Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Thromb Res ; 167: 128-134, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29843086

RESUMO

INTRODUCTION: Rivaroxaban selectively inhibits factor Xa (FXa), which plays a central role in blood coagulation. In addition, FXa activates protease-activated receptor-2 (PAR-2). We have shown that PAR-2-/- mice exhibit less cardiac dysfunction after cardiac injury. MATERIAL AND METHODS: Wild-type (WT) and PAR-2-/- mice were subjected to left anterior descending artery (LAD) ligation to induce cardiac injury and heart failure. Mice received either placebo or rivaroxaban chow either starting at the time of surgery or 3 days after surgery and continued up to 28 days. Cardiac function was measured by echocardiography pre-surgery and 3, 7 and 28 days after LAD ligation. We also measured anticoagulation, intravascular thrombi, infarct size, cardiac hypertrophy and inflammation at various times. RESULTS: Rivaroxaban increased the prothrombin time and inhibited the formation of intravascular thrombi in mice subjected to LAD ligation. WT mice receiving rivaroxaban immediately after surgery had similar infarct sizes at day 1 as controls but exhibited significantly less impairment of cardiac function at day 3 and beyond compared to the placebo group. Rivaroxaban also inhibited the expansion of the infarct at day 28. Rivaroxaban did not significantly affect the expression of inflammatory mediators or a neutrophil marker at day 2 after LAD ligation. Delaying the start of rivaroxaban administration until 3 days after surgery failed to preserve cardiac function. In addition, rivaroxaban did not reduce cardiac dysfunction in PAR-2-/- mice. CONCLUSIONS: Early administration of rivaroxaban preserves cardiac function in mice after LAD ligation.


Assuntos
Inibidores do Fator Xa/uso terapêutico , Cardiopatias/tratamento farmacológico , Infarto do Miocárdio/prevenção & controle , Rivaroxabana/uso terapêutico , Animais , Modelos Animais de Doenças , Inibidores do Fator Xa/farmacologia , Humanos , Camundongos , Rivaroxabana/farmacologia
2.
Am J Pathol ; 180(1): 141-52, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22067907

RESUMO

Proteases and proteinase-activated receptor (PAR) activation are involved in several intestinal inflammatory conditions. We hypothesized that serine proteases and PAR activation could also modulate the intestinal injury induced by ischemia-reperfusion (I-R). C57Bl/6 mice were subjected to 90 minutes of intestinal ischemia followed or not by reperfusion. Sham-operated animals served as controls. After ischemia, plasma and tissue serine protease activity levels were increased compared to the activity measured in plasma and tissues from sham-operated mice. This increase was maintained or further enhanced after 2 and 5 hours of reperfusion, respectively. Trypsin (25 kDa) was detected in tissues both after ischemia and 2 hours of reperfusion. Treatment with FUT-175 (10 mg/kg), a potent serine protease inhibitor, increased survival after I-R, inhibited tissue protease activity, and significantly decreased intestinal myeloperoxidase (MPO) activity and chemokine and adhesion molecule expression. We investigated whether serine proteases modulate granulocyte recruitment by a PAR-dependent mechanism. MPO levels and adhesion molecule expression were significantly reduced in I-R groups pre-treated with the PAR(1) antagonist SCH-79797 (5 mg/kg) and in Par(2)(-/-)mice, compared, respectively, to vehicle-treated group and wild-type littermates. Thus, increased proteolytic activity and PAR activation play a pathogenic role in intestinal I-R injury. Inhibition of PAR-activating serine proteases could be beneficial to reduce post-ischemic intestinal inflammation.


Assuntos
Granulócitos/fisiologia , Intestino Delgado/irrigação sanguínea , Isquemia/enzimologia , Traumatismo por Reperfusão/prevenção & controle , Inibidores de Serina Proteinase/farmacologia , Animais , Benzamidinas , Quimiocinas/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Granulócitos/enzimologia , Guanidinas/farmacologia , Isquemia/patologia , Leucina/análogos & derivados , Leucina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peroxidase/metabolismo , Inibidores de Proteases/farmacologia , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Tripsina/metabolismo , alfa-Macroglobulinas/metabolismo
3.
Inflamm Res ; 59(9): 699-709, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20339899

RESUMO

OBJECTIVE: Our aim was to determine the contribution of proteinase-activated receptor-2 (PAR(2))-expressing bone marrow-derived cells on the development of colonic inflammation. MATERIALS: Chimeric mice were generated by injecting bone marrow cells from wildtype (PAR (2) (+/+) ) or PAR(2) knockout mice (PAR (2) (-/-) ) into irradiated PAR (2) (+/+) or PAR (2) (-/-) mice. TREATMENTS: Colitis was induced by giving 2.5% dextran sodium sulfate (DSS) solution for 7 days or by a single intracolonic administration of trinitrobenzene sulphonic acid (TNBS, 2 mg dissolved in 40% ethanol). METHODS: Seven days after the induction of colitis, bowel thickness, inflammatory parameters [myeloperoxidase (MPO) activity, macroscopic/microscopic damage scores], and leukocyte trafficking (visualized via intravital microscopy) were assessed. RESULTS: Total deficiency of PAR(2) resulted in a marked reduction in severity of both TNBS and DSS induced colitis as assessed by MPO activity, macroscopic damage, bowel thickness, and leukocyte adherence. Colitis was attenuated in all chimeric lines in which there was loss of PAR(2) in the host, non-bone marrow-derived tissue, independent of the status of PAR expression by bone marrow-derived cells. Interestingly, TNBS colitis was attenuated in PAR (2) (+/+) chimeric mice with PAR (2) (-/-) derived bone marrow but these animals were not protected from DSS colitis. CONCLUSIONS: Expression of PAR(2) by host-derived tissues plays a dominant role in regulating colonic inflammation. PAR(2) expression by bone marrow-derived cells appears to play a role in TNBS colitis but not in DSS induced injury.


Assuntos
Colite/metabolismo , Receptor PAR-2/metabolismo , Animais , Células da Medula Óssea/efeitos da radiação , Transplante de Medula Óssea , Quimera , Colite/induzido quimicamente , Colite/patologia , Colo/patologia , Colo/efeitos da radiação , Sulfato de Dextrana/toxicidade , Inflamação , Leucócitos/patologia , Leucócitos/efeitos da radiação , Camundongos , Camundongos Knockout , Peroxidase/análise , Receptor PAR-2/análise , Ácido Trinitrobenzenossulfônico/toxicidade
4.
Am J Respir Crit Care Med ; 181(3): 247-53, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19875688

RESUMO

RATIONALE: Mast cells and neutrophils are key contributors to the pathophysiological inflammatory processes that underpin asthma and chronic obstructive pulmonary disease, partly through the release of noxious serine proteases, including cathepsin G (Cat G) and chymase. From this standpoint, a dual inhibitor of neutrophil Cat G and mast cell chymase could protect against these disease-related inflammatory responses. OBJECTIVES: We examined the antiinflammatory pharmacology of RWJ-355871, a dual inhibitor of Cat G and chymase, in animal models of inflammation that evince pathophysiological pathways relevant to asthma and chronic obstructive pulmonary disease to determine the therapeutic potential of this compound. METHODS: In an ovalbumin (OVA)-sensitized rat model, RWJ-355871 was administered to block the mast-cell-mediated increase in paw volume caused by OVA injection. In a sheep asthma model, antigen-induced airway responses were assessed with and without aerosol treatment with RWJ-355871. In a murine tobacco-smoke model of airway inflammation, the effect of RWJ-355871 on smoke-induced neutrophilia was determined. MEASUREMENTS AND MAIN RESULTS: Intravenous treatment of OVA-sensitized rats with RWJ-355871 provided dose-dependent reduction in the increase in rat paw volume. In allergic sheep, aerosol pretreatment with RWJ-355871 showed dose-dependent inhibition of the antigen-induced early response, late response, and post-antigen-induced airway hyperreponsiveness. In tobacco-smoke-exposed mice, nebulized RWJ-355871 significantly reduced the smoke-induced neutrophilia from the levels observed in untreated mice. CONCLUSIONS: The preclinical antiinflammatory effects of RWJ-355871 in these animal models of inflammation indicate that this dual inhibitor may have therapeutic utility for treating airway inflammatory diseases involving mechanisms that depend on Cat G and/or chymase.


Assuntos
Catepsina G/antagonistas & inibidores , Quimases/antagonistas & inibidores , Pneumopatias/enzimologia , Organofosfonatos/uso terapêutico , Piperidinas/uso terapêutico , Doença Pulmonar Obstrutiva Crônica/enzimologia , Animais , Biomarcadores/metabolismo , Líquido da Lavagem Broncoalveolar/química , Catepsina G/metabolismo , Quimases/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Injeções Intravenosas , Pneumopatias/tratamento farmacológico , Camundongos , Organofosfonatos/administração & dosagem , Piperidinas/administração & dosagem , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Ratos , Ovinos , Resultado do Tratamento
5.
Cancer Res ; 68(17): 7219-27, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18757438

RESUMO

The G protein-coupled protease-activated receptors (PAR) are key signaling components for proteases in vascular biology and tumor progression. To address the contributions of PAR1 and PAR2 to breast cancer development, we established cohorts of mouse mammary tumor virus-polyoma middle T (PyMT) PAR1(-/-) and PAR2(-/-) mice, considering that the PyMT model recapitulates aspects of human disease. Appearance of palpable tumors, tumor expansion, and metastasis was indistinguishable between wild-type and PAR1(-/-) mice. PAR1(-/-) breast cancer cells were no longer responsive to thrombin in vitro, excluding compensatory up-regulation of alternative thrombin receptors and indicating that thrombin-PAR1 signaling is dispensable in breast tumor microenvironments. In contrast, palpable tumors and multifocal disease developed slower in PAR2(-/-) mice, and as a consequence of delayed tumor onset, metastasis was reduced. Analysis of early tumors showed persistence of adenomas with delayed appearance of vascularized adenocarcinomas in PAR2(-/-) mice. Furthermore, CXCL1 production by early PAR2(-/-) tumors was reduced. These results are consistent with previous xenograft data that implicated breast cancer PAR2 signaling in the induction of proangiogenic growth factors and chemokines. This study establishes that protease signaling contributes to mammary tumor development and that PAR2, rather than the thrombin receptor PAR1, plays a crucial role in the angiogenic switch.


Assuntos
Adenocarcinoma/fisiopatologia , Antígenos Transformantes de Poliomavirus/genética , Neoplasias Mamárias Experimentais/fisiopatologia , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Transdução de Sinais , Adenocarcinoma/irrigação sanguínea , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Neoplasias Mamárias Experimentais/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/genética , Reação em Cadeia da Polimerase
6.
J Immunol ; 179(8): 5493-503, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17911636

RESUMO

The proteinase-activated receptors (PARs) are a novel family of G protein-coupled receptors, and their effects in neurodegenerative diseases remain uncertain. Alzheimer's disease (AD) is a neurodegenerative disorder defined by misfolded protein accumulation with concurrent neuroinflammation and neuronal death. We report suppression of proteinase-activated receptor-2 (PAR2) expression in neurons of brains from AD patients, whereas PAR2 expression was increased in proximate glial cells, together with up-regulation of proinflammatory cytokines and chemokines and reduced IL-4 expression (p < 0.05). Glial PAR2 activation increased expression of formyl peptide receptor-2 (p < 0.01), a cognate receptor for a fibrillar 42-aa form of beta-amyloid (Abeta(1-42)), enhanced microglia-mediated proinflammatory responses, and suppressed astrocytic IL-4 expression, resulting in neuronal death (p < 0.05). Conversely, neuronal PAR2 activation protected human neurons against the toxic effects of Abeta(1-42) (p < 0.05), a key component of AD neuropathogenesis. Amyloid precursor protein-transgenic mice, displayed glial fibrillary acidic protein and IL-4 induction (p < 0.05) in the absence of proinflammatory gene up-regulation and neuronal injury, whereas PAR2 was up-regulated at this early stage of disease progression. PAR2-deficient mice, after hippocampal Abeta(1-42) implantation, exhibited enhanced IL-4 induction and less neuroinflammation (p < 0.05), together with improved neurobehavioral outcomes (p < 0.05). Thus, PAR2 exerted protective properties in neurons, but its activation in glia was pathogenic with secretion of neurotoxic factors and suppression of astrocytic anti-inflammatory mechanisms contributing to Abeta(1-42)-mediated neurodegeneration.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/prevenção & controle , Receptor PAR-2/fisiologia , Idoso , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Animais , Astrócitos/metabolismo , Linhagem Celular , Células Cultivadas , Feminino , Humanos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/toxicidade , Ratos , Receptor PAR-2/biossíntese , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Receptores de Formil Peptídeo/biossíntese , Receptores de Formil Peptídeo/genética , Receptores de Lipoxinas/biossíntese , Receptores de Lipoxinas/genética
7.
BMC Physiol ; 7: 3, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17397547

RESUMO

BACKGROUND: Protease-activated receptors (PAR) are present in the urinary bladder, and their expression is altered in response to inflammation. PARs are a unique class of G protein-coupled that carry their own ligands, which remain cryptic until unmasked by proteolytic cleavage. Although the canonical signal transduction pathway downstream of PAR activation and coupling with various G proteins is known and leads to the rapid transcription of genes involved in inflammation, the effect of PAR activation on the downstream transcriptome is unknown. We have shown that intravesical administration of PAR-activating peptides leads to an inflammatory reaction characterized by edema and granulocyte infiltration. Moreover, the inflammatory response to intravesical instillation of known pro-inflammatory stimuli such as E. coli lipopolysaccharide (LPS), substance P (SP), and antigen was strongly attenuated by PAR1- and to a lesser extent by PAR2-deficiency. RESULTS: Here, cDNA array experiments determined inflammatory genes whose expression is dependent on PAR1 activation. For this purpose, we compared the alteration in gene expression in wild type and PAR1-/- mice induced by classical pro-inflammatory stimuli (LPS, SP, and antigen). 75 transcripts were considered to be dependent on PAR-1 activation and further annotated in silico by Ingenuity Pathways Analysis (IPA) and gene ontology (GO). Selected transcripts were target validated by quantitative PCR (Q-PCR). Among PAR1-dependent transcripts, the following have been implicated in the inflammatory process: b2m, ccl7, cd200, cd63, cdbpd, cfl1, dusp1, fkbp1a, fth1, hspb1, marcksl1, mmp2, myo5a, nfkbia, pax1, plaur, ppia, ptpn1, ptprcap, s100a10, sim2, and tnfaip2. However, a balanced response to signals of injury requires a transient cellular activation of a panel of genes together with inhibitory systems that temper the overwhelming inflammation. In this context, the activation of genes such as dusp1 and nfkbia seems to counter-balance the inflammatory response to PAR activation by limiting prolonged activation of p38 MAPK and increased cytokine production. In contrast, transcripts such as arf6 and dcnt1 that are involved in the mechanism of PAR re-sensitization would tend to perpetuate the inflammatory reaction in response to common pro-inflammatory stimuli. CONCLUSION: The combination of cDNA array results and genomic networks reveals an overriding participation of PAR1 in bladder inflammation, provides a working model for the involvement of downstream signaling, and evokes testable hypotheses regarding the transcriptome downstream of PAR1 activation. It remains to be determined whether or not mechanisms targeting PAR1 gene silencing or PAR1 blockade will ameliorate the clinical manifestation of cystitis.


Assuntos
Cistite/genética , Cistite/metabolismo , Regulação da Expressão Gênica , Receptor PAR-1/metabolismo , Animais , Antígenos/imunologia , Cálcio/metabolismo , Cromatina/metabolismo , Cistite/induzido quimicamente , Cistite/imunologia , Feminino , Expressão Gênica , Genoma , Imunoprecipitação , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Fosfolipases A/metabolismo , Reação em Cadeia da Polimerase/métodos , Receptor PAR-1/deficiência , Frações Subcelulares/metabolismo , Substância P , Bexiga Urinária/metabolismo
8.
BMC Physiol ; 7: 4, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17397548

RESUMO

BACKGROUND: In general, inflammation plays a role in most bladder pathologies and represents a defense reaction to injury that often times is two edged. In particular, bladder neurogenic inflammation involves the participation of mast cells and sensory nerves. Increased mast cell numbers and tryptase release represent one of the prevalent etiologic theories for interstitial cystitis and other urinary bladder inflammatory conditions. The activity of mast cell-derived tryptase as well as thrombin is significantly increased during inflammation. Those enzymes activate specific G-protein coupled proteinase-activated receptors (PAR)s. Four PARs have been cloned so far, and not only are all four receptors highly expressed in different cell types of the mouse urinary bladder, but their expression is altered during experimental bladder inflammation. We hypothesize that PARs may link mast cell-derived proteases to bladder inflammation and, therefore, play a fundamental role in the pathogenesis of cystitis. RESULTS: Here, we demonstrate that in addition to the mouse urinary bladder, all four PA receptors are also expressed in the J82 human urothelial cell line. Intravesical administration of PAR-activating peptides in mice leads to an inflammatory reaction characterized by edema and granulocyte infiltration. Moreover, the inflammatory response to intravesical instillation of known pro-inflammatory stimuli such as E. coli lipopolysaccharide (LPS), substance P, and antigen was strongly attenuated by PAR1-, and to a lesser extent, by PAR2-deficiency. CONCLUSION: Our results reveal an overriding participation of PAR1 in bladder inflammation, provide a working model for the involvement of downstream signaling, and evoke testable hypotheses regarding the role of PARs in bladder inflammation. It remains to be determined whether or not mechanisms targeting PAR1 gene silencing or PAR1 blockade will ameliorate the clinical manifestations of cystitis.


Assuntos
Cistite/metabolismo , Receptor PAR-1/metabolismo , Animais , Antígenos/imunologia , Linhagem Celular , Cistite/induzido quimicamente , Cistite/imunologia , Cistite/patologia , Edema/induzido quimicamente , Granulócitos/patologia , Humanos , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor PAR-1/agonistas , Receptor PAR-1/deficiência , Receptor PAR-2/efeitos dos fármacos , Receptor PAR-2/metabolismo , Receptores Ativados por Proteinase/metabolismo , Substância P , Bexiga Urinária/metabolismo , Urotélio/citologia , Urotélio/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 27(6): 1456-62, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17363687

RESUMO

OBJECTIVE: Tissue factor (TF) initiates coagulation and indirectly triggers thrombin-dependent protease activated receptor (PAR) signaling. The TF-VIIa complex also directly cleaves PAR2 and promotes angiogenesis in vitro in TF cytoplasmic domain-deleted (TF(deltaCT)) mice. Here we address the effect of PAR1 and PAR2 deficiency on angiogenesis in vivo. METHODS AND RESULTS: In hypoxia-driven angiogenesis of oxygen induced retinopathy (OIR), wild-type, PAR1-/-, PAR2-/-, and TF(deltaCT) mice showed a comparable regression of the superficial vascular plexus during the initial exposure of mice to hyperoxia. However, TF(deltaCT) mice revascularized areas of central vaso-obliteration significantly faster than wild-type animals. Pharmacological inhibition of the TF-VIIa complex, but not of Xa, and blockade of tyrosine kinase receptor pathways with Gleevec reversed accelerated angiogenesis of TF(deltaCT) mice to revascularization rates observed in wild-type mice. Genetic deletion of PAR2, but not of PAR1, abolished enhanced revascularization of TF(deltaCT) mice. PAR1 knock-out animals were indistinguishable from wild-type mice in the model of retinal neoangiogenesis and angiogenesis-dependent subcutaneous tumor growth was unaltered in PAR1- and PAR2-deficient animals. CONCLUSION: Loss of the TF cytoplasmic domain results in accelerated hypoxia-induced angiogenesis mediated by TF-VIIa signaling. PAR2 signaling is sufficient for this proangiogenic effect without apparent contributions of mouse host cell PAR1.


Assuntos
Hiperóxia/metabolismo , Hipóxia/complicações , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Neovascularização Retiniana/etiologia , Vasos Retinianos/metabolismo , Transdução de Sinais , Tromboplastina/metabolismo , Animais , Benzamidas , Inibidores dos Fatores de Coagulação Sanguínea/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fator VIIa/metabolismo , Hiperóxia/induzido quimicamente , Hiperóxia/patologia , Hipóxia/metabolismo , Hipóxia/patologia , Mesilato de Imatinib , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Oxigênio , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Receptor PAR-1/deficiência , Receptor PAR-1/genética , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Transdução de Sinais/efeitos dos fármacos , Tromboplastina/genética , Fatores de Tempo
10.
J Clin Invest ; 117(3): 636-47, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17304351

RESUMO

Mediators involved in the generation of symptoms in patients with irritable bowel syndrome (IBS) are poorly understood. Here we show that colonic biopsy samples from IBS patients release increased levels of proteolytic activity (arginine cleavage) compared to asymptomatic controls. This was dependent on the activation of NF-kappaB. In addition, increased proteolytic activity was measured in vivo, in colonic washes from IBS compared with control patients. Trypsin and tryptase expression and release were increased in colonic biopsies from IBS patients compared with control subjects. Biopsies from IBS patients (but not controls) released mediators that sensitized murine sensory neurons in culture. Sensitization was prevented by a serine protease inhibitor and was absent in neurons lacking functional protease-activated receptor-2 (PAR2). Supernatants from colonic biopsies of IBS patients, but not controls, also caused somatic and visceral hyperalgesia and allodynia in mice, when administered into the colon. These pronociceptive effects were inhibited by serine protease inhibitors and a PAR2 antagonist and were absent in PAR2-deficient mice. Our study establishes that proteases are released in IBS and that they can directly stimulate sensory neurons and generate hypersensitivity symptoms through the activation of PAR2.


Assuntos
Dor Abdominal/enzimologia , Endopeptidases/fisiologia , Hiperalgesia/enzimologia , Síndrome do Intestino Irritável/complicações , Neurônios Aferentes/metabolismo , Dor Abdominal/etiologia , Adulto , Colo/enzimologia , Colo/inervação , Colo/patologia , Endopeptidases/análise , Endopeptidases/efeitos dos fármacos , Feminino , Humanos , Hiperalgesia/etiologia , Síndrome do Intestino Irritável/enzimologia , Síndrome do Intestino Irritável/patologia , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Receptor PAR-2/agonistas , Inibidores de Serina Proteinase/farmacologia , Tripsina/análise , Tripsina/efeitos dos fármacos , Tripsina/metabolismo , Triptases/análise , Triptases/antagonistas & inibidores , Triptases/metabolismo , alfa 1-Antitripsina/análise , alfa 1-Antitripsina/metabolismo
11.
J Biol Chem ; 281(43): 32095-112, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-16885167

RESUMO

Serine proteinases like thrombin can signal to cells by the cleavage/activation of proteinase-activated receptors (PARs). Although thrombin is a recognized physiological activator of PAR(1) and PAR(4), the endogenous enzymes responsible for activating PAR(2) in settings other than the gastrointestinal system, where trypsin can activate PAR(2), are unknown. We tested the hypothesis that the human tissue kallikrein (hK) family of proteinases regulates PAR signaling by using the following: 1) a high pressure liquid chromatography (HPLC)-mass spectral analysis of the cleavage products yielded upon incubation of hK5, -6, and -14 with synthetic PAR N-terminal peptide sequences representing the cleavage/activation motifs of PAR(1), PAR(2), and PAR(4); 2) PAR-dependent calcium signaling responses in cells expressing PAR(1), PAR(2), and PAR(4) and in human platelets; 3) a vascular ring vasorelaxation assay; and 4) a PAR(4)-dependent rat and human platelet aggregation assay. We found that hK5, -6, and -14 all yielded PAR peptide cleavage sequences consistent with either receptor activation or inactivation/disarming. Furthermore, hK14 was able to activate PAR(1), PAR(2), and PAR(4) and to disarm/inhibit PAR(1). Although hK5 and -6 were also able to activate PAR(2), they failed to cause PAR(4)-dependent aggregation of rat and human platelets, although hK14 did. Furthermore, the relative potencies and maximum effects of hK14 and -6 to activate PAR(2)-mediated calcium signaling differed. Our data indicate that in physiological settings, hKs may represent important endogenous regulators of the PARs and that different hKs can have differential actions on PAR(1), PAR(2), and PAR(4).


Assuntos
Calicreínas/farmacologia , Receptores Ativados por Proteinase/fisiologia , Transdução de Sinais/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Aorta Torácica/efeitos dos fármacos , Baculoviridae/genética , Plaquetas/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Humanos , Calicreínas/síntese química , Calicreínas/química , Calicreínas/classificação , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Ativados por Proteinase/química , Receptores Ativados por Proteinase/efeitos dos fármacos , Receptores Ativados por Proteinase/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Suínos , Trombina/farmacologia , Tripsina/farmacologia
12.
Am J Pathol ; 168(4): 1189-99, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16565494

RESUMO

We have investigated the specific contribution of protease-activated receptor-2 (PAR(2)) to host defense during Porphyromonas gingivalis infection. Culture supernatants from P. gingivalis strains 33277 and W50 provoked Ca(2+) mobilization in cells transfected with PAR(2) (PAR(2)-KNRK) and desensitized the subsequent responses to PAR(2)-selective agonist. In addition, culture supernatants of P. gingivalis E8 (RgpA/RgpB double knockout) did not cause calcium response in PAR(2)-KNRK cells, evidencing the involvement of the arginine-specific cysteine proteases RgpA and RgpB in PAR(2) activation by P. gingivalis. Injection of P. gingivalis into mouse subcutaneous chambers provoked an increased proteolytic activity, which was inhibited by serine protease inhibitors. Fluids collected from chambers of P. gingivalis-injected mice were able to activate PAR(2) and this activation was inhibited by serine protease inhibitors. P. gingivalis inoculation into subcutaneous chambers of wild-type mice induced an inflammatory response that was inhibited by a serine protease inhibitor and was significantly reduced in PAR(2)-deficient mice. Finally, mice orally challenged with P. gingivalis developed alveolar bone loss, which was significantly reduced in PAR(2)-deficient mice at 42 and 60 days after P. gingivalis infection. We conclude that PAR(2) is activated on P. gingivalis infection, in which it plays an important role in the host inflammatory response.


Assuntos
Infecções por Bacteroidaceae/metabolismo , Periodontite/metabolismo , Porphyromonas gingivalis/patogenicidade , Receptor PAR-2/fisiologia , Adesinas Bacterianas/metabolismo , Perda do Osso Alveolar/microbiologia , Animais , Infecções por Bacteroidaceae/microbiologia , Sinalização do Cálcio , Cisteína Endopeptidases/metabolismo , Citocinas/metabolismo , Dinoprostona/metabolismo , Cisteína Endopeptidases Gingipaínas , Camundongos , Camundongos Knockout , Peptídeo Hidrolases/metabolismo , Periodontite/microbiologia , Receptor PAR-2/genética , Receptor PAR-2/metabolismo
13.
J Exp Med ; 203(2): 425-35, 2006 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-16476770

RESUMO

The proteinase-activated receptors (PARs) are widely recognized for their modulatory properties of inflammation and neurodegeneration. We investigated the role of PAR2 in the pathogenesis of multiple sclerosis (MS) in humans and experimental autoimmune encephalomyelitis (EAE) in mice. PAR2 expression was increased on astrocytes and infiltrating macrophages in human MS and murine EAE central nervous system (CNS) white matter (P < 0.05). Macrophages and astrocytes from PAR2 wild-type (WT) and knockout (KO) mice exhibited differential immune gene expression with PAR2 KO macrophages showing significantly higher interleukin 10 production after lipopolysaccharide stimulation (P < 0.001). PAR2 activation in macrophages resulted in the release of soluble oligodendrocyte cytotoxins (P < 0.01). Myelin oligodendrocyte glycoprotein-induced EAE caused more severe inflammatory gene expression in the CNS of PAR2 WT animals (P < 0.05), together with enhanced T cell proliferation and interferon gamma production (P < 0.05), compared with KO littermates. Indeed, PAR2 WT animals showed markedly greater microglial activation and T lymphocyte infiltration accompanied by worsened demyelination and axonal injury in the CNS compared with their PAR2 KO littermates. Enhanced neuropathological changes were associated with a more severe progressive relapsing disease phenotype (P < 0.001) in WT animals. These findings reveal previously unreported pathogenic interactions between CNS PAR2 expression and neuroinflammation with ensuing demyelination and axonal injury.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Receptor PAR-2/fisiologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Proliferação de Células , Células Cultivadas , Encefalomielite Autoimune Experimental/imunologia , Feminino , Lobo Frontal/metabolismo , Lobo Frontal/patologia , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação/genética , Inflamação/metabolismo , Interferon gama/biossíntese , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Linfócitos T/metabolismo , Linfócitos T/patologia
14.
Inflamm Bowel Dis ; 11(9): 792-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16116312

RESUMO

BACKGROUND: Activation of colonic proteinase activated receptor-1 (PAR1) provokes colonic inflammation and increases mucosal permeability in mice. The mechanism of inflammation is not neurogenic like in the paw of rats but depends on PAR1-mediated activation monocytic cells. PAR1 activation in the colon increases the release of lymphocyte T helper-1 (TH1) cytokines. Moreover, PAR1 expression is increased in biopsies from patients with inflammatory bowel disease, and its activation during TH1-mediated colitis in mice increases all of the hallmarks of inflammation. METHODS: This study aimed to characterize the effects of PAR1 activation in oxazolone-mediated colitis, involving a TH2 cytokine profile. RESULTS: Intracolonic administration of oxazolone increased myeloperoxidase activity, damage score, and interleukin (IL)-4, IL-10, tumor necrosis factor alpha, and IL-1beta mRNA expression but lowered interferon-gamma mRNA expression, indicating colonic inflammation of a TH2 profile. The concurrent intracolonic administration of a PAR1 agonist in oxazolone-treated mice inhibited colitis, resulting in a reduction of myeloperoxidase activity, damage score, and inflammatory cytokine mRNA expression. Using PAR1-deficient mice, we confirmed that the anti-inflammatory effects of PAR1 agonists were mediated by PAR1. Moreover, in PAR1-deficient mice or in mice treated with a PAR1 antagonist, oxazolone-induced colitis was exacerbated, showing an endogenous modulatory role for PAR1 in this TH2 cytokine profile of colitis. CONCLUSIONS: Thus, as opposed to a previously shown proinflammatory role for PAR1 in a TH1 cytokine-mediated colitis, our new data show anti-inflammatory role for PAR1 activation in the setting of TH2 cytokine colitis model.


Assuntos
Colite/imunologia , Colite/fisiopatologia , Receptor PAR-1/fisiologia , Animais , Colite/veterinária , Citocinas/imunologia , Inflamação/fisiopatologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Oxazolona , RNA Mensageiro/biossíntese , Receptor PAR-1/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Th1/imunologia
15.
J Immunol ; 174(11): 7320-9, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15905579

RESUMO

Proteinase-activated receptors (PARs), a newly discovered subgroup of G-protein coupled receptors, are widely expressed by neural cells, but their roles in the nervous system remain uncertain. In this study, we report that PAR-2 was up-regulated on neurons in conjunction with neuroinflammation in brain tissue from patients with HIV-1-associated dementia. The inflammatory cytokines TNF-alpha and IL-1beta were also increased in HIV-1-associated dementia brains compared with patients without dementia (p < 0.05), but these same cytokines induced PAR-2 expression on neurons. Enhanced PAR-2 expression and subsequent activation prevented neuronal cell death and induction of the tumor suppressor, p53, caused by the HIV-encoded protein, Tat (p < 0.01). Intrastriatal implantation of a PAR-2 peptide agonist also inhibited Tat-induced neurotoxicity in a mouse model of HIV neuropathogenesis (p < 0.05). Moreover, PAR-2 null animals showed more severe neuroinflammation and neuronal loss caused by Tat neurotoxicity (p < 0.05). TNF-alpha protected wild-type neurons from Tat-related neurotoxicity, but in PAR-2-deficient neurons, the same concentrations of TNF-alpha were cytotoxic (p < 0.001). Thus, neuroinflammation can exert protective effects by which it induces PAR-2 expression with the ensuing abrogation of neuronal death.


Assuntos
Complexo AIDS Demência/metabolismo , Complexo AIDS Demência/patologia , HIV-1/imunologia , Neurônios/metabolismo , Neurônios/patologia , Receptor PAR-2/biossíntese , Complexo AIDS Demência/imunologia , Adulto , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/imunologia , Sobrevivência Celular/imunologia , Sistema Livre de Células/imunologia , Sistema Livre de Células/virologia , Células Cultivadas , Citocinas/fisiologia , Feminino , Produtos do Gene tat/antagonistas & inibidores , Produtos do Gene tat/toxicidade , Gliose/genética , Gliose/patologia , Gliose/fisiopatologia , Gliose/virologia , HIV-1/patogenicidade , Humanos , Mediadores da Inflamação/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/imunologia , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Receptor PAR-2/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/biossíntese , Células U937 , Produtos do Gene tat do Vírus da Imunodeficiência Humana
16.
J Med Chem ; 48(6): 1725-8, 2005 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-15771419

RESUMO

Novel indolylindazolylmaleimides were synthesized and examined for kinase inhibition. We identified low-nanomolar inhibitors of PKC-beta with good to excellent selectivity vs other PKC isozymes and GSK-3beta. In a cell-based functional assay, 8f and 8i effectively blocked IL-8 release induced by PKC-betaII (IC(50) = 20-25 nM). In cardiovascular safety assessment, representative lead compounds bound to the hERG channel with high affinity, potently inhibited ion current in a patch-clamp experiment, and caused a dose-dependent increase of QT(c) in guinea pigs.


Assuntos
Indazóis/síntese química , Indóis/síntese química , Maleimidas/síntese química , Proteína Quinase C/antagonistas & inibidores , Animais , Linhagem Celular , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/química , Glicogênio Sintase Quinase 3 beta , Cobaias , Humanos , Indazóis/farmacologia , Indazóis/toxicidade , Indóis/farmacologia , Indóis/toxicidade , Interleucina-8/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Síndrome do QT Longo/induzido quimicamente , Maleimidas/farmacologia , Maleimidas/toxicidade , Modelos Moleculares , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/síntese química , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Proteína Quinase C/química , Proteína Quinase C beta , Relação Estrutura-Atividade
17.
Am J Physiol Gastrointest Liver Physiol ; 288(2): G337-45, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15345469

RESUMO

The proteinase-activated thrombin receptor-1 (PAR-1) belongs to a unique family of G protein-coupled receptors activated by proteolytic cleavage. We studied the effect of PAR-1 activation in the regulation of ion transport in mouse colon in vitro. Expression of PAR-1 in mouse colon was assessed by RT-PCR and immunohistochemistry. To study the role of PAR-1 activation in chloride secretion, mouse colon was mounted in Ussing chambers. Changes in short-circuit current (Isc) were measured in tissues exposed to either thrombin, saline, the PAR-1-activating peptide TFLLR-NH2, or the inactive reverse peptide RLLFT-NH2, before electrical field stimulation (EFS). Experiments were repeated in the presence of either a PAR-1 antagonist or in PAR-1-deficient mice to assess receptor specificity. In addition, studies were conducted in the presence of chloride-free buffer or the muscarinic antagonist atropine to assess chloride dependency and the role of cholinergic neurons in the PAR-1-induced effect. PAR-1 mRNA was expressed in full-thickness specimens and mucosal scrapings of mouse colon. PAR-1 immunoreactivity was found on epithelial cells and on neurons in submucosal ganglia where it was colocalized with both VIP and neuropeptide Y. After PAR-1 activation by thrombin or TFLLR-NH2, secretory responses to EFS but not those to forskolin or carbachol were significantly reduced. The reduction in the response to EFS was not observed in the presence of the PAR-1 antagonist, in PAR-1-deficient mice, when chloride was excluded from the bathing medium, or when atropine was present. PAR-1 is expressed in submucosal ganglia in the mouse colon and its activation leads to a decrease in neurally evoked epithelial chloride secretion.


Assuntos
Cloretos/metabolismo , Colo/fisiologia , Mucosa Intestinal/metabolismo , Receptor PAR-1/fisiologia , Animais , Colo/química , Colo/inervação , Expressão Gênica , Técnicas In Vitro , Mucosa Intestinal/química , Transporte de Íons , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeo Y/análise , RNA Mensageiro/análise , Receptor PAR-1/análise , Transdução de Sinais , Peptídeo Intestinal Vasoativo/análise
18.
Am J Physiol Gastrointest Liver Physiol ; 288(2): G388-95, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15458925

RESUMO

Protease-activated receptor-2 (PAR-2) is a widely expressed tethered ligand receptor that can be activated by trypsin and other trypsin-like serine proteases. In the exocrine pancreas, PAR-2 activation modulates acinar cell secretion of digestive enzymes and duct cell ion channel function. During acute pancreatitis, digestive enzyme zymogens, including trypsinogen, are activated within the pancreas. We hypothesized that trypsin, acting via PAR-2, might regulate the severity of that disease, and to test this hypothesis, we examined the effect of either genetically deleting or pharmacologically activating PAR-2 on the severity of secretagogue-induced experimental pancreatitis. We found that experimental acute pancreatitis is more severe in PAR-2(-/-) than in wild-type mice and that in vivo activation of PAR-2, achieved by parenteral administration of the PAR-2-activating peptide SLIGRL-NH2, reduces the severity of pancreatitis. In the pancreas during the early stages of pancreatitis, the MAPK ERK1/2 is activated and translocated to the nucleus, but nuclear translocation is reduced by activation of PAR-2. Our findings indicate that PAR-2 exerts a protective effect on pancreatitis and that activation of PAR-2 ameliorates pancreatitis, possibly by inhibiting ERK1/2 translocation to the nucleus. Our observations suggest that PAR-2 activation may be of therapeutic value in the treatment and/or prevention of severe clinical pancreatitis, and they lead us to speculate that, from a teleological standpoint, PAR-2 may have evolved in the pancreas as a protective mechanism designed to dampen the injurious effects of intrapancreatic trypsinogen activation.


Assuntos
Pancreatite/fisiopatologia , Receptor PAR-2/fisiologia , Animais , Ceruletídeo , Ativação Enzimática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oligopeptídeos/farmacologia , Pancreatite/induzido quimicamente , Pancreatite/metabolismo , Peptídeos/farmacologia , Receptor PAR-2/genética , Receptor PAR-2/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 24(6): 1118-23, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15130911

RESUMO

OBJECTIVE: Recently, mice made deficient in growth arrest-specific gene 6 product (Gas6) or in which Gas6 gene expression was inhibited were shown to have platelet dysfunction and to be less susceptible to thrombosis. The aim of this study was to define and characterize the relevant Gas6 receptor or receptors involved in platelet function. METHODS AND RESULTS: Using RT-PCR and Western blot analysis we found that mer was the predominantly expressed subtype in mouse and human platelets, whereas axl and rse were not detected. We generated mer-deficient mice by targeted disruption of the mer receptor gene. Platelets derived from mer-deficient mice had decreased platelet aggregation in responses to low concentrations of collagen, U46619, and PAR4 thrombin receptor agonist peptide in vitro. However, the response to ADP was not different from wild-type platelets. Knockout of the mer gene protected mice from collagen/epinephrine-induced pulmonary thromoembolism and inhibited ferric chloride-induced thrombosis in vivo. Tail bleeding times, coagulation parameters, and peripheral blood cell counts in mer-deficient mice were similar to wild-type mice. CONCLUSIONS: Our data provide the first evidence that mer, presumably through activation by its ligand Gas6, participates in regulation of platelet function in vitro and platelet-dependent thrombosis in vivo.


Assuntos
Plaquetas/enzimologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Testes de Coagulação Sanguínea , Plaquetas/fisiologia , Western Blotting , Cloretos , Colágeno/farmacologia , Colágeno/toxicidade , Epinefrina/toxicidade , Feminino , Compostos Férricos/toxicidade , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligopeptídeos/farmacologia , Proteínas Oncogênicas/análise , Agregação Plaquetária/efeitos dos fármacos , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/análise , Receptores Proteína Tirosina Quinases/deficiência , Receptores Proteína Tirosina Quinases/genética , Receptores de Trombina/agonistas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tromboembolia/induzido quimicamente , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
20.
Blood ; 103(4): 1342-7, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14576054

RESUMO

Sepsis is associated with a systemic activation of coagulation and an excessive inflammatory response. Anticoagulants have been shown to inhibit both coagulation and inflammation in sepsis. In this study, we used both genetic and pharmacologic approaches to analyze the role of tissue factor and protease-activated receptors in coagulation and inflammation in a mouse endotoxemia model. We used mice expressing low levels of the procoagulant molecule, tissue factor (TF), to analyze the effects of TF deficiency either in all tissues or selectively in hematopoietic cells. Low TF mice had reduced coagulation, inflammation, and mortality compared with control mice. Similarly, a deficiency of TF expression by hematopoietic cells reduced lipopolysaccharide (LPS)-induced coagulation, inflammation, and mortality. Inhibition of the down-stream coagulation protease, thrombin, reduced fibrin deposition and prolonged survival without affecting inflammation. Deficiency of either protease activated receptor-1 (PAR-1) or protease activated receptor-2 (PAR-2) alone did not affect inflammation or survival. However, a combination of thrombin inhibition and PAR-2 deficiency reduced inflammation and mortality. These data demonstrate that hematopoietic cells are the major pathologic site of TF expression during endotoxemia and suggest that multiple protease-activated receptors mediate crosstalk between coagulation and inflammation.


Assuntos
Moléculas de Adesão Celular , Endotoxemia/imunologia , Endotoxemia/metabolismo , Receptor PAR-1/metabolismo , Tromboplastina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/imunologia , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Modelos Animais de Doenças , Endotoxemia/mortalidade , Fibrina/metabolismo , Hematopoese/fisiologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Receptor Cross-Talk/fisiologia , Receptor PAR-2/metabolismo , Receptores Ativados por Proteinase/metabolismo , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA