Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Am J Respir Crit Care Med ; 205(7): 769-782, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35073247

RESUMO

Rationale: Although the cysteine protease cathepsin S has been implicated in the pathogenesis of several inflammatory lung diseases, its role has not been examined in the context of acute respiratory distress syndrome, a condition that still lacks specific and effective pharmacological treatments. Objectives: To characterize the status of cathepsin S in acute lung inflammation and examine the role of cathepsin S in disease pathogenesis. Methods: Human and mouse model BAL fluid samples were analyzed for the presence and activity of cathepsin S and its endogenous inhibitors. Recombinant cathepsin S was instilled directly into the lungs of mice. The effects of cathepsin S knockout and pharmacological inhibition were examined in two models of acute lung injury. Protease-activated receptor-1 antagonism was used to test a possible mechanism for cathepsin S-mediated inflammation. Measurements and Main Results: Pulmonary cathepsin S concentrations and activity were elevated in acute respiratory distress syndrome, a phenotype possibly exacerbated by the loss of the endogenous antiprotease cystatin SN. Direct cathepsin S instillation into the lungs induced key pathologies of acute respiratory distress syndrome, including neutrophilia and alveolar leakage. Conversely, in murine models of acute lung injury, genetic knockdown and prophylactic or therapeutic inhibition of cathepsin S reduced neutrophil recruitment and protein leakage. Cathepsin S may partly mediate its pathogenic effects via protease-activated receptor-1, because antagonism of this receptor abrogated cathepsin S-induced airway inflammation. Conclusions: Cathepsin S contributes to acute lung injury and may represent a novel therapeutic target for acute respiratory distress syndrome.


Assuntos
Pneumonia , Síndrome do Desconforto Respiratório , Animais , Líquido da Lavagem Broncoalveolar , Catepsinas , Modelos Animais de Doenças , Humanos , Pulmão/patologia , Camundongos
2.
Eur Respir J ; 53(3)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30655278

RESUMO

Cathepsin S (CatS) is upregulated in the lungs of patients with cystic fibrosis (CF). However, its role in CF lung disease pathogenesis remains unclear.In this study, ß-epithelial Na+ channel-overexpressing transgenic (ßENaC-Tg) mice, a model of CF-like lung disease, were crossed with CatS null (CatS-/-) mice or treated with the CatS inhibitor VBY-999.Levels of active CatS were elevated in the lungs of ßENaC-Tg mice compared with wild-type (WT) littermates. CatS-/-ßENaC-Tg mice exhibited decreased pulmonary inflammation, mucus obstruction and structural lung damage compared with ßENaC-Tg mice. Pharmacological inhibition of CatS resulted in a significant decrease in pulmonary inflammation, lung damage and mucus plugging in the lungs of ßENaC-Tg mice. In addition, instillation of CatS into the lungs of WT mice resulted in inflammation, lung remodelling and upregulation of mucin expression. Inhibition of the CatS target, protease-activated receptor 2 (PAR2), in ßENaC-Tg mice resulted in a reduction in airway inflammation and mucin expression, indicating a role for this receptor in CatS-induced lung pathology.Our data indicate an important role for CatS in the pathogenesis of CF-like lung disease mediated in part by PAR2 and highlight CatS as a therapeutic target.


Assuntos
Catepsinas/metabolismo , Fibrose Cística/metabolismo , Muco/metabolismo , Pneumonia/metabolismo , Receptor PAR-2/metabolismo , Obstrução das Vias Respiratórias/metabolismo , Animais , Catepsinas/genética , Modelos Animais de Doenças , Canais Epiteliais de Sódio/genética , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia/etiologia
3.
J Infect Dis ; 217(9): 1462-1471, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29415278

RESUMO

Streptococcus pneumoniae is the most common causative pathogen in community-acquired pneumonia. Protease-activated receptor 2 (PAR2) is expressed by different cell types in the lungs and can mediate inflammatory responses. We sought to determine the role of PAR2 during pneumococcal pneumonia. Pneumococcal pneumonia or sepsis was induced in wild-type and PAR2 knock-out (Par2-/-) mice by infection with viable S. pneumoniae. Par2-/- mice demonstrated improved host defense, a largely preserved lung barrier integrity, and reduced mortality during pneumococcal pneumonia. PAR2 deficiency did not influence bacterial growth after intravenous infection. Inhibition of the endogenous PAR2 activating proteases tissue factor/factor VIIa or tryptase did not impact on bacterial burdens during pneumonia. In a PAR2 reporter cell line it was demonstrated that S. pneumoniae-derived proteases are able to cleave PAR2. These results show that S. pneumoniae is able to cleave and exploit PAR2 to disseminate systemically from the airways.


Assuntos
Pneumonia Pneumocócica/microbiologia , Receptor PAR-2 , Streptococcus pneumoniae/fisiologia , Animais , Carga Bacteriana , Coagulação Sanguínea , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Proteínas de Helminto/farmacologia , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Pneumocócica/patologia , Organismos Livres de Patógenos Específicos
4.
Eur Respir J ; 49(2)2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28179435

RESUMO

Proteases were traditionally viewed as mere protein-degrading enzymes with a very restricted spectrum of substrates. A major expansion in protease research has uncovered a variety of novel substrates, and it is now evident that proteases are critical pleiotropic actors orchestrating pathophysiological processes. Recent findings evidenced that the net proteolytic activity also relies upon interconnections between different protease and protease inhibitor families in the protease web.In this review, we provide an overview of these novel concepts with a particular focus on pulmonary pathophysiology. We describe the emerging roles of several protease families including cysteine and serine proteases.The complexity of the protease web is exemplified in the light of multidimensional regulation of serine protease activity by matrix metalloproteases through cognate serine protease inhibitor processing. Finally, we will highlight how deregulated protease activity during pulmonary pathogenesis may be exploited for diagnosis/prognosis purposes, and utilised as a therapeutic tool using nanotechnologies.Considering proteases as part of an integrative biology perspective may pave the way for the development of new therapeutic targets to treat pulmonary diseases related to intrinsic protease deregulation.


Assuntos
Pneumopatias/enzimologia , Pulmão/enzimologia , Metaloproteinases da Matriz/metabolismo , Animais , Humanos , Pulmão/imunologia , Pneumopatias/tratamento farmacológico , Pneumopatias/imunologia , Camundongos , Inibidores de Proteases/uso terapêutico , Proteólise/efeitos dos fármacos
5.
Am J Respir Crit Care Med ; 193(8): 847-60, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26599507

RESUMO

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is a devastating disease that remains refractory to current therapies. OBJECTIVES: To characterize the expression and activity of the membrane-anchored serine protease matriptase in IPF in humans and unravel its potential role in human and experimental pulmonary fibrogenesis. METHODS: Matriptase expression was assessed in tissue specimens from patients with IPF versus control subjects using quantitative reverse transcriptase-polymerase chain reaction, immunohistochemistry, and Western blotting, while matriptase activity was monitored by fluorogenic substrate cleavage. Matriptase-induced fibroproliferative responses and the receptor involved were characterized in human primary pulmonary fibroblasts by Western blot, viability, and migration assays. In the murine model of bleomycin-induced pulmonary fibrosis, the consequences of matriptase depletion, either by using the pharmacological inhibitor camostat mesilate (CM), or by genetic down-regulation using matriptase hypomorphic mice, were characterized by quantification of secreted collagen and immunostainings. MEASUREMENTS AND MAIN RESULTS: Matriptase expression and activity were up-regulated in IPF and bleomycin-induced pulmonary fibrosis. In cultured human pulmonary fibroblasts, matriptase expression was significantly induced by transforming growth factor-ß. Furthermore, matriptase elicited signaling via protease-activated receptor-2 (PAR-2), and promoted fibroblast activation, proliferation, and migration. In the experimental bleomycin model, matriptase depletion, by the pharmacological inhibitor CM or by genetic down-regulation, diminished lung injury, collagen production, and transforming growth factor-ß expression and signaling. CONCLUSIONS: These results implicate increased matriptase expression and activity in the pathogenesis of pulmonary fibrosis in human IPF and in an experimental mouse model. Overall, targeting matriptase, or treatment by CM, which is already in clinical use for other diseases, may represent potential therapies for IPF.


Assuntos
Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/fisiopatologia , Pulmão/metabolismo , Pulmão/fisiopatologia , Serina Endopeptidases/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Serina Proteases/metabolismo
6.
Proc Natl Acad Sci U S A ; 109(23): 9113-8, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22615380

RESUMO

CCAAT/enhancer-binding protein δ (C/EBPδ) recently emerged as an essential player in the inflammatory response to bacterial infections. C/EBPδ levels increase rapidly after a proinflammatory stimulus, and increasing C/EBPδ levels seem to be indispensable for amplification of the inflammatory response. Here we aimed to elucidate the role of C/EBPδ in host defense in community-acquired pneumococcal pneumonia. We show that C/EBPδ(-/-) mice are relatively resistant to pneumococcal pneumonia, as indicated by delayed and reduced mortality, diminished outgrowth of pneumococci in lungs, and reduced dissemination of the infection. Moreover, expression of platelet-activating factor receptor (PAFR), which is known to potentiate bacterial translocation of gram-positive bacteria, was significantly reduced during infection in C/EBPδ(-/-) mice compared with WT controls. Importantly, cell stimulation experiments revealed that C/EBPδ potentiates PAFR expression induced by lipoteichoic acid and pneumococci. Thus, C/EBPδ exaggerates bacterial dissemination during Streptococcus pneumoniae-induced pulmonary infection, suggesting an important role for PAFR-dependent bacterial translocation.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/imunologia , Regulação da Expressão Gênica/fisiologia , Glicoproteínas da Membrana de Plaquetas/metabolismo , Pneumonia Pneumocócica/imunologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Western Blotting , Proteína delta de Ligação ao Facilitador CCAAT/genética , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Técnicas Histológicas , Humanos , Luciferases , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Knockout , Permeabilidade , Pneumonia Pneumocócica/microbiologia , Reação em Cadeia da Polimerase em Tempo Real , Estatísticas não Paramétricas
7.
Lab Invest ; 94(1): 89-97, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24247561

RESUMO

CCAAT-enhancer-binding protein delta (C/EBPδ) is a transcription factor mainly known for its role in inflammation and apoptosis/proliferation. Considering that these are key processes in renal fibrosis, we hypothesized that C/EBPδ would potentiate renal fibrosis. In line with this hypothesis, C/EBPδ has recently been suggested to regulate the fibrotic response during glomerulonephritis. Here we determined the importance of C/EBPδ in the development of renal tubulointerstitial fibrosis by subjecting 8- to 12-week-old C/EBPδ-deficient mice and age- and sex-matched wild-type controls to the unilateral ureteral obstruction model. Mice were killed at 1, 3, or 7 days post surgery, and renal tissues were obtained for RNA, protein, and immunohistochemical analysis. We show that C/EBPδ deficiency resulted in a more profound fibrotic response as evident from enhanced tubular injury, collagen deposition in the interstitial area, and higher expression of transforming growth factor-ß. Moreover, we show that the increase in renal fibrosis in C/EBPδ-deficient mice does not depend on an altered proliferation/apoptosis balance or on a differential inflammatory response in the obstructed kidney. In conclusion, our study provides direct evidence that C/EBPδ is a novel mediator of renal fibrosis. Modulating C/EBPδ expression could consequently be a potential antifibrotic strategy in patients with chronic kidney disease.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Animais , Apoptose , Proteína delta de Ligação ao Facilitador CCAAT/genética , Processos de Crescimento Celular/fisiologia , Feminino , Fibrose/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estatísticas não Paramétricas , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
8.
Mol Med ; 20: 410-6, 2014 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-24842054

RESUMO

Accumulating evidence shows that protease-activated receptor-1 (PAR-1) plays an important role in the development of fibrosis, including lung fibrosis. However, whether PAR-1 also plays a role in the development of skin fibrosis remains elusive. The aim of this study was to determine the role of PAR-1 in the development of skin fibrosis. To explore possible mechanisms by which PAR-1 could play a role, human dermal fibroblasts and keratinocytes were stimulated with specific PAR-1 agonists or antagonists. To investigate the role of PAR-1 in skin fibrosis, we subjected wild-type and PAR-1-deficient mice to a model of bleomycin-induced skin fibrosis. PAR-1 activation leads to increased proliferation and extra cellular matrix (ECM) production, but not migration of human dermal fibroblasts (HDF) in vitro. Moreover, transforming growth factor (TGF)-ß production was increased in keratinocytes upon PAR-1 activation, but not in HDF. The loss of PAR-1 in vivo significantly attenuated bleomycin-induced skin fibrosis. The bleomycin-induced increase in dermal thickness and ECM production was reduced significantly in PAR-1-deficient mice compared with wild-type mice. Moreover, TGF-ß expression and the number of proliferating fibroblasts were reduced in PAR-1-deficient mice although the difference did not reach statistical significance. This study demonstrates that PAR-1 contributes to the development of skin fibrosis and we suggest that PAR-1 potentiates the fibrotic response mainly by inducing fibroblast proliferation and ECM production.


Assuntos
Fibroblastos/patologia , Queratinócitos/patologia , Receptor PAR-1/metabolismo , Dermatopatias/metabolismo , Pele/patologia , Animais , Bleomicina , Linhagem Celular , Proliferação de Células , Células Cultivadas , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrose , Humanos , Queratinócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor PAR-1/agonistas , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-1/genética , Pele/efeitos dos fármacos , Dermatopatias/induzido quimicamente , Dermatopatias/patologia
9.
J Cell Mol Med ; 16(9): 2035-48, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22128761

RESUMO

Prolactin is best known as the polypeptide anterior pituitary hormone, which regulates the development of the mammary gland. However, it became clear over the last decade that prolactin contributes to a broad range of pathologies, including breast cancer. Prolactin is also involved in angiogenesis via the release of pro-angiogenic factors by leukocytes and epithelial cells. However, whether prolactin also influences endothelial cells, and whether there are functional consequences of prolactin-induced signalling in the perspective of angiogenesis, remains so far elusive. In the present study, we show that prolactin induces phosphorylation of ERK1/2 and STAT5 and induces tube formation of endothelial cells on Matrigel. These effects are blocked by a specific prolactin receptor antagonist, del1-9-G129R-hPRL. Moreover, in an in vivo model of the chorioallantoic membrane of the chicken embryo, prolactin enhances vessel density and the tortuosity of the vasculature and pillar formation, which are hallmarks of intussusceptive angiogenesis. Interestingly, while prolactin has only little effect on endothelial cell proliferation, it markedly stimulates endothelial cell migration. Again, migration was reverted by del1-9-G129R-hPRL, indicating a direct effect of prolactin on its receptor. Immunohistochemistry and spectral imaging revealed that the prolactin receptor is present in the microvasculature of human breast carcinoma tissue. Altogether, these results suggest that prolactin may directly stimulate angiogenesis, which could be one of the mechanisms by which prolactin contributes to breast cancer progression, thereby providing a potential tool for intervention.


Assuntos
Células Endoteliais/patologia , Neovascularização Patológica/patologia , Prolactina/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Indutores da Angiogênese/efeitos adversos , Animais , Neoplasias da Mama/patologia , Linhagem Celular , Embrião de Galinha , Colágeno/metabolismo , Combinação de Medicamentos , Células Endoteliais/metabolismo , Feminino , Imuno-Histoquímica , Laminina/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Fosforilação , Proteoglicanas/metabolismo , Receptores da Prolactina/antagonistas & inibidores , Receptores da Prolactina/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo
10.
Am J Pathol ; 178(2): 924-34, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21281823

RESUMO

Insulin-like growth factor-1 (IGF-1) signaling is important for the maintenance of plaque stability in atherosclerosis due to its effects on vascular smooth muscle cell (vSMC) phenotype. To investigate this hypothesis, we studied the effects of the highly inflammatory milieu of the atherosclerotic plaque on IGF-1 signaling and stability-related phenotypic parameters of murine vSMCs in vitro, and the effects of IGF-1 supplementation on plaque phenotype in an atherosclerotic mouse model. M1-polarized, macrophage-conditioned medium inhibited IGF-1 signaling by ablating IGF-1 and increasing IGF-binding protein 3, increased vSMC apoptosis, and decreased proliferation. Expression of α-actin and col3a1 genes was strongly attenuated by macrophage-conditioned medium, whereas expression of matrix-degrading enzymes was increased. Importantly, all of these effects could be corrected by supplementation with IGF-1. In vivo, treatment with the stable IGF-1 analog Long R3 IGF-1 in apolipoprotein E knockout mice reduced stenosis and core size, and doubled cap/core ratio in early atherosclerosis. In advanced plaques, Long R3 IGF-1 increased the vSMC content of the plaque by more than twofold and significantly reduced the rate of intraplaque hemorrhage. We believe that IGF-1 in atherosclerotic plaques may have a role in preventing plaque instability, not only by modulating smooth muscle cell turnover, but also by altering smooth muscle cell phenotype.


Assuntos
Aterosclerose/patologia , Fator de Crescimento Insulin-Like I/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Placa Aterosclerótica/patologia , Animais , Aterosclerose/metabolismo , Western Blotting , Movimento Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fator de Crescimento Insulin-Like I/genética , Camundongos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fenótipo , Placa Aterosclerótica/metabolismo , Isoformas de Proteínas/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
Thromb Res ; 123(6): 914-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19108874

RESUMO

INTRODUCTION: Factor X (FX) is a serine-protease playing a crucial role in the blood coagulation pathway and triggering intracellular signalling in a variety of cells via protease-activated receptors (PARs). By exploiting naturally occurring variants (V342A and G381D, catalytic domain; E19A, gamma-carboxyglutamic acid (GLA)-rich domain), we investigated the relationship between the pro-coagulant activity and the signal transduction capacity of FX. MATERIALS AND METHODS: Recombinant FX (rFX) variants were expressed in Human Embryonic Kidney cells and purified by immunoaffinity chromatography. Activated rFX (rFXa) variants were characterized for pro-coagulant, amidolytic and thrombin generation activity. rFXa signalling was assessed through evaluation of extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation in C2C12 myoblasts. RESULTS AND CONCLUSIONS: rFX variants showed reduced (rFX-342A, 29%; rFX-19A, 12%) or not detectable (rFX-381D) amidolytic activity. Thrombin generation activity in a plasma system was also decreased either upon activation by Russell's viper venom (rFX-342A, 38%; rFX-19A, 7%; rFX-381D, not detectable) or by the extrinsic pathway (rFX-342A, 36%; rFX-19A, rFX-381D, not detectable). The rFXa-381D mutant displayed little or no enzymatic activity, and did not induce any appreciable signal transduction capacity. The rFXa-342A mutant induced a dose-dependent signalling with a 50% reduced signalling capacity. At the highest concentration (174 nM), signalling progressed with a time course similar to that of rFXa-wt. Zymogen rFX-19A showed defective and incomplete activation resulting in strongly reduced enzymatic activity and signalling. Taken together our data are consistent with a close correlation between pro-coagulant activity and intracellular signalling capacity.


Assuntos
Fator Xa/genética , Fator Xa/metabolismo , Mutação , Animais , Linhagem Celular , Coagulantes/metabolismo , Variação Genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Protrombina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Especificidade por Substrato
12.
Pathophysiol Haemost Thromb ; 36(3-4): 137-47, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19176987

RESUMO

Protease-activated receptors (PARs) are G-protein-coupled receptors (GPCRs) that are activated by a unique proteolytic mechanism. Besides the important role of blood coagulation factors in preventing bleeding after vascular injury, these serine proteinases actively engage target cells thereby fulfilling critical functions in cell biology. Cellular responses triggered by coagulation factor-induced PAR activation suggest that PARs play an important role in proliferation, survival and/or malignant transformation of tumor cells. Indeed, PAR expression correlates with cancer malignancy and clinical studies show that anticoagulant treatment is beneficial in cancer patients. In this review, we provide an overview on the PAR family, their mode of activation and mechanisms by which PAR signaling is terminated. In addition, we discuss the relationship between blood coagulation and cancer biology focusing on the potential role of PAR-induced modulation of cell survival, apoptosis and tumor growth.


Assuntos
Apoptose/fisiologia , Fatores de Coagulação Sanguínea/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/patologia , Receptores Ativados por Proteinase/fisiologia , Sequência de Aminoácidos , Animais , Caspases/fisiologia , Divisão Celular/fisiologia , Transformação Celular Neoplásica , Sequência Conservada , Ativação Enzimática , Proteínas Heterotriméricas de Ligação ao GTP/fisiologia , Humanos , Camundongos , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Neoplasias/sangue , Conformação Proteica , Estrutura Terciária de Proteína , Receptores Ativados por Proteinase/agonistas , Receptores Ativados por Proteinase/química , Receptores Ativados por Proteinase/genética , Rodopsina/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Trombofilia/etiologia , Trombofilia/fisiopatologia , Tromboplastina/fisiologia
13.
Cell Signal ; 19(12): 2596-604, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17884337

RESUMO

Sonic hedgehog (Shh) is a morphogen pivotal for development and tissue maintenance. Biological effects of Shh are mediated through a pathway that involves binding to patched1 (Ptch1), thereby releasing Smoothened (Smo) from inhibition resulting in the activation of Gli transcription factors, which mediate the induction of Shh target genes. Here, we describe a novel signal transduction pathway for Shh, which is transcription/translation-independent, SuFu insensitive, and consequently independent of Gli-mediated induction of transcription. Through this alternative pathway Shh, transduced via Smo, induced altered cell morphology together with lamellipodia formation. Migration assays demonstrate that this cytoskeletal rearrangement mediates the migratory response to Shh. This Shh-induced, Smo mediated migration utilizes and requires the metabolism of arachidonic acid through the 5-lipoxygenase pathway. These data provide a link between a seemingly novel Gli-independent Hh signaling pathway and the leukotriene metabolism, and might explain the developmental abnormalities observed in both patients with defective leukotriene metabolism as well as in rodent models of defective Rho family GTPase signaling.


Assuntos
Ácido Araquidônico/metabolismo , Movimento Celular , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Actinas/metabolismo , Animais , Araquidonato 5-Lipoxigenase/metabolismo , Linhagem Celular , Forma Celular , Quimiotaxia , Fibroblastos/enzimologia , Cinética , Leucotrienos/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Pseudópodes/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Receptor Smoothened , Transativadores/genética , Transativadores/metabolismo , Transcrição Gênica , Transfecção , Proteína GLI1 em Dedos de Zinco
14.
J Clin Transl Res ; 3(Suppl 2): 358-365, 2018 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-30873483

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis is a devastating fibrotic diffuse parenchymal lung disorder that remains refractory to pharmacological therapies. Therefore, novel treatments are urgently required. CCAAT/enhancer binding protein delta (C/EBPδ) is a transcription factor that mediates critical cellular functions in pathophysiology and which was recently suggested to be a key regulatory component in IPF. The purpose of this study was to prove or refute the importance of C/EBPδ in pulmonary fibrosis. METHODS: Pulmonary fibrosis was induced by intranasal instillation of bleomycin into wild-type and C/EBPδ deficient mice. At different time intervals after bleomycin instillation, fibrosis was assessed by hydroxyproline analysis, histochemistry and q-PCR for fibrotic marker expression. RESULTS: C/EBPδ deficient mice developed pulmonary fibrosis to a similar degree as wildtype mice as evident from similar Ashcroft scores, hydroxyproline levels and expression levels of collagen, fibronectin and α-smooth muscle actin at both 14 and 21 days after bleomycin instillation. The resolution of fibrosis, assessed at 48 days after bleomycin instillation, was also similar in wildtype and C/EBPδ deficient mice. In line with the lack of effect of C/EBPδ on fibrosis progression/resolution, macrophage recruitment and/or differentiation were also not different in wildtype or C/EBPδ deficient mice. CONCLUSIONS: Overall, C/EBPδ does not seem to affect bleomycin-induced experimental pulmonary fibrosis and we challenge the importance of C/EBPδ in pulmonary fibrosis. RELEVANCE FOR PATIENTS: This study shows that the transcription factor C/EBPδ does not play a major role in the development of pulmonary fibrosis. Pharmacological targeting of C/EBPδ is therefore not likely to have a beneficial effect for patients suffering from pulmonary fibrosis.

15.
Thromb Haemost ; 114(3): 530-6, 2015 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-25994568

RESUMO

Already since the early 1800s, it has been recognised that malignancies may provoke thromboembolic complications, and indeed cancer patients are at increased risk of developing venous thrombosis. Interestingly, case control studies of deep-vein thrombosis suggested that low-molecular-weight heparin (LMWH) improved survival of cancer patients. This led to the hypothesis that cancer cells might 'take advantage' of a hypercoagulable state to more efficiently metastasise. Initial randomised placebo control trials showed that LMWH improve overall survival of cancer patients, especially in those patients with a relatively good prognosis. The failure of recent phase III trials, however, tempers enthusiasm for anticoagulant treatment in cancer patients despite an overwhelming body of literature showing beneficial effects of anticoagulants in preclinical models. Instead of discarding LMWH as potential (co)treatment modality in cancer patients, these disappointing recent trials should guide future preclinical research on anticoagulants in cancer biology. Most and for all, the underlying mechanisms by which coagulation drives tumour progression need to be elucidated. This could ultimately allow selection of cancer patients most likely to benefit from anticoagulant treatment and/or from targeted therapy downstream of coagulation factor signalling.


Assuntos
Anticoagulantes/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Tromboembolia/prevenção & controle , Humanos , Neoplasias/sangue , Neoplasias/complicações , Neoplasias/mortalidade , Seleção de Pacientes , Medição de Risco , Fatores de Risco , Tromboembolia/sangue , Tromboembolia/etiologia , Tromboembolia/mortalidade , Resultado do Tratamento
16.
Curr Pharm Des ; 17(1): 9-16, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21222644

RESUMO

Chronic inflammatory disorders constitute a heterogeneous group of complex and multifactorial diseases, which are often associated with increased cardiovascular morbidity and mortality, independent of the established cardiovascular risk factors. In keeping with this observation, hypercoagulability is frequently observed in patients suffering from atherosclerosis, chronic obstructive pulmonary disease and rheumatoid arthritis although the physiological significance of activated coagulation remained elusive. However, the identification of protease activated receptors (PAR) seem to provide a link between coagulation activation and disease progression as their activation by coagulation factors triggers a broad range of signaling pathways relevant for chronic inflammatory disorders. In experimental animal models, anticoagulation and/or genetic ablation of PAR signaling affords protection against the perpetuation of atherosclerosis, chronic obstructive pulmonary disease and rheumatoid arthritis. It is thus tempting to speculate that targeting the coagulation-PAR axis might have clinical relevance in the setting of chronic inflammatory disorders. In the current review, we discuss the current knowledge on coagulation activation in inflammatory disorders, we discuss the relationship between atherosclerosis, chronic obstructive pulmonary disease and rheumatoid arthritis and we review the current knowledge on PAR signaling in these disorders.


Assuntos
Coagulação Sanguínea/fisiologia , Inflamação/fisiopatologia , Receptores Ativados por Proteinase/fisiologia , Artrite Reumatoide/complicações , Artrite Reumatoide/fisiopatologia , Aterosclerose/complicações , Aterosclerose/fisiopatologia , Doença Crônica , Humanos , Inflamação/complicações , Modelos Biológicos , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Transdução de Sinais/fisiologia
17.
Curr Pharm Des ; 16(9): 1091-105, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20030609

RESUMO

The autonomous nervous system of the gut is increasingly recognized as an important regulatory factor in intestinal permeability and immune cell activation. Neuropeptides released by neurons -or inflammatory cells- have emerged as neuro-immune modulators that can relay, for instance, stress-induced neuronal activity to immune processes. Such peptides can participate in processes reducing inflammatory responses, or augment resolution of inflammation. Neuropeptides and hormones such as vasoactive intestinal peptide, urocortin, ghrelin, and cortistatin have been shown to modulate the disease activity in a variety of experimental models of inflammatory and autoimmune disease via modulation of immune or neuronal cell activity. We review here the potential of neuropeptide receptor activation to modulate inflammatory diseases of the intestine. We will highlight the role of neuropeptides in gastrointestinal (GI) physiology and immune regulation, and we will speculate on the therapeutic potential of peptides that bind G protein coupled receptors (GPCRs) in the management of inflammation in the GI tract.


Assuntos
Gastroenterite/tratamento farmacológico , Trato Gastrointestinal/fisiologia , Neuropeptídeos/uso terapêutico , Receptores de Neuropeptídeos/efeitos dos fármacos , Receptores de Neuropeptídeos/fisiologia , Animais , Sistema Nervoso Central/fisiologia , Gastroenterite/metabolismo , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/inervação , Humanos , Sistema Imunitário/fisiologia , Modelos Biológicos , Neuropeptídeos/farmacologia
18.
Exp Cell Res ; 313(12): 2622-33, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17531220

RESUMO

Coagulation Factor (F)Xa is a serine protease that plays a crucial role during blood coagulation by converting prothrombin into active thrombin. Recently, however, it emerged that besides this role in coagulation, FXa induces intracellular signaling leading to different cellular effects. Here, we show that coagulation factor (F)Xa drives tumor cells of epithelial origin, but not endothelial cells or monocytes, into apoptosis, whereas it even enhances fibroblast survival. FXa signals through the protease activated receptor (PAR)-1 to activate extracellular-signal regulated kinase (ERK) 1/2 and p38. This activation is associated with phosphorylation of the transcription factor CREB, and in tumor cells with up-regulation of the BH3-only pro-apoptotic protein Bim, leading to caspase-3 cleavage, the main hallmark of apoptosis. Transfection of tumor cells with dominant negative forms of CREB or siRNA for either PAR-1, Bim, ERK1 and/or p38 inhibited the pro-apoptotic effect of FXa. In fibroblasts, FXa-induced PAR-1 activation leads to down-regulation of Bim and pre-treatment with PAR-1 or Bim siRNA abolishes proliferation. We thus provide evidence that beyond its role in blood coagulation, FXa plays a key role in cellular processes in which Bim is the central player in determining cell survival.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/efeitos dos fármacos , Fator Xa/farmacologia , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas/genética , Regulação para Cima/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2 , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Receptor PAR-1/metabolismo , Transcrição Gênica/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA