Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cell Biol ; 222(11)2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37737895

RESUMO

The transmembrane serine protease matriptase is a key regulator of both barrier-disruptive and protective epithelial cell-cell interactions. Elevated matriptase is a consistent feature of epithelial ovarian cancers (OvCa), where multicellular spheroids shed from the primary tumor into the peritoneal cavity are critical drivers of metastasis. Dynamic cell-to-cell adhesive contacts are required for spheroid formation and maintenance. Here, we show that overactive matriptase, reflected in an increased ratio of matriptase to its inhibitor hepatocyte growth factor activator inhibitor 1 (HAI-1), disrupts cell-cell contacts to produce loose prometastatic spheroids that display increased mesothelial cell adhesion and submesothelial invasion. We show that these activities are dependent on the matriptase activation of a protease-activated receptor-2 (PAR-2) signaling pathway involving PI3K/Akt and MMP9-induced disruption of cell-cell adhesion by the release of the soluble E-cadherin ectodomain. These data reveal a novel pathological connection between matriptase activation of PAR-2 and disruption of cell-cell adhesion, and support the clinical investigation of this signaling axis as a therapeutic strategy for aggressive metastatic OvCa.


Assuntos
Neoplasias Ovarianas , Serina Endopeptidases , Transdução de Sinais , Feminino , Humanos , Metaloproteinase 9 da Matriz/genética , Neoplasias Ovarianas/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Esferoides Celulares , Serina Endopeptidases/metabolismo
2.
mBio ; 14(5): e0120823, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37768050

RESUMO

IMPORTANCE: Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, impacts millions of individuals worldwide and severely impairs the quality of life for patients. Dysregulation of innate immune signaling pathways reduces barrier function and exacerbates disease progression. Macrophage (Mφ) signaling pathways are potential targets for IBD therapies. While multiple treatments are available for IBD, (i) not all patients respond, (ii) responses may diminish over time, and (iii) treatments often have undesirable side effects. Genetic studies have shown that the inheritance of two co-segregating SNPs expressed in the innate immune receptor, TLR4, is associated with human IBD. Mice expressing homologous SNPs ("TLR4-SNP" mice) exhibited more severe colitis than WT mice in a DSS-induced colonic inflammation/repair model. We identified a critical role for M2a "tissue repair" Mφ in the resolution of colitis. Our findings provide insight into potential development of novel therapies targeting Mφ signaling pathways that aim to alleviate the debilitating symptoms experienced by individuals with IBD.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Humanos , Camundongos , Animais , Receptor 4 Toll-Like , Polimorfismo de Nucleotídeo Único , Qualidade de Vida , Colite/induzido quimicamente , Macrófagos , Doenças Inflamatórias Intestinais/induzido quimicamente , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
3.
Int J Mol Sci ; 24(11)2023 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-37298257

RESUMO

Hemostasis is a delicate balance between coagulation and fibrinolysis that regulates the formation and removal of fibrin, respectively. Positive and negative feedback loops and crosstalk between coagulation and fibrinolytic serine proteases maintain the hemostatic balance to prevent both excessive bleeding and thrombosis. Here, we identify a novel role for the glycosylphosphatidylinositol (GPI)-anchored serine protease testisin in the regulation of pericellular hemostasis. Using in vitro cell-based fibrin generation assays, we found that the expression of catalytically active testisin on the cell surface accelerates thrombin-dependent fibrin polymerization, and intriguingly, that it subsequently promotes accelerated fibrinolysis. We find that the testisin-dependent fibrin formation is inhibited by rivaroxaban, a specific inhibitor of the central prothrombin-activating serine protease factor Xa (FXa), demonstrating that cell-surface testisin acts upstream of factor X (FX) to promote fibrin formation at the cell surface. Unexpectedly, testisin was also found to accelerate fibrinolysis by stimulating the plasmin-dependent degradation of fibrin and enhancing plasmin-dependent cell invasion through polymerized fibrin. Testisin was not a direct activator of plasminogen, but it is able to induce zymogen cleavage and the activation of pro-urokinase plasminogen activator (pro-uPA), which converts plasminogen to plasmin. These data identify a new proteolytic component that can regulate pericellular hemostatic cascades at the cell surface, which has implications for angiogenesis, cancer biology, and male fertility.


Assuntos
Fibrinólise , Hemostáticos , Masculino , Humanos , Fibrinólise/fisiologia , Fibrinolisina/metabolismo , Glicosilfosfatidilinositóis , Serina Proteases , Serina Endopeptidases/metabolismo , Plasminogênio/metabolismo , Ativador de Plasminogênio Tipo Uroquinase , Fibrina/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(28): e2201423119, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35867758

RESUMO

Treatments for advanced and recurrent ovarian cancer remain a challenge due to a lack of potent, selective, and effective therapeutics. Here, we developed the basis for a transformative anticancer strategy based on anthrax toxin that has been engineered to be selectively activated by the catalytic power of zymogen-activating proteases on the surface of malignant tumor cells to induce cell death. Exposure to the engineered toxin is cytotoxic to ovarian tumor cell lines and ovarian tumor spheroids derived from patient ascites. Preclinical studies demonstrate that toxin treatment induces tumor regression in several in vivo ovarian cancer models, including patient-derived xenografts, without adverse side effects, supportive of progression toward clinical evaluation. These data lay the groundwork for developing therapeutics for treating women with late-stage and recurrent ovarian cancers, utilizing a mechanism distinct from current anticancer therapies.


Assuntos
Antígenos de Bactérias , Antineoplásicos , Toxinas Bacterianas , Neoplasias Ovarianas , Pró-Fármacos , Serina Proteases , Antígenos de Bactérias/farmacologia , Antígenos de Bactérias/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Toxinas Bacterianas/farmacologia , Toxinas Bacterianas/uso terapêutico , Linhagem Celular Tumoral , Precursores Enzimáticos/metabolismo , Feminino , Humanos , Recidiva Local de Neoplasia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Serina Proteases/metabolismo , Esferoides Celulares , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Front Immunol ; 10: 1348, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31258531

RESUMO

Clinical observations and accumulating laboratory evidence support a complex interplay between coagulation, inflammation, innate immunity and fibrinolysis in venous thromboembolism (VTE). VTE, which includes deep vein thrombosis (DVT) and pulmonary embolism (PE), and the subsequent complications of post-thrombotic syndrome (PTS), are significant causes of morbidity and mortality in patients. Clinical risk factors for VTE include cancer, major trauma, surgery, sepsis, inflammatory bowel disease, paralysis, prolonged periods of immobility, and aging. Abnormalities in venous blood flow or stasis initiates the activation of endothelial cells, and in concert with platelets, neutrophils and monocytes, propagates VTE in an intact vein. In addition, inflammatory cells play crucial roles in thrombus recanalization and restoration of blood flow via fibrinolysis and vascular remodeling. Faster resolution of the thrombus is key for improved disease prognosis. While in the clinical setting, anticoagulation therapy is successful in preventing propagation of venous thrombi, current therapies are not designed to inhibit inflammation, which can lead to the development of PTS. Animal models of DVT have provided many insights into the molecular and cellular mechanisms involved in the formation, propagation, and resolution of venous thrombi as well as the roles of key components of the fibrinolytic system in these processes. Here, we review the recent advances in our understanding of fibrinolysis and inflammation in the resolution of VTE.


Assuntos
Células Endoteliais/fisiologia , Inflamação/imunologia , Trombose Venosa/imunologia , Animais , Coagulação Sanguínea , Modelos Animais de Doenças , Fibrinólise , Humanos , Imunidade Inata
7.
J Mol Med (Berl) ; 97(5): 691-709, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30911775

RESUMO

Ovarian cancer is the leading cause of death among all the gynecological cancers in the USA. Ovarian cancer employs a unique mode of metastasis, as exfoliated tumor cells disseminate within the peritoneal cavity, colonizing in several sites as well as accumulating ascites. Tumor recurrence and widespread metastasis are significant factors contributing to poor prognosis. PRSS21 is a metastasis-associated ovarian cancer gene that encodes the glycosyl-phosphatidylinositol-linked serine protease, testisin. Testisin expression is increased in multiple ovarian tumor types, with relatively little expression in normal tissues, but is differentially decreased in metastatic ovarian serous carcinomas compared to primary tumors. Here we explored the function of testisin in late-stage ovarian cancer progression using a murine xenograft model of ovarian intraperitoneal tumor metastasis. Increased tumor testisin expression inhibited intra-peritoneal tumor seeding and colonization, ascites accumulation, and metastatic tumor burden that was dependent on catalytically active testisin. The known testisin substrate, protease-activated receptor-2 (PAR-2), is a target of testisin activity. Gene profiling and mechanistic studies demonstrate that testisin activity suppresses the synthesis and secretion of pro-angiogenic angiopoietins, ANG2 and ANGPTL4, which normally promote vascular leak and edema. These observations support a model wherein testisin activates PAR-2 to antagonize proangiogenic angiopoietins that modulate vascular permeability and ascites accumulation associated with ovarian tumor metastasis. KEY MESSAGES: Testisin inhibits metastatic ovarian tumor burden and ascites production. Testisin activity antagonizes ANG2 and ANGPTL4 synthesis and secretion. PAR-2 is a proteolytic target of testisin on the surface of ovarian cancer cells.


Assuntos
Proteína 4 Semelhante a Angiopoietina/metabolismo , Neoplasias Ovarianas/metabolismo , Ribonuclease Pancreático/metabolismo , Serina Endopeptidases/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Proteínas Ligadas por GPI/metabolismo , Humanos , Camundongos Nus , Metástase Neoplásica/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia , Proteólise , Proteínas de Transporte Vesicular/metabolismo
8.
Cancer Res ; 79(2): 301-310, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30610085

RESUMO

Pericellular proteolysis provides a significant advantage to developing tumors through the ability to remodel the extracellular matrix, promote cell invasion and migration, and facilitate angiogenesis. Recent advances demonstrate that pericellular proteases can also communicate directly to cells by activation of a unique group of transmembrane G-protein-coupled receptors (GPCR) known as protease-activated receptors (PAR). In this review, we discuss the specific roles of one of four mammalian PARs, namely PAR-2, which is overexpressed in advanced stage tumors and is activated by trypsin-like serine proteases that are highly expressed or otherwise dysregulated in many cancers. We highlight recent insights into the ability of different protease agonists to bias PAR-2 signaling and the newly emerging evidence for an interplay between PAR-2 and membrane-anchored serine proteases, which may co-conspire to promote tumor progression and metastasis. Interfering with these pathways might provide unique opportunities for the development of new mechanism-based strategies for the treatment of advanced and metastatic cancers.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Receptor PAR-2/metabolismo , Serina Proteases/metabolismo , Animais , Membrana Celular/enzimologia , Membrana Celular/metabolismo , Progressão da Doença , Glicosilfosfatidilinositóis/metabolismo , Humanos , Neoplasias/enzimologia , Transdução de Sinais
9.
J Biol Chem ; 292(26): 10801-10812, 2017 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-28490634

RESUMO

Compromised gastrointestinal barrier function is strongly associated with the progressive and destructive pathologies of the two main forms of irritable bowel disease (IBD), ulcerative colitis (UC), and Crohn's disease (CD). Matriptase is a membrane-anchored serine protease encoded by suppression of tumorigenicity-14 (ST14) gene, which is critical for epithelial barrier development and homeostasis. Matriptase barrier-protective activity is linked with the glycosylphosphatidylinositol (GPI)-anchored serine protease prostasin, which is a co-factor for matriptase zymogen activation. Here we show that mRNA and protein expression of both matriptase and prostasin are rapidly down-regulated in the initiating inflammatory phases of dextran sulfate sodium (DSS)-induced experimental colitis in mice, and, significantly, the loss of these proteases precedes the appearance of clinical symptoms, suggesting their loss may contribute to disease susceptibility. We used heterozygous St14 hypomorphic mice expressing a promoter-linked ß-gal reporter to show that inflammatory colitis suppresses the activity of the St14 gene promoter. Studies in colonic T84 cell monolayers revealed that barrier disruption by the colitis-associated Th2-type cytokines, IL-4 and IL-13, down-regulates matriptase as well as prostasin through phosphorylation of the transcriptional regulator STAT6 and that inhibition of STAT6 with suberoylanilide hydroxamic acid (SAHA) restores protease expression and reverses cytokine-induced barrier dysfunction. Both matriptase and prostasin are significantly down-regulated in colonic tissues from human subjects with active ulcerative colitis or Crohn's disease, implicating the loss of this barrier-protective protease pathway in the pathogenesis of irritable bowel disease.


Assuntos
Colite Ulcerativa/metabolismo , Doença de Crohn/metabolismo , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Serina Endopeptidases/metabolismo , Animais , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/genética , Colite Ulcerativa/patologia , Colo/metabolismo , Colo/patologia , Doença de Crohn/induzido quimicamente , Doença de Crohn/genética , Doença de Crohn/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Humanos , Ácidos Hidroxâmicos/farmacologia , Interleucina-13/genética , Interleucina-4/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Mutantes , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Serina Endopeptidases/genética , Vorinostat
10.
Oncotarget ; 6(32): 33534-53, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26392335

RESUMO

The membrane-anchored serine proteases are a unique group of trypsin-like serine proteases that are tethered to the cell surface via transmembrane domains or glycosyl-phosphatidylinositol-anchors. Overexpressed in tumors, with pro-tumorigenic properties, they are attractive targets for protease-activated prodrug-like anti-tumor therapies. Here, we sought to engineer anthrax toxin protective antigen (PrAg), which is proteolytically activated on the cell surface by the proprotein convertase furin to instead be activated by tumor cell-expressed membrane-anchored serine proteases to function as a tumoricidal agent. PrAg's native activation sequence was mutated to a sequence derived from protein C inhibitor (PCI) that can be cleaved by membrane-anchored serine proteases, to generate the mutant protein PrAg-PCIS. PrAg-PCIS was resistant to furin cleavage in vitro, yet cytotoxic to multiple human tumor cell lines when combined with FP59, a chimeric anthrax toxin lethal factor-Pseudomonas exotoxin fusion protein. Molecular analyses showed that PrAg-PCIS can be cleaved in vitro by several serine proteases including the membrane-anchored serine protease testisin, and mediates increased killing of testisin-expressing tumor cells. Treatment with PrAg-PCIS also potently attenuated the growth of testisin-expressing xenograft tumors in mice. The data indicates PrAg can be engineered to target tumor cell-expressed membrane-anchored serine proteases to function as a potent tumoricidal agent.


Assuntos
Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Pró-Fármacos/farmacologia , Serina Endopeptidases/farmacologia , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/genética , Antineoplásicos/farmacologia , Toxinas Bacterianas/genética , Linhagem Celular Tumoral , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/farmacologia , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Nus , Engenharia de Proteínas , Serina Endopeptidases/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Biol Chem ; 290(6): 3529-41, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25519908

RESUMO

Protease-activated receptors (PARs) are a family of seven-transmembrane, G-protein-coupled receptors that are activated by multiple serine proteases through specific N-terminal proteolytic cleavage and the unmasking of a tethered ligand. The majority of PAR-activating proteases described to date are soluble proteases that are active during injury, coagulation, and inflammation. Less investigation, however, has focused on the potential for membrane-anchored serine proteases to regulate PAR activation. Testisin is a unique trypsin-like serine protease that is tethered to the extracellular membrane of cells through a glycophosphatidylinositol (GPI) anchor. Here, we show that the N-terminal domain of PAR-2 is a substrate for testisin and that proteolytic cleavage of PAR-2 by recombinant testisin activates downstream signaling pathways, including intracellular Ca(2+) mobilization and ERK1/2 phosphorylation. When testisin and PAR-2 are co-expressed in HeLa cells, GPI-anchored testisin specifically releases the PAR-2 tethered ligand. Conversely, knockdown of endogenous testisin in NCI/ADR-Res ovarian tumor cells reduces PAR-2 N-terminal proteolytic cleavage. The cleavage of PAR-2 by testisin induces activation of the intracellular serum-response element and NFκB signaling pathways and the induction of IL-8 and IL-6 cytokine gene expression. Furthermore, the activation of PAR-2 by testisin results in the loss and internalization of PAR-2 from the cell surface. This study reveals a new biological substrate for testisin and is the first demonstration of the activation of a PAR by a serine protease GPI-linked to the cell surface.


Assuntos
Proteólise , Receptor PAR-2/metabolismo , Serina Endopeptidases/metabolismo , Sinalização do Cálcio , Membrana Celular/metabolismo , Proteínas Ligadas por GPI/metabolismo , Células HEK293 , Células HeLa , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Receptor PAR-2/química , Elementos de Resposta
12.
Physiol Rep ; 1(4)2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24179676

RESUMO

The lack of bile flow from the liver into the intestine can have devastating complications including hepatic failure, sepsis and even death. This pathologic condition known as cholestasis can result from etiologies as diverse as total parenteral nutrition (TPN), hepatitis and pancreatic cancer. The intestinal injury associated with cholestasis has been shown to result in decreased intestinal resistance, increased bacterial translocation and increased endotoxemia. Anecdotal clinical evidence suggests a genetic predisposition to exaggerated injury. Recent animal research on two different strains of inbred mice demonstrating different rates of bacterial translocation with different mortality rates supports this premise. In this study, a microarray analysis of intestinal tissue following common bile duct ligation (CBDL) performed under general anesthesia on these same two strains of inbred mice was done with the goal of identifying the potential molecular mechanistic pathways responsible. Over 500 genes were increased more than 2.0 fold following CBDL. The most promising candidate genes included MUPs, Serpina1a and LCN-2. RT-PCR validated the microarray results for these candidate genes. In an in vitro experiment using differentiated intestinal epithelial cells, inhibition of MUP-1 by siRNA resulted in increased intestinal epithelial cell permeability. Diverse novel mechanisms involving the growth hormone pathway, the acute phase response and the innate immune response are thus potential avenues for limiting cholestatic intestinal injury. Changes in gene expression were at times found to be not only due to the CBDL but also due to the murine strain. Should further studies in cholestatic patients demonstrate inter-individual variability similar to what we have shown in mice, then a "personalized medicine" approach to cholestatic patients may become possible.

13.
PLoS One ; 8(3): e57855, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23472114

RESUMO

SerpinB2 or plasminogen activator inhibitor type 2 (PAI-2) is highly induced in macrophages in response to inflammatory stimuli and is linked to the modulation of innate immunity, macrophage survival, and inhibition of plasminogen activators. Lipopolysaccharide (LPS), a potent bacterial endotoxin, can induce SerpinB2 expression via the toll-like receptor 4 (TLR4) by ∼1000-fold over a period of 24 hrs in murine macrophages. To map the LPS-regulated SerpinB2 promoter regions, we transfected reporter constructs driven by the ∼5 kb 5'-flanking region of the murine SerpinB2 gene and several deletion mutants into murine macrophages. In addition, we compared the DNA sequence of the murine 5' flanking sequence with the sequence of the human gene for homologous functional regulatory elements and identified several regulatory cis-acting elements in the human SERPINB2 promoter conserved in the mouse. Mutation analyses revealed that a CCAAT enhancer binding (C/EBP) element, a cyclic AMP response element (CRE) and two activator protein 1 (AP-1) response elements in the murine SerpinB2 proximal promoter are essential for optimal LPS-inducibility. Electrophoretic mobility shift (EMSA) and chromatin immunoprecipitation (ChIP) assays demonstrated that LPS induces the formation of C/EBP-ß containing complexes with the SerpinB2 promoter. Importantly, both constitutive and LPS-induced SerpinB2 expression was severely abrogated in C/EBP-ß-null mouse embryonic fibroblasts (MEFs) and primary C/EBP-ß-deficient peritoneal macrophages. Together, these data provide new insight into C/EBP-ß-dependent regulation of inflammation-associated SerpinB2 expression.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Regulação da Expressão Gênica , Lipopolissacarídeos/metabolismo , Inibidor 2 de Ativador de Plasminogênio/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Sequência Conservada , Análise Mutacional de DNA , Fibroblastos/citologia , Humanos , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas
14.
J Biol Chem ; 288(15): 10328-37, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23443662

RESUMO

The type II transmembrane serine protease matriptase is a key regulator of epithelial barriers in skin and intestine. In skin, matriptase acts upstream of the glycosylphosphatidylinositol-anchored serine protease, prostasin, to activate the prostasin zymogen and initiate a proteolytic cascade that is required for stratum corneum barrier functionality. Here, we have investigated the relationship between prostasin and matriptase in intestinal epithelial barrier function. We find that similar to skin, matriptase and prostasin are components of a common intestinal epithelial barrier-forming pathway. Depletion of prostasin by siRNA silencing in Caco-2 intestinal epithelium inhibits barrier development similar to loss of matriptase, and the addition of recombinant prostasin to the basal side of polarized Caco-2 epithelium stimulates barrier forming changes similar to the addition of recombinant matriptase. However, in contrast to the proteolytic cascade in skin, prostasin functions upstream of matriptase to activate the endogenous matriptase zymogen. Prostasin is unable to proteolytically activate the matriptase zymogen directly but induces matriptase activation indirectly. Prostasin requires expression of endogenous matriptase to stimulate barrier formation since matriptase depletion by siRNA silencing abrogates prostasin barrier-forming activity. Active recombinant matriptase, however, does not require the expression of endogenous prostasin for barrier-forming activity. Together, these data show that matriptase and not prostasin is the primary effector protease of tight junction assembly in simple columnar epithelia and further highlight a spatial and tissue-specific aspect of cell surface proteolytic cascades.


Assuntos
Precursores Enzimáticos/biossíntese , Células Epiteliais/enzimologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Mucosa Intestinal/enzimologia , Serina Endopeptidases/biossíntese , Serina Endopeptidases/metabolismo , Células CACO-2 , Ativação Enzimática/fisiologia , Precursores Enzimáticos/genética , Células Epiteliais/citologia , Inativação Gênica , Humanos , Mucosa Intestinal/citologia , Proteólise , Serina Endopeptidases/genética , Junções Íntimas/enzimologia , Junções Íntimas/genética
15.
Inflamm Bowel Dis ; 18(7): 1303-14, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22081509

RESUMO

BACKGROUND: Matriptase is a membrane-anchored serine protease encoded by suppression of tumorigenicity-14 (ST14) that is required for epithelial barrier homeostasis. However, its functional role in inflammatory bowel disease (IBD) is unexplored. METHODS: Matriptase expression in control, Crohn's disease, and ulcerative colitis tissue specimens was studied by quantitative polymerase chain reaction (qPCR) and immunostaining. Matriptase function was investigated by subjecting St14 hypomorphic and control littermates to dextran sodium sulfate (DSS)-induced colitis and by siRNA silencing in cultured monolayers. Mice were analyzed for clinical, histological, molecular, and cellular effects. RESULTS: Matriptase protein and ST14 mRNA levels are significantly downregulated in inflamed colonic tissues from Crohn's disease and ulcerative colitis patients. Matriptase-deficient St14 hypomorphic mice administered DSS for 7 days followed by water without DSS for 3 days develop a severe colitis, with only 30% of the St14 hypomorphic mice surviving to day 14, compared with 100% of control littermates. Persistent colitis in surviving St14 hypomorphic mice was associated with sustained cytokine production, an inability to recover barrier integrity, and enhanced claudin-2 expression. Cytokines implicated in barrier disruption during IBD suppress matriptase expression in T84 epithelial monolayers and restoration of matriptase improves barrier integrity in the cytokine-perturbed monolayers. CONCLUSIONS: These data demonstrate a critical role for matriptase in restoring barrier function to injured intestinal mucosa during colitis, which is suppressed by excessive activation of the immune system. Strategies to enhance matriptase-mediated barrier recovery could be important for intervening in the cycle of inflammation associated with IBD.


Assuntos
Colite Ulcerativa/metabolismo , Colite/prevenção & controle , Doença de Crohn/metabolismo , Intestinos/enzimologia , Serina Endopeptidases/metabolismo , Serina Endopeptidases/fisiologia , Animais , Western Blotting , Colite/induzido quimicamente , Colite/patologia , Colite Ulcerativa/genética , Colite Ulcerativa/patologia , Colo/enzimologia , Colo/patologia , Doença de Crohn/genética , Doença de Crohn/patologia , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Impedância Elétrica , Feminino , Humanos , Técnicas Imunoenzimáticas , Intestinos/lesões , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina Endopeptidases/genética
16.
Cancer Res ; 68(14): 5648-57, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18632617

RESUMO

The tumor suppressor retinoblastoma protein (Rb) plays a pivotal role in the regulation of cell proliferation and sensitivity to apoptosis through binding to E2F transcription factors. Loss of Rb in response to genotoxic stress or inflammatory cytokines can enhance cell death, in part, by eliminating Rb-mediated repression of proapoptotic gene transcription. Here we show that calpain cleavage of Rb facilitates Rb loss by proteasome degradation and that this may occur during tumor necrosis factor alpha-induced apoptosis. The cytoprotective, Rb-binding protein SerpinB2 (plasminogen activator inhibitor type 2) protects Rb from calpain cleavage, increasing Rb levels and enhancing cell survival. Chromatin immunoprecipitation assays show that the increased Rb levels selectively enhance Rb repression of proapoptotic gene transcription. This cytoprotective role of SerpinB2 is illustrated by reduced susceptibility of SerpinB2-deficient mice to multistage skin carcinogenesis, where Rb-dependent cell proliferation competes with apoptosis during initiation of papilloma development. These data identify SerpinB2 as a cell survival factor that modulates Rb repression of proapoptotic signal transduction and define a new posttranslational mechanism for selective regulation of the intracellular levels of Rb.


Assuntos
Calpaína/metabolismo , Sobrevivência Celular , Regulação Neoplásica da Expressão Gênica , Inibidor 2 de Ativador de Plasminogênio/metabolismo , Inibidor 2 de Ativador de Plasminogênio/fisiologia , Proteína do Retinoblastoma/metabolismo , Animais , Apoptose , Fibroblastos/metabolismo , Predisposição Genética para Doença , Células HeLa , Humanos , Células Jurkat , Camundongos , Inibidor 2 de Ativador de Plasminogênio/genética , Transdução de Sinais , Neoplasias Cutâneas/metabolismo
17.
J Biol Chem ; 280(25): 23549-58, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15843372

RESUMO

Human granzyme B (GrB) released from cytotoxic lymphocytes plays a key role in the induction of target cell apoptosis when internalized in the presence of perforin. Here we demonstrate that GrB also possesses a potent extracellular matrix remodeling activity. Both native and recombinant GrB caused detachment of immortalized and transformed cell lines, primary endothelial cells, and chondrocytes. Cell detachment by GrB induced endothelial cell death (anoikis). GrB also inhibited tumor cell spreading, migration, and invasion in vitro. Investigation into the underlying mechanism revealed that GrB efficiently cleaves three proteins involved in extracellular matrix structure and function: vitronectin, fibronectin, and laminin. In vitronectin, GrB cleaves after an Arg-Lys-Asp (RGD) motif, which is part of the integrin-binding site found in matrix proteins. We propose that targeting of the integrin-extracellular matrix interface by GrB may allow perforin-independent killing of target cells via anoikis, restrict motility of tumor cells, facilitate lymphocyte migration, or directly reduce virus infectivity. It may also contribute to tissue destruction in diseases in which extracellular GrB is evident, such as rheumatoid arthritis and atherosclerosis.


Assuntos
Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Laminina/metabolismo , Serina Endopeptidases/metabolismo , Vitronectina/metabolismo , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Adesão Celular , Linhagem Celular Tumoral , Granzimas , Humanos , Hidrólise , Proteínas Recombinantes/metabolismo
18.
J Immunol ; 170(2): 805-15, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12517944

RESUMO

Granzyme B (grB) is a serine proteinase released by cytotoxic lymphocytes (CLs) to kill abnormal cells. GrB-mediated apoptotic pathways are conserved in nucleated cells; hence, CLs require mechanisms to protect against ectopic or misdirected grB. The nucleocytoplasmic serpin, proteinase inhibitor 9 (PI-9), is a potent inhibitor of grB that protects cells from grB-mediated apoptosis in model systems. Here we show that PI-9 is present in CD4(+) cells, CD8(+) T cells, NK cells, and at lower levels in B cells and myeloid cells. PI-9 is up-regulated in response to grB production and degranulation, and associates with grB-containing granules in activated CTLs and NK cells. Intracellular complexes of PI-9 and grB are evident in NK cells, and overexpression of PI-9 enhances CTL potency, suggesting that cytoplasmic grB, which may threaten CL viability, is rapidly inactivated by PI-9. Because dendritic cells (DCs) acquire characteristics similar to those of target cells to activate naive CD8(+) T cells and therefore may also require protection against grB, we investigated the expression of PI-9 in DCs. PI-9 is evident in thymic DCs (CD3(-), CD4(+), CD8(-), CD45(+)), tonsillar DCs, and DC subsets purified from peripheral blood (CD16(+) monocytes and CD123(+) plasmacytoid DCs). Furthermore, PI-9 is expressed in monocyte-derived DCs and is up-regulated upon TNF-alpha-induced maturation of monocyte-derived DCs. In conclusion, the presence and subcellular localization of PI-9 in leukocytes and DCs are consistent with a protective role against ectopic or misdirected grB during an immune response.


Assuntos
Células Apresentadoras de Antígenos/citologia , Degranulação Celular/imunologia , Citotoxicidade Imunológica , Serina Endopeptidases/metabolismo , Inibidores de Serina Proteinase/biossíntese , Serpinas/biossíntese , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Regulação para Cima/imunologia , Adjuvantes Imunológicos/biossíntese , Adjuvantes Imunológicos/sangue , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/fisiologia , Células Apresentadoras de Antígenos/metabolismo , Diferenciação Celular/imunologia , Linhagem Celular , Células Cultivadas , Citotoxicidade Imunológica/genética , Células Dendríticas/classificação , Células Dendríticas/metabolismo , Granzimas , Humanos , Líquido Intracelular/enzimologia , Líquido Intracelular/metabolismo , Leucócitos/metabolismo , Serina Endopeptidases/biossíntese , Inibidores de Serina Proteinase/sangue , Inibidores de Serina Proteinase/genética , Inibidores de Serina Proteinase/fisiologia , Serpinas/sangue , Serpinas/genética , Serpinas/fisiologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Citotóxicos/enzimologia , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA