity of HK and HKa for endothelial cells are KD=7-52 nM. The contribution of each protein is unknown. We examined the direct binding of HK and HKa to the soluble extracellular form of uPAR (suPAR), gC1qR and CK1 using surface plasmon resonance. Each binding protein linked to a CM-5 chip and the association, dissociation and KD (equilibrium constant) were measured. The interaction of HK and HKa with each binding protein was zinc-dependent. The affinity for HK and HKa was gC1qR>CK1>suPAR, indicating that gC1qR is dominant for binding. The affinity for HKa compared to HK was the same for gC1qR, 2.6-fold tighter for CK1 but 53-fold tighter for suPAR. Complex between binding proteins was only observed between gC1qR and CK1 indicating that a binary CK1-gC1qR complex can form independently of kininogen. Although suPAR has the weakest affinity of the three binding proteins, it is the only one that distinguished between HK and HKa. This finding indicates that uPAR may be a key membrane binding protein for differential binding and signalling between the cleaved and uncleaved forms of kininogen. The role of CK1 and gC1qR may be to initially bind HK to the membrane surface before productive cleavage to HKa.


Assuntos
Queratinas/metabolismo , Cininogênio de Alto Peso Molecular/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Complemento/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ressonância de Plasmônio de Superfície , Coagulação Sanguínea , Endotélio/metabolismo , Humanos , Queratina-1/metabolismo , Queratinas/química , Cininogênio de Alto Peso Molecular/química , Glicoproteínas de Membrana/química , Ligação Proteica , Receptores de Complemento/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Transdução de Sinais
2.
J Thromb Haemost ; 8(1): 185-93, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19874467

RESUMO

BACKGROUND AND OBJECTIVE: Endothelial progenitor cells (EPCs) contribute to postnatal neovascularization, thus promoting wide interest in their therapeutic potential in vascular injury and prevention of their dysfunction in cardiovascular diseases. Cleaved high molecular weight kininogen (HKa), an activation product of the plasma kallikrein-kinin system (KKS), inhibits the functions of differentiated endothelial cells including in vitro and in vivo angiogenesis. In this study, our results provided the first evidence that HKa is able to target EPCs and inhibits their tube forming capacity. METHODS AND RESULTS: We determined the effect of HKa on EPCs using a three-dimensional vasculogenesis assay. Upon stimulation with vascular endothelial growth factor (VEGF) alone, EPCs formed vacuoles and tubes, and differentiated into capillary-like networks. As detected by gelatinolytic activity assay, VEGF stimulated secretion and activation of matrix metallopeptidase 2 (MMP-2), but not MMP-9, in the conditioned medium of 3D culture of EPCs. Specific inhibition or gene ablation of MMP-2, but not MMP-9, blocked the vacuole and tube formation by EPCs. Thus, MMP-2 is selectively required for EPC vasculogenesis. In a concentration-dependent manner, HKa significantly inhibited tube formation by EPCs and the conversion of pro-MMP-2 to MMP-2. Moreover, HKa completely blocked the association between pro-MMP-2 and alphavbeta3 integrin, and its inhibition of MMP-2 activation was dependent on the presence of alphavbeta3 integrin. In a purified system, HKa did not directly inhibit MMP-2 activity. CONCLUSIONS: HKa inhibits tube forming capacity of EPCs by suppression of MMP-2 activation, which may constitute a novel link between activation of the KKS and EPC dysfunction.


Assuntos
Células Endoteliais/enzimologia , Cininogênio de Alto Peso Molecular/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Neovascularização Fisiológica , Células-Tronco/enzimologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Regulação para Baixo , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática , Precursores Enzimáticos/metabolismo , Gelatinases/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Neovascularização Fisiológica/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Interferência de RNA , Células-Tronco/efeitos dos fármacos , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Oncogene ; 28(30): 2756-65, 2009 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-19483730

RESUMO

Upregulation and activation of epidermal growth factor receptor and/or urokinase-type plasminogen activator receptor in a variety of cancers have been shown to be associated with poor prognosis. High-molecular-weight kininogen can be hydrolysed by plasma kallikrein to bradykinin and cleaved high-molecular-weight kininogen (HKa). HKa and its domain 5 (D5) both have been shown to have potent anti-angiogenic activity. We now show that HKa blocks human prostate cancer cell (DU145) migration by 76.0+/-2.4% at 300 nM and invasion by 78.0+/-12.9% at 11.1 nM. D5 inhibits tumor migration and invasion in a concentration-dependent manner. Stimulation by basic fibroblast growth factor (bFGF) or vascular endothelial growth factor results in clustering of urokinase-type plasminogen activator receptor (uPAR) and epidermal growth factor receptor (EGFR) on the surface of DU145 cells. The co-localization of uPAR and EGFR is prevented by HKa. Immunoprecipitation suggests that uPAR, EGFR and alpha5beta1 integrin formed a ternary complex. Immunoblotting shows that HKa significantly decreases the bFGF-transactivated phosphorylation of EGFR at Tyr 1173 between 30 min and 4 h. The phosphorylation of extracellular signal-regulated kinase (ERK) and AKT, which are downstream effectors of EGFR, is also inhibited by HKa. These novel data indicate that HKa and D5 inhibit migration and invasion of human prostate cancer cells through an EGFR/uPAR pathway, suggesting the therapeutic potential of HKa and D5 to decrease metastasis of human prostate cancer.


Assuntos
Receptores ErbB/fisiologia , Cininogênio de Alto Peso Molecular/farmacologia , Neoplasias da Próstata/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Cininogênio de Alto Peso Molecular/química , Cininogênio de Alto Peso Molecular/uso terapêutico , Masculino , Invasividade Neoplásica , Neoplasias da Próstata/tratamento farmacológico , Estrutura Terciária de Proteína , Quinazolinas , Receptores de Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Transdução de Sinais/efeitos dos fármacos , Tirfostinas/farmacologia
4.
J Thromb Haemost ; 5(2): 403-11, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17155949

RESUMO

BACKGROUND: Domain 5 (D5) of kininogen inhibits endothelial cell adhesion, migration, proliferation and angiogenesis by inducing apoptosis and disrupting a signaling pathway initiated by binding to the urokinase receptor (uPAR). OBJECTIVES: Because tumor cells frequently overexpress uPAR, we hypothesized that D5 can directly inhibit proliferation of colon carcinoma cells. METHODS AND RESULTS: A recombinant fusion protein of D5 and glutathione S-transferase (GST-D5) but not GST at 280 nm inhibited proliferation of human colon carcinoma cells (HCT-116) in vitro by 75-86%. We found that treatment with GST-D5 did not affect the survival pathway, phosphatidylinositol 3-kinase or the apoptotic pathway. In contrast, the G1/S phase transition of the cell cycle was downregulated as evidenced by an increase of cells in G0/G1 and a decrease in cells in S by flow cytometry. We found a decrease in serine phosphorylation of the retinoblastoma protein Rb (p107) after incubation with GST-D5. Less E2F-1 transcription factor and p107 were released and fewer cells overcame the G1/S growth restriction point. Expression levels of cyclins D1, A and E were reduced as measured by densiometric analysis of western blots. Cyclin-dependent protein kinase activities were downregulated and p27, the cyclin-dependent kinase inhibitor, was activated by GST-D5. CONCLUSIONS: These findings indicate that D5 of high molecular weight kininogen interferes with the G1 to S phase transition, reducing the proliferation of human colon carcinoma cells.


Assuntos
Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/patologia , Interfase/efeitos dos fármacos , Cininogênios/fisiologia , Fragmentos de Peptídeos/farmacologia , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/análise , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Fase G1 , Humanos , Cininogênios/química , Proteínas Recombinantes de Fusão/farmacologia , Proteína do Retinoblastoma/metabolismo , Fase S
5.
Dig Liver Dis ; 37(9): 665-73, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15949977

RESUMO

BACKGROUND: Tissue kallikrein and its natural inhibitor, kallistatin, play opposite roles in the generation of bradykinin, a potent mediator of inflammation. Observations on experimental models and humans with ulcerative colitis suggest a pathogenetic role of the kallikrein-kinin system in inflammatory bowel diseases. AIM: To evaluate tissue kallikrein and kallistatin in intestinal tissue samples from Crohn's disease and ulcerative colitis patients with different degrees of disease involvement. PATIENTS AND METHODS: Full-thickness surgical intestinal samples were obtained from 144 subjects (38 normal controls, 32 inflammatory controls, 38 Crohn's disease, 36 ulcerative colitis) and tested for kallikrein and kallistatin by immunoperoxidase techniques. RESULTS: Compared with controls, kallikrein immunoreactivity was significantly weaker in goblet cells (p=0.0001) and significantly stronger in interstitium (p=0.0001) of the Crohn's disease and ulcerative colitis samples. Kallistatin colocalised with kallikrein, with almost no reactivity in goblet cells but strong reactivity in interstitium of inflammatory bowel disease patients (p=0.0001 versus controls). The kallikrein and kallistatin depletion of goblet cells and the increased interstitial kallikrein and kallistatin reactivity correlated with the degree of tissue inflammation (p=0.0001). Disease-free samples had normal kallikrein and kallistatin patterns. CONCLUSIONS: Kallikrein-kinin system is actively involved in inflammatory bowel disease as a result of the release of kallikrein in the intestinal extracellular space; this involvement correlates with the degree of tissue inflammation. The normal pattern observed in the disease-free samples seems to rule out a genetic defect of kallikrein and kallistatin in inflammatory bowel diseases.


Assuntos
Doenças Inflamatórias Intestinais/patologia , Sistema Calicreína-Cinina , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas de Transporte/análise , Colo/química , Colo/patologia , Feminino , Humanos , Imuno-Histoquímica , Doenças Inflamatórias Intestinais/fisiopatologia , Calicreínas/análise , Masculino , Pessoa de Meia-Idade , Serpinas/análise
6.
J Thromb Haemost ; 3(4): 670-6, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15733059

RESUMO

High-molecular-weight kininogen (HK) is a plasma protein that possesses multiple physiological functions. Originally identified as a precursor of bradykinin, a bioactive peptide that regulates many cardiovascular processes, it is now recognized that HK plays important roles in fibrinolysis, thrombosis, and inflammation. HK binds to endothelial cells where it can be cleaved by plasma kallikrein to release bradykinin (BK). The remaining portion of the molecule, cleaved HK, is designated cleaved high-molecular-weight kininogen or HKa. While BK has been intensively studied, the physiological implication of the generation of HKa is not clear. Recent studies have revealed that HKa inhibits angiogenesis while BK promotes angiogenesis. These findings represent novel functions of the kallikrein-kinin system that have not yet been fully appreciated. In this review, we will briefly discuss the recent progress in the studies of the molecular mechanisms that mediate the antiangiogenic effect of HKa and the proangiogenic activity of BK.


Assuntos
Bradicinina/fisiologia , Sistema Calicreína-Cinina , Cininogênio de Alto Peso Molecular/química , Cininogênio de Alto Peso Molecular/fisiologia , Neovascularização Fisiológica , Inibidores da Angiogênese/química , Animais , Adesão Celular , Ciclo Celular , Membrana Celular/metabolismo , Proliferação de Células , Humanos , Camundongos , Modelos Biológicos , Neovascularização Patológica , Peptídeos/química , Estrutura Terciária de Proteína
7.
FASEB J ; 18(7): 887-9, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15001555

RESUMO

Bradykinin (BK), a vasoactive, proinflammatory nonapeptide, promotes cell adhesion molecule (CAM) expression, leukocyte sequestration, inter-endothelial gap formation, and protein extravasation in postcapillary venules. These effects are mediated by bradykinin-1 (B1R) and-2 (B2R) receptors. We delineated some of the mechanisms by which BK could influence chronic inflammation by altering CAM expression on leukocytes, endothelium, and synovium in joint sections of peptidoglycan-polysaccharide-injected Lewis rats. Blocking B1R results in significantly increased joint inflammation. Immunohistochemistry of the B1R antagonist group revealed increased leukocyte and synovial CD11b and CD54 expression and increased CD11b and CD44 endothelial expression. B2R antagonism decreased leukocyte and synovial CD44 and CD54 and endothelial CD11b expression. Although these findings implicate B2R involvement in the acute phase of inflammation by facilitating leukocyte activation (CD11b), homing (CD44), and transmigration (CD54). Treatment with a B2R antagonist did not affect the disease evolution in this model. In contrast, when both BK receptors are blocked, the aggravation of inflammation by B1R blockade is neutralized and there is no difference from the disease-untreated model. Our findings suggest that B1R and B2R signaling show physiologic antagonism. B1R signaling suggests involvement in down-regulation of leukocyte activation, transmigration, and homing. Further studies are needed to evaluate the B1 receptor agonist's role in this model.


Assuntos
Artrite/metabolismo , Bradicinina/fisiologia , Moléculas de Adesão Celular/biossíntese , Endotélio Vascular/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Membrana Sinovial/efeitos dos fármacos , Animais , Artrite/induzido quimicamente , Artrite/genética , Artrite Reumatoide/metabolismo , Bradicinina/análogos & derivados , Bradicinina/biossíntese , Bradicinina/genética , Bradicinina/farmacologia , Antagonistas de Receptor B1 da Bradicinina , Antagonistas de Receptor B2 da Bradicinina , Antígeno CD11b/biossíntese , Antígeno CD11b/genética , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/genética , Quimiotaxia de Leucócito/efeitos dos fármacos , Modelos Animais de Doenças , Endotélio Vascular/patologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/genética , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Selectina L/biossíntese , Selectina L/genética , Leucócitos/metabolismo , Masculino , Oligopeptídeos/farmacologia , Peptidoglicano/toxicidade , Pré-Calicreína/análise , Ratos , Ratos Endogâmicos Lew , Receptor B1 da Bradicinina/fisiologia , Receptor B2 da Bradicinina/fisiologia , Organismos Livres de Patógenos Específicos , Membrana Sinovial/irrigação sanguínea , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia
8.
J Thromb Haemost ; 1(8): 1791-8, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12911595

RESUMO

We previously localized the heparin binding region on high molecular weight kininogen to domain 5 (D5) by quantifying the binding using surface plasmon resonance of D5 fused at its N-terminal to glutathione-S-transferase. We further examined GST-(H475-S626) which at 100 nm was previously shown to be ineffective in reversing the heparin acceleration of antithrombin inhibition of thrombin. However, we now show that at a concentration of 400 nm, complete reversal of accelerated inhibition occurred. To characterize the interacting sequences on D5, four peptides representing surface loops of a molecular model were synthesized. Peptides H475-H485 and G440-G455, rich in histidine and low in lysine, showed weak or no detectable binding in the absence of Zn++, but tighter binding in the presence of Zn++. H483-K497 containing three histidines and six lysines showed tight binding without Zn++, and increased in avidity with Zn++. In contrast, G486-K502, low in histidine and high in lysine, showed tight binding (KD = 0.8 microm) in the absence and presence of Zn++. Both H483-K497 and G486-K502 were effective in neutralizing the accelerated inhibition by heparin of thrombin by antithrombin in the absence of Zn++. Therefore, a set of lysine residues in the sequence of K487-K502 is responsible for Zn++-independent binding of heparin. Further, a group of histidine residues in sequence range of H475-H485 contributes to Zn++-dependent binding of heparin to HK-D5.


Assuntos
Heparina/química , Cininogênio de Alto Peso Molecular/química , Zinco/química , Sequência de Aminoácidos , Antitrombinas/química , Biotinilação , Relação Dose-Resposta a Droga , Ácido Edético/farmacologia , Deleção de Genes , Glutationa Transferase/metabolismo , Histidina/química , Humanos , Cinética , Cininogênio de Alto Peso Molecular/genética , Lisina/química , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície , Trombina/química , Fatores de Tempo
9.
J Thromb Haemost ; 1(1): 164-70, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12871554

RESUMO

Previously we demonstrated that domain 5 (D5) of high-molecular-weight kininogen (HK) inhibits neovascularization in the chicken chorioallantoic membrane (CAM) assay and further found that kallikrein cleaved HK (HKa) inhibited FGF2-and VEGF-induced neovascularization, and thus was antiangiogenic. In this study, we sought to demonstrate whether uncleaved HK stimulates neovascularization and thus is proangiogenic. The chick chorioallantoic membrane was used as an in ovo assay of angiogenesis. Low-molecular-weight kininogen stimulates angiogenesis, indicating that D5 is not involved. Bradykinin stimulates neovascularization equally to HK and LK and is likely to be responsible for the effect of HK. A murine monoclonal antibody to HK (C11C1) also recognizes a similar component in chicken plasma as detected by surface plasmon resonance. Angiogenesis induced by FGF2 and VEGF is inhibited by this monoclonal antibody and is a more potent inhibitor of neovascularization induced by VEGF than an integrin alphavbeta3 antibody (LM 609). Our postulate that C11C1 inhibits the stimulation of angiogenesis by HK was confirmed when either C11C1 or D5 completely inhibited angiogenesis in the CAM induced by HK. Growth of human fibrosarcoma (HT-1080) on the CAM was inhibited by GST-D5 and C11C1. These results indicate HK is proangiogenic probably by releasing bradykinin and that a monoclonal antibody directed to HK could serve as an antiangiogenic agent with a potential for inhibiting tumor angiogenesis and other angiogenesis-mediated disorders.


Assuntos
Inibidores da Angiogênese/farmacologia , Anticorpos Monoclonais/farmacologia , Cininogênio de Alto Peso Molecular/antagonistas & inibidores , Neovascularização Fisiológica/imunologia , Alantoide/irrigação sanguínea , Animais , Anticorpos Monoclonais/imunologia , Bradicinina/farmacologia , Embrião de Galinha , Córion/irrigação sanguínea , Relação Dose-Resposta a Droga , Fatores de Crescimento Endotelial/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibrossarcoma/metabolismo , Glutationa Transferase/genética , Glutationa Transferase/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Cininogênio de Alto Peso Molecular/imunologia , Cininogênio de Alto Peso Molecular/farmacologia , Cininogênio de Baixo Peso Molecular/farmacologia , Linfocinas/farmacologia , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Ressonância de Plasmônio de Superfície , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
10.
Inflamm Res ; 52(2): 86-93, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12665127

RESUMO

OBJECTIVE AND DESIGN: The goal of this study is to investigate the consequence of the interaction between Mac-1 and uPAR and determine the mechanisms by which uPAR regulates Mac-1 dependent adhesion to fibrinogen. MATERIAL: Human embryonic kidney 293 cells transfected with Mac-1 or uPAR or co-transfected with both Mac-1 and uPAR. METHODS: Cell adhesion and binding assays and Western Blotting for protein tyrosine phosphorylation analysis. RESULTS: The adhesion to fibrinogen was increased two-fold for Mac-1-uPAR co-transfected cells comparing to the Mac-1 transfected cells alone. The increased adhesion was inhibited when cells were treated with phosphatidylinositol-specific phospholipase C to remove uPAR. Occupancy of uPAR with urokinase-type plasminogen activator further enhanced the cell adhesion to fibrinogen. Phosphorylation of focal adhesion kinase (FAK) and mitogen-activated protein kinase (MAPK) was increased in Mac-1-uPAR co-transfected cells but not in Mac-1 transfected cells. CONCLUSIONS: uPAR up-regulated the Mac-1 adhesion to fibrinogen and FAK and MAPK were involved in this regulation.


Assuntos
Antígeno CD11b/fisiologia , Antígenos CD18/fisiologia , Fibrinogênio/fisiologia , Antígeno de Macrófago 1/fisiologia , Receptores de Superfície Celular/fisiologia , Adesão Celular/fisiologia , Linhagem Celular , Humanos , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transdução de Sinais/fisiologia , Transfecção , Regulação para Cima , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
11.
Mol Pharmacol ; 62(3): 514-20, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12181427

RESUMO

To identify amino acid residues involved in PDE3-selective inhibitor binding, we selected eight presumed interacting residues in the substrate-binding pocket of PDE3A using a model created on basis of homology to the PDE4B crystal structure. We changed the residues to alanine using site-directed mutagenesis technique, expressed the mutants in a baculovirus/Sf9 cell system, and analyzed the kinetic characteristics of inhibition of the mutant enzymes by milrinone and cilostazol, specific inhibitors of PDE3. The mutants displayed differential sensitivity to the inhibitors. Mutants Y751A, D950A, and F1004A had reduced sensitivity to milrinone (K(i) changed from 0.66 microM for the recombinant PDE3A to 7.5 to 156 microM for the mutants), and diminished sensitivity to cilostazol (K(i) of the mutants were 18- to 371-fold higher than that of the recombinant PDE3A). In contrast, the mutants T844A, F972A and Q975A showed increased K(i) for cilostazol but no difference for milrinone from the recombinant PDE3A. Molecular models show that the PDE3 inhibitors cilostazol and milrinone share some of common residues but interact with distinct residues at the active site, suggesting that selective inhibitors can be designed with flexible size against PDE3 active site. Our study implies that highly conserved residuals Y751, D950 and F1004 in the PDE families are key residues for binding of both substrate and inhibitors, and nonconserved T844 may be responsible for the cilostazol selectivity of PDE3A. Detailed knowledge of the structure of inhibitory sites should contribute to development of more potent and specific inhibitory drugs.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Milrinona/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Tetrazóis/farmacologia , 3',5'-AMP Cíclico Fosfodiesterases/efeitos dos fármacos , 3',5'-AMP Cíclico Fosfodiesterases/genética , Sítios de Ligação , Cilostazol , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida
12.
Thromb Haemost ; 87(5): 873-9, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12038792

RESUMO

Platelets contain two cAMP phosphodiesterases (PDEs) which regulate intracellular cAMP levels, cGMP-inhibited cAMP PDE (PDE3A) and cGMP-stimulated PDE (PDE2A). Using the PDE3 inhibitor, milrinone and the PDE2 inhibitor, erythro-9-(2-hydroxyl-3-nonyl)adenine (EHNA), we have explored the contribution of each PDE to the regulation of platelet function. Inhibition of PDE2 resulted in higher levels of intracellular cAMP than inhibition of PDE3A suggesting this PDE may be the more important regulator of cAMP in human platelets. However, a concentration-dependent inhibition of agonist-induced aggregation was observed with milrinone while little effect was seen with EHNA. In addition, we observed a concentration-dependent inhibition in the increase of intracellular Ca2+ with PDE3 inhibition and significantly less with PDE2 inhibition. PDE3 inhibition also resulted in a concentration-dependent increase in cAMP-mediated phosphorylation of the vasodilator-stimulated phospho-protein (VASP) whereas there was no significant increase with PDE2 inhibition. In each of these experiments, synergism was noted with the combination of milrinone and EHNA. These results suggest that cAMP pools may be localized and the various PDEs regulate specific pools. These data also suggest that inhibitors of PDE3A may be more effective antiplatelet agents.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/fisiologia , Adenina/análogos & derivados , Plaquetas/enzimologia , AMP Cíclico/fisiologia , Diester Fosfórico Hidrolases/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Sistemas do Segundo Mensageiro/fisiologia , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , Adenina/farmacologia , Adulto , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Cálcio/sangue , Sinalização do Cálcio/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , GMP Cíclico/farmacologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2 , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Inibidores Enzimáticos/farmacologia , Humanos , Proteínas dos Microfilamentos , Milrinona/farmacologia , Fosfoproteínas/metabolismo , Fosforilação , Inibidores da Agregação Plaquetária/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Sistemas do Segundo Mensageiro/efeitos dos fármacos
13.
FASEB J ; 15(13): 2365-76, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11689462

RESUMO

Proteolytic cleavage of single-chain, high molecular weight kininogen (HK) by kallikrein releases the short-lived vasodilator bradykinin and leaves behind a two-chain, high molecular weight kininogen (HKa) reported to bind to the beta2-integrin Mac-1 (CR3, CD11b/CD18, alphaMbeta2) on neutrophils and exert antiadhesive properties by binding to the urokinase receptor (uPAR) and vitronectin. We define the molecular mechanisms for the antiadhesive effects of HK related to disruption of beta2-integrin-mediated cellular interactions in vitro and in vivo. In a purified system, HK and HKa inhibited the binding of soluble fibrinogen and ICAM-1 to immobilized Mac-1, but not the binding of ICAM-1 to immobilized LFA-1 (CD11a/CD18, alphaLbeta2). This inhibitory effect could be attributed to HK domain 5 and to a lesser degree to HK domain 3, consistent with the requirement of both domains for binding to Mac-1. Accordingly, HK, HKa, and domain 5 inhibited the adhesion of Mac-1 but not LFA-1-transfected K562 human erythroleukemic cells to ICAM-1. Moreover, adhesion of human monocytic cells to fibrinogen and to human endothelial cells was blocked by HK, HKa, and domain 5. By using peptides derived from HK domain 5, the sequences including amino acids H475-G497 (and to a lesser extent, G440-H455) were identified as responsible for the antiadhesive effect, which was independent of uPAR. Finally, administration of domain 5 into mice, followed by induction of thioglycollate-provoked peritonitis, decreased the recruitment of neutrophils by approximately 70% in this model of acute inflammation. Taken together, HKa (and particularly domain 5) specifically interacts with Mac-1 but not with LFA-1, thereby blocking Mac-1-dependent leukocyte adhesion to fibrinogen and endothelial cells in vitro and in vivo and serving as a novel endogenous regulator of leukocyte recruitment into the inflamed tissue.


Assuntos
Cininogênio de Alto Peso Molecular/farmacologia , Leucócitos/efeitos dos fármacos , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Ligação Competitiva/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Fibrinogênio/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Células K562 , Cininogênio de Alto Peso Molecular/química , Leucócitos/citologia , Leucócitos/metabolismo , Antígeno-1 Associado à Função Linfocitária/genética , Antígeno-1 Associado à Função Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Infiltração de Neutrófilos/efeitos dos fármacos , Peritonite/induzido quimicamente , Peritonite/patologia , Peritonite/prevenção & controle , Plasmídeos/genética , Tioglicolatos/administração & dosagem , Células U937
14.
Arterioscler Thromb Vasc Biol ; 21(9): 1427-33, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11557667

RESUMO

We recently reported that domain 5 (D5) of high-molecular-weight kininogen inhibited critical steps required for angiogenesis. Thus, it was named kininostatin. To understand its mechanism of action, we further investigated the effects of D5 on basic fibroblast growth factor (bFGF)-induced endothelial cell proliferation and cell viability. We report here that D5-inhibited cell proliferation of human endothelial cells stimulated by bFGF was associated with a significant reduction of cyclin D1 expression, which is a critical component required for the transition from G(1) to S phase of the cell cycle. However, inhibition of cell proliferation by D5 was not due to an inhibition of extracellular signal-regulated protein kinase activity. Endothelial cells underwent apoptosis when cultured in a serum-free medium, which was prevented by bFGF. D5 reversed the protective effect of bFGF by 80%. Cells treated with D5 in the presence of bFGF showed typical morphological features of apoptosis, which was further confirmed by 2 additional assays: Hoechst 33258 cell staining and DNA fragmentation analysis. We conclude that the inhibition of endothelial cell proliferation and induction of apoptosis together represent a major contribution to the antiangiogenic activity of D5.


Assuntos
Inibidores da Angiogênese/farmacologia , Apoptose , Endotélio Vascular/citologia , Cininogênio de Alto Peso Molecular/química , Cininogênio de Alto Peso Molecular/farmacologia , Neovascularização Fisiológica , Inibidores da Angiogênese/química , Divisão Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Sobrevivência Celular/efeitos dos fármacos , Ciclina D1/metabolismo , DNA/biossíntese , Fragmentação do DNA , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estrutura Terciária de Proteína
15.
Protein Sci ; 10(8): 1481-9, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11468344

RESUMO

Cyclic nucleotide phosphodiesterase 3A (PDE3A) hydrolyzes cAMP to AMP, but is competitively inhibited by cGMP due to a low k(cat) despite a tight K(m). Cyclic AMP elevation is known to inhibit all pathways of platelet activation, and thus regulation of PDE3 activity is significant. Although cGMP elevation will inhibit platelet function, the major action of cGMP in platelets is to elevate cAMP by inhibiting PDE3A. To investigate the molecular details of how cGMP, a similar but not identical molecule to cAMP, behaves as an inhibitor of PDE3A, we constructed a molecular model of the catalytic domain of PDE3A based on homology to the recently determined X-ray crystal structure of PDE4B. Based on the excellent fit of this model structure, we mutated nine amino acids in the putative catalytic cleft of PDE3A to alanine using site-directed mutagenesis. Six of the nine mutants (Y751A, H840A, D950A, F972A, Q975A, and F1004A) significantly decreased catalytic efficiency, and had k(cat)/K(m) less than 10% of the wild-type PDE3A using cAMP as substrate. Mutants N845A, F972A, and F1004A showed a 3- to 12-fold increase of K(m) for cAMP. Four mutants (Y751A, H840A, D950A, and F1004A) had a 9- to 200-fold increase of K(i) for cGMP in comparison to the wild-type PDE3A. Studies of these mutants and our previous study identified two groups of amino acids: E866 and F1004 contribute commonly to both cAMP and cGMP interactions while N845, E971, and F972 residues are unique for cAMP and the residues Y751, H836, H840, and D950 interact with cGMP. Therefore, our results provide biochemical evidence that cGMP interacts with the active site residues differently from cAMP.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/química , 3',5'-AMP Cíclico Fosfodiesterases/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Mutagênese Sítio-Dirigida , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , 3',5'-AMP Cíclico Fosfodiesterases/genética , Sítios de Ligação , Plaquetas/enzimologia , Domínio Catalítico , Cristalografia por Raios X , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Humanos , Immunoblotting , Cinética , Modelos Moleculares , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
Inflamm Res ; 50(3): 149-55, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11339503

RESUMO

OBJECTIVE AND DESIGN: We studied the ability of bradykinin (BK) receptor antagonists type 1 and 2 (B1-RA, B2-RA) to prevent acute inflammation. MATERIAL: A peptidoglycan-polysaccharide (PG-APS)-induced model of arthritis in the Lewis rat was analyzed. TREATMENT: Four groups of animals were studied for 5 days. Treatment was administered subcutaneously (s.c.) 1 mg/kg every 12 h. Group I received PG-APS and was treated with the B2-RA, CP-0597 (DArg-Arg-Pro-Hyp-Gly-Thi-Ser-DTic-NChg-Arg). Group II received PG-APS and was treated with a combined B1 and B2-RA, B9430 (DArg-Arg-Pro-Hyp-Gly-Igl-Ser-Dlgl-Oic-Arg). Group III received PG-APS and albumin control. Group IV received albumin control. METHODS: Joint diameter, liver weight, hematocrit, white blood count and plasma concentrations of prekallikrein, high molecular weight kininogen, HK and IL-beta were measured. Groups were compared by ANOVA. RESULTS: Acute arthritis and hepatomegaly were attenuated in the B2-RA-treated animals (p<0.05). Weight loss was more pronounced in the B1/B2-RA-treated animals. Anemia induced by PG-APS was prevented by B2-RA and B1/B2-RA treatment (p<0.001). A marked decrease in plasma HK to 64% of normal was found in the disease-untreated animals, which was completely normalized by B2-RA treatment and partially attenuated by the B1/B2-RA (78%). The decrease in plasma prekallikrein levels was prevented by combined B1/B2-RA treatment (p<0.05). Finally, elevated plasma IL-1beta levels were lowered by B1/B2-RA treatment and were below detection limits with the B2-RA treatment. CONCLUSIONS: These results indicate that the systemic inflammation is due in part to BK generation which can be blocked by B2-RA, while inhibiting the B1 receptor prevents an anti-inflammatory response.


Assuntos
Artrite/tratamento farmacológico , Antagonistas dos Receptores da Bradicinina , Inflamação/prevenção & controle , Oligopeptídeos/uso terapêutico , Peptidoglicano/toxicidade , Doença Aguda , Animais , Artrite/induzido quimicamente , Bradicinina/análogos & derivados , Bradicinina/fisiologia , Bradicinina/uso terapêutico , Feminino , Interleucina-1/sangue , Calicreínas/sangue , Neutrófilos/fisiologia , Ratos , Ratos Endogâmicos Lew , Receptor B2 da Bradicinina
17.
Biol Chem ; 382(1): 65-70, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11258675

RESUMO

Cleavage of high molecular weight kininogen (HK) by plasma kallikrein results in a light chain and a heavy chain (HK). The light chain has two domains: D6, which binds (pre)kallikrein, and D5, which binds to anionic surfaces, including heparin as well as zinc. Initially, HK was thought to be important for surface-activated coagulation. HKa or D5 binds to the urokinase receptor on endothelial cells, thereby enhancing the conversion of prourokinase to urokinase by kallikrein, and, thus, cell-associated fibrinolysis. HKa or D5 is antiadhesive by competing with vitronectin binding to the urokinase receptor and/or forming a complex with vitronectin. D5 inhibits endothelial cell migration, proliferation, tube formation and angiogenesis, thus modulating inflammation and neovascularization.


Assuntos
Adesão Celular/fisiologia , Cininogênios/fisiologia , Neovascularização Fisiológica/fisiologia , Sequência de Aminoácidos , Animais , Humanos , Cininogênios/química , Cininogênios/genética , Cininogênios/metabolismo , Dados de Sequência Molecular , Peso Molecular
18.
Hematol J ; 2(6): 371-7, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11920276

RESUMO

INTRODUCTION: Preliminary studies have shown that high molecular mass kininogen (HK) inhibits cathepsin G-induced platelet activation. However, the potential mechanism underlying this inhibitory effect remains to be elucidated. MATERIALS AND METHODS: Suspensions of washed and gel-filtered platelets were used in radioligand binding and aggregation studies. The amidolytic activity of cathepsin G was measured using specific chromogenic substrate. Western blot technique was utilised to explore the potential complex formation between cathepsin G and HK. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis was used to analyse the cleavage products of HK. RESULTS: At a concentration of 1 microM, HK completely blocked cathepsin G-induced platelet shape change and secretion of ATP. HK inhibited cathepsin G-induced platelet aggregation in a concentration-dependent manner with an IC(50) of 0.48 microM. Moreover, HK was found to inhibit binding of (125)I-cathepsin G to gel-filtered platelets. (125)I-cathepsin G forms a complex with HK. The complex formation did not affect the amidolytic activity of cathepsin G. HK was proteolysed upon interaction with cathepsin G. CONCLUSION: Our results show that high molecular mass kininogen down-regulates cathepsin G-induced platelet activation by forming a complex with cathepsin G and thus prevents binding of cathepsin G to platelets. These kininogen-cathepsin G interactions may be potential targets for pharmacological intervention.


Assuntos
Catepsinas/antagonistas & inibidores , Cininogênio de Alto Peso Molecular/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Plaquetas/metabolismo , Catepsina G , Catepsinas/metabolismo , Catepsinas/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Cinética , Cininogênio de Alto Peso Molecular/metabolismo , Fragmentos de Peptídeos/análise , Ligação Proteica , Serina Endopeptidases
19.
J Lab Clin Med ; 136(4): 292-302, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11039850

RESUMO

The thrombospondin-1 (TSP1) structural requirements within its heparin-binding domain (HBD)(30 kd) or within the other domains of the molecule (450 kd) that interact with neutrophils (PMNs) have not been delineated. Synthetic peptides based on the HBD, a TSP1 proteolytic fragment lacking the HBD, a large C-terminal domain of TSP1 (210 kd), a TSP1 recombinant fragment (rTSP1(784-932)), and a monoclonal antibody directed against the TSP1 type 3 repeats (mAb D4.6) were utilized to map such structural requirements on TSP1. Synthetic peptides containing a heparin-binding motif and encompassing residues F16-G33 or A74-S95 of TSP1 competed quantitatively with iodine 125-labeled TSP1 for binding to heparinagarose beads. However, only F16-G33 was a competitor of TSP1 binding to PMNs, suggesting that the sequence F16-G33 within the HBD plays a role in PMN binding. The interaction site within the 450-kd fragment was further narrowed. A TSP1 -derived proteolytic fragment (210 kd), a recombinant TSP1 fragment (rTSP1(784-932)), and a type 3 repeat anti-TSP1 monoclonal antibody (mAb D4.6) competed for the binding of 125I-labeled TSP1 to PMNs. The N-terminal of rTSP1(784-932) and C-terminal sequence analysis of TSP1-210 kd delineated the structural requirements for the second binding region for PMNs-namely, residues A784-N823.


Assuntos
Plaquetas/metabolismo , Neutrófilos/metabolismo , Trombospondina 1/metabolismo , Sequência de Aminoácidos , Anticorpos Monoclonais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/imunologia , Plaquetas/química , Cálcio/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Fibrinolíticos/metabolismo , Fibrinolíticos/farmacologia , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Heparina/metabolismo , Heparina/farmacologia , Humanos , Radioisótopos do Iodo , Dados de Sequência Molecular , Neutrófilos/citologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Mapeamento de Peptídeos , Estrutura Terciária de Proteína , Sensibilidade e Especificidade , Trombospondina 1/química , Trombospondina 1/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...