Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Dent Res ; 102(9): 972-978, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37506226

RESUMO

The hemostatic and inflammatory systems work hand in hand to maintain homeostasis at mucosal barrier sites. Among the factors of the hemostatic system, fibrin is well recognized for its role in mucosal homeostasis, wound healing, and inflammation. Here, we present a basic overview of the fibrinolytic system, discuss fibrin as an innate immune regulator, and provide recent work uncovering the role of fibrin-neutrophil activation as a regulator of mucosal/periodontal homeostasis. We reason that the role of fibrin in periodontitis becomes most evident in individuals with the Mendelian genetic defect, congenital plasminogen (PLG) deficiency, who are predisposed to severe periodontitis in childhood due to a defect in fibrinolysis. Consistent with plasminogen deficiency being a risk factor for periodontitis, recent genomics studies uncover genetic polymorphisms in PLG, encoding plasminogen, being significantly associated with periodontal disease, and suggesting PLG variants as candidate risk indicators for common forms of periodontitis.


Assuntos
Hemostáticos , Periodontite , Humanos , Fibrinólise/fisiologia , Fibrinolisina , Plasminogênio/genética , Fibrina/fisiologia , Periodontite/genética
3.
J Thromb Haemost ; 14(8): 1618-28, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27079292

RESUMO

UNLABELLED: Essentials C57BL/6J-tissue plasminogen activator (tPA)-deficient mice are widely used to study tPA function. Congenic C57BL/6J-tPA-deficient mice harbor large 129-derived chromosomal segments. The 129-derived chromosomal segments contain gene mutations that may confound data interpretation. Passenger mutation-free isogenic tPA-deficient mice were generated for study of tPA function. SUMMARY: Background The ability to generate defined null mutations in mice revolutionized the analysis of gene function in mammals. However, gene-deficient mice generated by using 129-derived embryonic stem cells may carry large segments of 129 DNA, even when extensively backcrossed to reference strains, such as C57BL/6J, and this may confound interpretation of experiments performed in these mice. Tissue plasminogen activator (tPA), encoded by the PLAT gene, is a fibrinolytic serine protease that is widely expressed in the brain. A number of neurological abnormalities have been reported in tPA-deficient mice. Objectives To study genetic contamination of tPA-deficient mice. Materials and methods Whole genome expression array analysis, RNAseq expression profiling, low- and high-density single nucleotide polymorphism (SNP) analysis, bioinformatics and genome editing were used to analyze gene expression in tPA-deficient mouse brains. Results and conclusions Genes differentially expressed in the brain of Plat(-/-) mice from two independent colonies highly backcrossed onto the C57BL/6J strain clustered near Plat on chromosome 8. SNP analysis attributed this anomaly to about 20 Mbp of DNA flanking Plat being of 129 origin in both strains. Bioinformatic analysis of these 129-derived chromosomal segments identified a significant number of mutations in genes co-segregating with the targeted Plat allele, including several potential null mutations. Using zinc finger nuclease technology, we generated novel 'passenger mutation'-free isogenic C57BL/6J-Plat(-/-) and FVB/NJ-Plat(-/-) mouse strains by introducing an 11 bp deletion into the exon encoding the signal peptide. These novel mouse strains will be a useful community resource for further exploration of tPA function in physiological and pathological processes.


Assuntos
Mutação , Ativador de Plasminogênio Tecidual/genética , Alelos , Animais , Encéfalo/metabolismo , Cromossomos/ultraestrutura , Biologia Computacional , Cruzamentos Genéticos , Células-Tronco Embrionárias/citologia , Éxons , Feminino , Fibrinólise , Edição de Genes , Regulação da Expressão Gênica , Marcação de Genes , Genótipo , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Polimorfismo de Nucleotídeo Único , Sinais Direcionadores de Proteínas , Serina Proteases/metabolismo , Dedos de Zinco
4.
Oncogene ; 34(35): 4664-72, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-25486433

RESUMO

Deregulation of matriptase is a consistent feature of human epithelial cancers and correlates with poor disease outcome. We have previously shown that matriptase promotes multi-stage squamous cell carcinogenesis in transgenic mice through dual activation of pro-hepatocyte growth factor-cMet-Akt-mTor proliferation/survival signaling and PAR-2-Gαi-NFκB inflammatory signaling. Matriptase was congenitally and constitutively deregulated in our prior studies, and therefore it was unclear if aberrant matriptase signaling supports only initiation of tumor formation or if it is also critical for the progression of established tumors. To determine this, we here have generated triple-transgenic mice with constitutive deregulation of matriptase and simultaneous inducible expression of the cognate matriptase inhibitor, hepatocyte growth factor inhibitor (HAI)-2. As expected, constitutive expression of HAI-2 suppressed the formation of matriptase-dependent tumors in 7,12-Dimethylbenz(a)anthracene-treated mouse skin. Interestingly, however, the induction of HAI-2 expression in already established tumors markedly impaired malignant progression and caused regression of individual tumors. Tumor regression correlated with reduced accumulation of tumor-associated inflammatory cells, likely caused by diminished expression of pro-tumorigenic inflammatory cytokines. The data suggest that matriptase-dependent signaling may be a therapeutic target for both squamous cell carcinoma chemoprevention and for the treatment of established tumors.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Serina Endopeptidases/fisiologia , Neoplasias Cutâneas/enzimologia , Animais , Carcinogênese/imunologia , Carcinogênese/metabolismo , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/patologia , Citocinas/metabolismo , Progressão da Doença , Feminino , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Transgênicos , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia
5.
Oncogene ; 34(3): 346-56, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24469043

RESUMO

The membrane-anchored serine protease, matriptase, is consistently dysregulated in a range of human carcinomas, and high matriptase activity correlates with poor prognosis. Furthermore, matriptase is unique among tumor-associated proteases in that epithelial stem cell expression of the protease suffices to induce malignant transformation. Here, we use genetic epistasis analysis to identify proteinase-activated receptor (PAR)-2-dependent inflammatory signaling as an essential component of matriptase-mediated oncogenesis. In cell-based assays, matriptase was a potent activator of PAR-2, and PAR-2 activation by matriptase caused robust induction of nuclear factor (NF)κB through Gαi. Importantly, genetic elimination of PAR-2 from mice completely prevented matriptase-induced pre-malignant progression, including inflammatory cytokine production, inflammatory cell recruitment, epidermal hyperplasia and dermal fibrosis. Selective ablation of PAR-2 from bone marrow-derived cells did not prevent matriptase-driven pre-malignant progression, indicating that matriptase activates keratinocyte stem cell PAR-2 to elicit its pro-inflammatory and pro-tumorigenic effects. When combined with previous studies, our data suggest that dual induction of PAR-2-NFκB inflammatory signaling and PI3K-Akt-mTor survival/proliferative signaling underlies the transforming potential of matriptase and may contribute to pro-tumorigenic signaling in human epithelial carcinogenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Receptor PAR-2/metabolismo , Serina Endopeptidases/metabolismo , Proteínas ras/metabolismo , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular , Transformação Celular Neoplásica/genética , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Progressão da Doença , Células Epiteliais/patologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Imuno-Histoquímica , Queratinócitos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Receptor PAR-2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina Endopeptidases/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Proteínas ras/genética
6.
Thromb Haemost ; 112(2): 402-11, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24696092

RESUMO

Matriptase is a type-II transmembrane serine protease involved in epithelial homeostasis in both health and disease, and is implicated in the development and progression of a variety of cancers. Matriptase mediates its biological effects both via as yet undefined substrates and pathways, and also by proteolytic cleavage of a variety of well-defined protein substrates, several of which it shares with the closely-related protease hepsin. Development of targeted therapeutic strategies will require discrimination between these proteases. Here we have investigated cyclic microproteins of the squash Momordica cochinchinensis trypsin-inhibitor family (generated by total chemical synthesis) and found MCoTI-II to be a high-affinity (Ki 9 nM) and highly selective (> 1,000-fold) inhibitor of matriptase. MCoTI-II efficiently inhibited the proteolytic activation of pro-hepatocyte growth factor (HGF) by matriptase but not by hepsin, in both purified and cell-based systems, and inhibited HGF-dependent cell scattering. MCoTI-II also selectively inhibited the invasion of matriptase-expressing prostate cancer cells. Using a model of epithelial cell tight junction assembly, we also found that MCoTI-II could effectively inhibit the re-establishment of tight junctions and epithelial barrier function in MDCK-I cells after disruption, consistent with the role of matriptase in regulating epithelial integrity. Surprisingly, MCoTI-II was unable to inhibit matriptase-dependent proteolytic activation of prostasin, a GPI-anchored serine protease also implicated in epithelial homeostasis. These observations suggest that the unusually high selectivity afforded by MCoTI-II and its biological effectiveness might represent a useful starting point for the development of therapeutic inhibitors, and further highlight the role of matriptase in epithelial maintenance.


Assuntos
Ciclotídeos/farmacologia , Serina Endopeptidases/metabolismo , Inibidores de Serina Proteinase/farmacologia , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Cães , Impedância Elétrica , Células HEK293 , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Células Madin Darby de Rim Canino , Masculino , Terapia de Alvo Molecular , Invasividade Neoplásica , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Precursores de Proteínas/metabolismo , Serina Endopeptidases/genética , Especificidade por Substrato , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/enzimologia , Fatores de Tempo , Transfecção
7.
Oncogene ; 31(32): 3679-95, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22139080

RESUMO

Colitis-associated colorectal cancers are an etiologically distinct subgroup of colon cancers that occur in individuals suffering from inflammatory bowel disease and arise as a consequence of persistent exposure of hyperproliferative epithelial stem cells to an inflammatory microenvironment. An intrinsic defect in the intestinal epithelial barrier has been proposed to be one of several factors that contribute to the inappropriate immune response to the commensal microbiota that underlies inflammatory bowel disease. Matriptase is a membrane-anchored serine protease encoded by Suppression of Tumorigenicity-14 (ST14) that strengthens the intestinal epithelial barrier by promoting tight junction formation. Here, we show that intestinal epithelial-specific ablation of St14 in mice causes formation of colon adenocarcinoma with very early onset and high penetrance. Neoplastic progression is preceded by a chronic inflammation of the colon that resembles human inflammatory bowel disease and is promoted by the commensal microbiota. This study demonstrates that inflammation-associated colon carcinogenesis can be initiated and promoted solely by an intrinsic intestinal permeability barrier perturbation, establishes St14 as a critical tumor-suppressor gene in the mouse gastrointestinal tract and adds matriptase to the expanding list of pericellular proteases with tumor-suppressive functions.


Assuntos
Adenocarcinoma/enzimologia , Colite/enzimologia , Neoplasias do Colo/enzimologia , Genes Supressores de Tumor , Serina Endopeptidases/genética , Adenocarcinoma/patologia , Adenoma/enzimologia , Animais , Antibacterianos/farmacologia , Membrana Basal/enzimologia , Membrana Basal/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Colite/microbiologia , Colite/patologia , Colo/patologia , Neoplasias do Colo/patologia , Epitélio/enzimologia , Epitélio/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Doenças Inflamatórias Intestinais/enzimologia , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Absorção Intestinal , Proteínas de Membrana , Metagenoma/efeitos dos fármacos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica , Lesões Pré-Cancerosas , Serina Endopeptidases/metabolismo , Transdução de Sinais , beta Catenina/metabolismo
8.
Oncogene ; 30(17): 2003-16, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21217780

RESUMO

The progression and negative outcome of a variety of human carcinomas are intimately associated with aberrant activity of the c-Met oncogene. The underlying cause of this dysregulation, however, remains a subject of discussion, as the majority of cancer patients do not present with activating mutations in c-Met receptor itself. In this study, we show that the oncogenic protease matriptase is ubiquitously co-expressed with the c-Met in human squamous cell carcinomas and amplifies migratory and proliferative responses of primary epithelial cells to the cognate ligand for c-Met, pro-hepatocyte growth factor/scatter factor (proHGF/SF), through c-Met and Gab1 signaling. Furthermore, the selective genetic ablation of c-Met from matriptase-expressing keratinocytes completely negates the oncogenic potential of matriptase. In addition, matriptase-dependent carcinoma formation could be blocked by the pharmacological inhibition of the Akt-mammalian target of Rapamycin (mTor) pathway. Our data identify matriptase as an initiator of c-Met-Akt-mTor-dependent signaling axis in tumors and reveal mTor activation as an essential component of matriptase/c-Met-induced carcinogenesis. The study provides a specific example of how epithelial transformation can be promoted by epigenetic acquisition of the capacity to convert a widely available paracrine growth factor precursor to its signaling competent state.


Assuntos
Carcinoma de Células Escamosas/patologia , Transformação Celular Neoplásica , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Neoplasias de Cabeça e Pescoço/patologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Serina Endopeptidases/metabolismo , Animais , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/enzimologia , Neoplasias de Cabeça e Pescoço/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Queratinócitos/enzimologia , Queratinócitos/metabolismo , Queratinócitos/patologia , Camundongos , Precursores de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-met/deficiência , Proteínas Proto-Oncogênicas c-met/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima
9.
J Thromb Haemost ; 5 Suppl 1: 24-31, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17635705

RESUMO

Bacterial pathogens have frequently evolved and maintained the capacity to engage and/or activate hemostatic system components of their vertebrate hosts. Recent studies of mice with selected alterations in host plasminogen and other hemostatic factors have begun to reveal a seminal role of bacterial plasminogen activators and fibrin clearance in microbial pathogenesis. Bacterial pathogens appear to exploit host plasmin-mediated proteolysis to both support microbial dissemination and evade innate immune surveillance systems. The contribution of bacterial plasminogen activation to the evasion of the inflammatory response is particularly conspicuous with the plague agent, Yersinia pestis. Infection of control mice with wild-type Y. pestis leads to the formation of widespread foci containing massive numbers of free bacteria with little inflammatory cell infiltrate, whereas the loss of either the bacterial plasminogen activator, Pla, or the elimination of host plasminogen results in the accumulation of robust inflammatory cell infiltrates at sites of infection and greatly improved survival. Interestingly, fibrin(ogen) deficiency undermines the local inflammatory response observed with Pla-deficient Y. pestis and effectively eliminates the survival benefits posed by the elimination of either host plasminogen or bacterial Pla. These studies, and complementary studies with other human pathogens, illustrate that plasminogen and fibrinogen are extremely effective modifiers of the inflammatory response in vivo and critical determinants of bacterial virulence and host defense. Detailed studies of the inflammatory response in mice with genetically-imposed modifications in coagulation and fibrinolytic factors underscore the regulatory crosstalk between the hemostatic and immune systems.


Assuntos
Infecções Bacterianas/fisiopatologia , Fibrina/fisiologia , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Fibrinólise , Humanos , Camundongos , Plasminogênio/fisiologia , Yersinia pestis/fisiologia
10.
Oncogene ; 26(11): 1546-56, 2007 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-16983341

RESUMO

Hepatocyte growth factor activator inhibitor-1 (HAI-1) is a Kunitz-type transmembrane serine protease inhibitor that forms inhibitor complexes with several trypsin-like serine proteases and is required for mouse placental development and embryo survival. Here we show that the essential function of HAI-1 in placentation and all other embryonic processes is to restrict the activity of the type II transmembrane serine protease, matriptase. Enzymatic gene trapping of matriptase combined with HAI-1 immunohistochemistry revealed that matriptase is co-expressed with HAI-1 in both extraembryonic and embryonic tissues. As early as embryonic day 8.5, matriptase and HAI-1 were expressed in a population of chorionic trophoblasts. Ablation of HAI-1 disrupted the epithelial integrity of this cell population, causing disorganized laminin deposition and altered expression of E-cadherin and beta-catenin. This led to a complete loss of undifferentiated chorionic trophoblasts after embryonic day 9.5 and prevented the formation of the placental labyrinth. Genetic ablation of matriptase activity in HAI-1-deficient embryos, however, restored the integrity of chorionic trophoblasts and enabled placental labyrinth formation and development to term. Furthermore, matriptase/HAI-1 double-deficient mice were phenotypically indistinguishable from matriptase single-deficient littermates.


Assuntos
Glicoproteínas de Membrana/fisiologia , Placentação , Serina Endopeptidases/efeitos dos fármacos , Animais , Sequência de Bases , Primers do DNA , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Proteínas Secretadas Inibidoras de Proteinases
11.
Lab Invest ; 81(10): 1403-14, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11598153

RESUMO

The urokinase plasminogen activator receptor-associated protein/Endo180 (uPARAP/Endo180) is a newly discovered member of the macrophage mannose receptor family that was reported to interact with ligand-bound urokinase plasminogen activator receptor (uPAR), matrix metalloprotease-13 (MMP-13), and collagen V on the cell surface. We have determined the sites of expression of this novel receptor during murine postimplantation development. uPARAP/Endo180 was expressed in all tissues undergoing primary ossification, including the developing bones of the viscerocranium and calvarium that ossify intramembranously, and developing long bones undergoing endochondral ossification. uPARAP/Endo180 mRNA was expressed by both immature osteoblasts and by mature osteocalcin-producing osteoblasts-osteocytes, and was coexpressed with MMP-13. Interestingly, osteoblasts also expressed uPAR. Besides bone-forming tissues, uPARAP/Endo180 expression was detected only in a mesenchymal condensation of the midbrain and in the developing lungs. The data suggest a function of this novel protease receptor in bone development, possibly mediated through its interactions with uPAR, MMP-13, or collagen V.


Assuntos
Osso e Ossos/fisiologia , Colagenases/biossíntese , Receptores de Superfície Celular/biossíntese , Receptores Mitogênicos/biossíntese , Animais , Osso e Ossos/embriologia , Desenvolvimento Embrionário e Fetal , Feminino , Imuno-Histoquímica , Metaloproteinase 13 da Matriz , Camundongos , Osteogênese/fisiologia , Gravidez , Receptores de Ativador de Plasminogênio Tipo Uroquinase
12.
J Biol Chem ; 276(21): 17976-84, 2001 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-11278833

RESUMO

Urokinase plasminogen activator receptor (uPAR) binds pro-urokinase plasminogen activator (pro-uPA) and thereby localizes it near plasminogen, causing the generation of active uPA and plasmin on the cell surface. uPAR and uPA are overexpressed in a variety of human tumors and tumor cell lines, and expression of uPAR and uPA is highly correlated to tumor invasion and metastasis. To exploit these characteristics in the design of tumor cell-selective cytotoxins, we constructed mutated anthrax toxin-protective antigen (PrAg) proteins in which the furin cleavage site is replaced by sequences cleaved specifically by uPA. These uPA-targeted PrAg proteins were activated selectively on the surface of uPAR-expressing tumor cells in the presence of pro-uPA and plasminogen. The activated PrAg proteins caused internalization of a recombinant cytotoxin, FP59, consisting of anthrax toxin lethal factor residues 1-254 fused to the ADP-ribosylation domain of Pseudomonas exotoxin A, thereby killing the uPAR-expressing tumor cells. The activation and cytotoxicity of these uPA-targeted PrAg proteins were strictly dependent on the integrity of the tumor cell surface-associated plasminogen activation system. We also constructed a mutated PrAg protein that selectively killed tissue plasminogen activator-expressing cells. These mutated PrAg proteins may be useful as new therapeutic agents for cancer treatment.


Assuntos
Antígenos de Bactérias , Toxinas Bacterianas/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/uso terapêutico , Toxinas Bacterianas/toxicidade , Morte Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Humanos , Camundongos , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Plasminogênio/metabolismo , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas
13.
Am J Pathol ; 158(3): 921-9, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238040

RESUMO

Tissue repair requires an adequate cellular proliferation coordinated with the timely proteolysis of matrix elements. Based on the properties of plasminogen activators in liver cell proliferation and tissue proteolysis, we explored the regulatory role of tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) in liver repair. Using carbon tetrachloride (CCl(4)) intoxication as a model of acute liver injury, we found that tPA-deficient mice displayed a mild defect in hepatic repair, whereas livers of uPA-deficient mice had a more substantial delay in repair, with injury of centrilobular hepatocytes persisting up to 14 days after CCl(4). Notably, functional cooperativity between plasminogen activators was strongly inferred from the profound reparative defect in livers of mice lacking tPA and uPA simultaneously, with persistence of centrilobular injury as far out as 35 days. The defective repair was not because of increased susceptibility of experimental mice to the toxin or to inadequate cellular proliferation. Instead, lack of plasminogen activators led to the accumulation of fibrin and fibronectin within injured areas and poor removal of necrotic cells. These data demonstrate that tPA and uPA play a critical role in hepatic repair via proteolysis of matrix elements and clearance of cellular debris from the field of injury.


Assuntos
Hepatopatias/enzimologia , Regeneração Hepática , Fígado/enzimologia , Ativador de Plasminogênio Tecidual/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Doença Aguda , Animais , Tetracloreto de Carbono , Divisão Celular , Proteínas da Matriz Extracelular/metabolismo , Fibrina/metabolismo , Marcação de Genes , Fígado/citologia , Fígado/patologia , Hepatopatias/etiologia , Hepatopatias/patologia , Camundongos , Camundongos Mutantes , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tipo Uroquinase/genética
14.
Blood ; 96(10): 3302-9, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11071621

RESUMO

Detailed studies of tumor cell-associated procoagulants and fibrinolytic factors have implied that local thrombin generation and fibrin deposition and dissolution may be important in tumor growth and dissemination. To directly determine whether fibrin(ogen) or plasmin(ogen) are determinants of the metastatic potential of circulating tumor cells, this study examined the impact of genetic deficits in each of these key hemostatic factors on the hematogenous pulmonary metastasis of 2 established murine tumors, Lewis lung carcinoma and the B16-BL6 melanoma. In both tumor models, fibrinogen deficiency strongly diminished, but did not prevent, the development of lung metastasis. The quantitative reduction in metastasis in fibrinogen-deficient mice was not due to any appreciable difference in tumor stroma formation or tumor growth. Rather, tumor cell fate studies indicated an important role for fibrin(ogen) in sustained adhesion and survival of tumor cells within the lung. The specific thrombin inhibitor, hirudin, further diminished the metastatic potential of circulating tumor cells in fibrinogen-deficient mice, although the inhibitor had no apparent effect on tumor cell proliferation in vitro. The absence of plasminogen and plasmin-mediated fibrinolysis had no significant impact on hematogenous metastasis. The authors concluded that fibrin(ogen) is a critical determinant of the metastatic potential of circulating tumor cells. Furthermore, thrombin appears to facilitate tumor dissemination through at least one fibrin(ogen)-independent mechanism. These findings suggest that therapeutic strategies focusing on multiple distinct hemostatic factors might be beneficial in the containment of tumor metastasis.


Assuntos
Fibrinogênio/farmacologia , Metástase Neoplásica/fisiopatologia , Células Neoplásicas Circulantes/efeitos dos fármacos , Animais , Carcinoma Pulmonar de Lewis/sangue , Carcinoma Pulmonar de Lewis/patologia , Adesão Celular/efeitos dos fármacos , Modelos Animais de Doenças , Fibrinogênio/genética , Fibrinogênio/fisiologia , Fibrinolisina/farmacologia , Fibrinólise/efeitos dos fármacos , Fibrinólise/fisiologia , Fibrinolíticos/farmacologia , Hemostasia , Hirudinas/farmacologia , Histocitoquímica , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/sangue , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Metástase Neoplásica/patologia , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patologia , Neovascularização Patológica , Trombina/antagonistas & inibidores , Trombina/farmacologia
15.
Development ; 127(20): 4481-92, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11003846

RESUMO

Urokinase-type plasminogen activator expression is induced in the mouse mammary gland during development and post-lactational involution. We now show that primiparous plasminogen-deficient (Plg(-/-)) mice have seriously compromised mammary gland development and involution. All mammary glands were underdeveloped and one-quarter of the mice failed to lactate. Although the glands from lactating Plg(-/-) mice were initially smaller, they failed to involute after weaning, and in most cases they failed to support a second litter. Alveolar regression was markedly reduced and a fibrotic stroma accumulated in Plg(-/-) mice. Nevertheless, urokinase and matrix metalloproteinases (MMPs) were upregulated normally in involuting glands of Plg(-/-) mice, and fibrin did not accumulate in the glands. Heterozygous Plg(+/-) mice exhibited haploinsufficiency, with a definite, but less severe mammary phenotype. These data demonstrate a critical, dose-dependent requirement for Plg in lactational differentiation and mammary gland remodeling during involution.


Assuntos
Lactação/fisiologia , Glândulas Mamárias Animais/fisiologia , Plasminogênio/deficiência , Animais , Diferenciação Celular , Matriz Extracelular/metabolismo , Feminino , Fertilidade/fisiologia , Fibrina/metabolismo , Glândulas Mamárias Animais/citologia , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Mutantes , Plasminogênio/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
16.
Circ Res ; 87(2): 133-9, 2000 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-10903997

RESUMO

Extracellular proteolysis is likely to be a feature of vascular remodeling associated with atherosclerotic and restenotic arteries. To investigate the role of plasminogen-mediated proteolysis in remodeling, polyethylene cuffs were placed around the femoral arteries of mice with single and combined deficiencies in plasminogen and fibrinogen. Neointimal development occurred in all mice and was unaffected by genotype. Significant compensatory medial remodeling occurred in the cuffed arteries of control mice but not in plasminogen-deficient mice. Furthermore, focal areas of medial atrophy were frequently observed in plasminogen-deficient mice but not in control animals. A simultaneous deficit of fibrinogen restored the potential of the arteries of plasminogen-deficient mice to enlarge in association with neointimal development but did not eliminate the focal medial atrophy. An intense inflammatory infiltrate occurred in the adventitia of cuffed arteries, which was associated with enhanced matrix deposition. Adventitial collagen deposition was apparent after 28 days in control and fibrinogen-deficient arteries but not in plasminogen-deficient arteries, which contained persistent fibrin. These studies demonstrate that plasmin(ogen) contributes to favorable arterial remodeling and adventitial collagen deposition via a mechanism that is related to fibrinogen, presumably fibrinolysis. In addition, these studies reveal a fibrin-independent role of plasminogen in preventing medial atrophy in challenged vessels.


Assuntos
Artéria Femoral/fisiopatologia , Fibrinogênio/fisiologia , Plasminogênio/fisiologia , Túnica Íntima/fisiopatologia , Animais , Colágeno/metabolismo , Cruzamentos Genéticos , Artéria Femoral/patologia , Artéria Femoral/fisiologia , Inflamação , Macrófagos/fisiologia , Camundongos , Camundongos Knockout , Plasminogênio/deficiência , Túnica Íntima/patologia , Túnica Íntima/fisiologia
17.
J Cell Biol ; 148(6): 1295-304, 2000 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-10725341

RESUMO

Short seizure episodes are associated with remodeling of neuronal connections. One region where such reorganization occurs is the hippocampus, and in particular, the mossy fiber pathway. Using genetic and pharmacological approaches, we show here a critical role in vivo for tissue plasminogen activator (tPA), an extracellular protease that converts plasminogen to plasmin, to induce mossy fiber sprouting. We identify DSD-1-PG/phosphacan, an extracellular matrix component associated with neurite reorganization, as a physiological target of plasmin. Mice lacking tPA displayed decreased mossy fiber outgrowth and an aberrant band at the border of the supragranular region of the dentate gyrus that coincides with the deposition of unprocessed DSD-1-PG/phosphacan and excessive Timm-positive, mossy fiber termini. Plasminogen-deficient mice also exhibit the laminar band and DSD- 1-PG/phosphacan deposition, but mossy fiber outgrowth through the supragranular region is normal. These results demonstrate that tPA functions acutely, both through and independently of plasmin, to mediate mossy fiber reorganization.


Assuntos
Proteoglicanas de Sulfatos de Condroitina/fisiologia , Fibrinolisina/metabolismo , Hipocampo/fisiologia , Fibras Nervosas/fisiologia , Neuritos/fisiologia , Plasminogênio/metabolismo , Convulsões/fisiopatologia , Ativador de Plasminogênio Tecidual/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Tonsila do Cerebelo/fisiopatologia , Animais , Hipocampo/fisiopatologia , Hipocampo/ultraestrutura , Ácido Caínico/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/ultraestrutura , Neuritos/efeitos dos fármacos , Neuritos/ultraestrutura , Plasminogênio/deficiência , Plasminogênio/genética , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores , Convulsões/induzido quimicamente , Ativador de Plasminogênio Tecidual/deficiência , Ativador de Plasminogênio Tecidual/genética
18.
Biochemistry ; 39(3): 508-15, 2000 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-10642175

RESUMO

The plasminogen activation (PA) system is involved in the degradation of fibrin and various extracellular matrix proteins, taking part in a number of physiological and pathological tissue remodeling processes including cancer invasion. This system is organized as a classical proteolytic cascade, and as for other cascade systems, understanding the physiological initiation mechanism is of central importance. The attempts to identify initiation routes for activation of the proform of the key enzyme urokinase-type plasminogen activator (pro-uPA) in vivo have been hampered by the strong activator potency of the plasmin, that is generated during the progress of the cascade. Using gene-targeted mice deficient in plasminogen (Plg -/- mice) [Bugge, T. H., Flick, M. J., Daugherty, C. C., and Degen, J. L. (1995) Genes Dev. 9, 794-807], we have now demonstrated and identified a component capable of initiating the cascade by activating pro-uPA. The urine from Plg -/- mice contained active two-chain uPA as well as a proteinase capable of activating exogenously added pro-uPA. The active component was purified and identified by mass spectrometry-based peptide mapping as mouse glandular kallikrein mGK-6 (true tissue kallikrein). The pro-uPA converting activity of the mGK-6 enzyme, as well as its ability to cleave a synthetic substrate for glandular kallikrein, was inhibited by the serine proteinase inhibitor leupeptin but not by other serine proteinase inhibitors such as aprotinin, antithrombin III, or alpha(1)-antitrypsin. We suggest that mouse glandular kallikrein mGK-6 is an activator of pro-uPA in the mouse urinary tract in vivo. Since this kallikrein is expressed in a number of tissues and also occurs in plasma, it can also be considered a candidate for a physiological pro-uPA activator in other locations.


Assuntos
Plasminogênio/metabolismo , Calicreínas Teciduais/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Animais , Ativação Enzimática , Precursores Enzimáticos/metabolismo , Fibrinolisina/metabolismo , Humanos , Substâncias Macromoleculares , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Plasminogênio/deficiência , Plasminogênio/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Trombina/metabolismo , Calicreínas Teciduais/isolamento & purificação , Calicreínas Teciduais/urina , Ativador de Plasminogênio Tipo Uroquinase/química
19.
Invest Ophthalmol Vis Sci ; 41(1): 67-72, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10634603

RESUMO

PURPOSE: Excimer laser photorefractive keratectomy creates a nonvascular wound of the cornea. Fibrin deposition and resolution after excimer laser photokeratectomy were investigated in relation to corneal repair and restoration of clarity in mice with a genetic deficiency of plasminogen. METHODS: A Summit Apex Laser (Summit, Waltham, MA) was used to perform 2-mm, 175-pulse, transepithelial photoablations that resulted in deep stromal keratectomies. Photokeratectomy was performed on the corneas of plasminogen-deficient (Plg-/-) mice and littermate control animals. Eyes were examined for re-epithelialization and clarity throughout the 21-day observational period. Histologic sections were taken during the observational period and fibrin(ogen) was detected immunohistochemically. RESULTS: Re-epithelialization was rapid and complete within 3 days in both control and Plg-/- animals. Exuberant corneal fibrin(ogen) deposition was noted in Plg-/- mice and sparse fibrin(ogen) deposition in control mice on days 1 and 3 after injury. Fibrin(ogen) deposits resolved in control mice but persisted in Plg-/- mice (74% of eyes at 21 days; P < 0.004). Corneal opacification, scarring, and the presence of anterior chamber fibrin(ogen) occurred in plasminogen-deficient mice but not in control mice. CONCLUSIONS: Fibrin(ogen) deposition occurs during corneal wound repair after photokeratectomy. Impaired fibrinolysis in Plg-/- mice caused persistent stromal fibrin deposits that correlated with the development of corneal opacity.


Assuntos
Córnea/patologia , Opacidade da Córnea/etiologia , Ceratectomia Fotorrefrativa/efeitos adversos , Plasminogênio/deficiência , Animais , Córnea/metabolismo , Córnea/cirurgia , Opacidade da Córnea/metabolismo , Opacidade da Córnea/patologia , Fibrina/metabolismo , Fibrinogênio/metabolismo , Técnicas Imunoenzimáticas , Lasers de Excimer , Camundongos , Camundongos Mutantes , Cicatrização
20.
Proc Natl Acad Sci U S A ; 96(26): 15143-8, 1999 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-10611352

RESUMO

Cellular proliferation and tissue remodeling are central to the regenerative response after a toxic injury to the liver. To explore the role of plasminogen in hepatic tissue remodeling and regeneration, we used carbon tetrachloride to induce an acute liver injury in plasminogen-deficient (Plg(o)) mice and nontransgenic littermates (Plg(+)). On day 2 after CCl(4), livers of Plg(+) and Plg(o) mice had a similar diseased pale/lacy appearance, followed by restoration of normal appearance in Plg(+) livers by day 7. In contrast, Plg(o) livers remained diseased for as long as 2.5 months, with a diffuse pale/lacy appearance and persistent damage to centrilobular hepatocytes. The persistent centrilobular lesions were not a consequence of impaired proliferative response in Plg(o) mice. Notably, fibrin deposition was a prominent feature in diseased centrilobular areas in Plg(o) livers for at least 30 days after injury. Nonetheless, the genetically superimposed loss of the Aalpha fibrinogen chain (Plg(o)/Fib(o) mice) did not correct the abnormal phenotype. These data show that plasminogen deficiency impedes the clearance of necrotic tissue from a diseased hepatic microenvironment and the subsequent reconstitution of normal liver architecture in a fashion that is unrelated to circulating fibrinogen.


Assuntos
Fígado/fisiologia , Plasminogênio/deficiência , Afibrinogenemia/genética , Animais , Divisão Celular/efeitos dos fármacos , Clorofórmio/toxicidade , Fibrina/biossíntese , Fibrinogênio/genética , Fígado/citologia , Fígado/efeitos dos fármacos , Regeneração Hepática/fisiologia , Camundongos , Camundongos Transgênicos , Plasminogênio/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA