Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(28): e2201423119, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35867758

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos , Antineoplásicos , Toxinas Bacterianas , Neoplasias Ováricas , Profármacos , Serina Proteasas , Antígenos Bacterianos/farmacología , Antígenos Bacterianos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Toxinas Bacterianas/farmacología , Toxinas Bacterianas/uso terapéutico , Línea Celular Tumoral , Precursores Enzimáticos/metabolismo , Femenino , Humanos , Recurrencia Local de Neoplasia , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Profármacos/farmacología , Profármacos/uso terapéutico , Serina Proteasas/metabolismo , Esferoides Celulares , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Mol Med (Berl) ; 97(5): 691-709, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30911775

RESUMEN

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.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/metabolismo , Neoplasias Ováricas/metabolismo , Ribonucleasa Pancreática/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Línea Celular Tumoral , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Ratones Desnudos , Metástasis de la Neoplasia/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Ováricas/patología , Proteolisis , Proteínas de Transporte Vesicular/metabolismo
4.
J Biol Chem ; 292(26): 10801-10812, 2017 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-28490634

RESUMEN

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.


Asunto(s)
Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/metabolismo , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/genética , Colitis Ulcerosa/patología , Colon/metabolismo , Colon/patología , Enfermedad de Crohn/inducido químicamente , Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Humanos , Ácidos Hidroxámicos/farmacología , Interleucina-13/genética , Interleucina-4/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Serina Endopeptidasas/genética , Vorinostat
5.
Curr Opin Hematol ; 23(3): 243-52, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26906027

RESUMEN

PURPOSE OF REVIEW: The endothelial cell plasma membrane is a metabolically active, dynamic, and fluid microenvironment where pericellular proteolysis plays a critical role. Membrane-anchored proteases may be expressed by endothelial cells as well as mural cells and leukocytes with distribution both inside and outside of the vascular system. Here, we will review the recent advances in our understanding of the direct and indirect roles of membrane-anchored proteases in vascular biology and the possible conservation of their extravascular functions in endothelial cell biology. RECENT FINDINGS: Membrane-anchored proteases belonging to the serine or metalloprotease families contain amino-terminal or carboxy-terminal domains, which serve to tether their extracellular protease domains directly at the plasma membrane. This architecture enables protease function and substrate repertoire to be regulated through dynamic localization in distinct areas of the cell membrane. These proteases are proving to be key components of the cell machinery for regulating vascular permeability, generation of vasoactive peptides, receptor tyrosine kinase transactivation, extracellular matrix proteolysis, and angiogenesis. SUMMARY: A complex picture of the interdependence between membrane-anchored protease localization and function is emerging that may provide a mechanism for precise coordination of extracellular signals and intracellular responses through communication with the cytoskeleton and with cellular signaling molecules.


Asunto(s)
Membrana Celular/enzimología , Células Endoteliales/enzimología , Células Endoteliales/fisiología , Serina Endopeptidasas/metabolismo , Células Endoteliales/citología , Humanos
6.
J Biol Chem ; 290(6): 3529-41, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25519908

RESUMEN

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.


Asunto(s)
Proteolisis , Receptor PAR-2/metabolismo , Serina Endopeptidasas/metabolismo , Señalización del Calcio , Membrana Celular/metabolismo , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Células HeLa , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Receptor PAR-2/química , Elementos de Respuesta
7.
Glia ; 60(12): 1944-53, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22927334

RESUMEN

The molecular mechanisms controlling human oligodendrocyte development are poorly characterized. Microarray analysis of human oligodendrocyte progenitor cells (OPCs) and immature oligodendrocytes revealed that specific-class I histone deacetylase (HDAC) target genes were actively repressed during oligodendrocyte commitment. Although epigenetic regulation of oligodendrocyte differentiation has been established in rodent development, the role of HDACs in human OPCs remains undefined. We used HDAC inhibitors (HDACi) trichostatin A (TSA) and sodium butyrate to determine the importance of HDAC activity in human primary OPC differentiation. Treatment with either drug resulted in significant dose-dependent inhibition of O4(+) oligodendrocyte cell differentiation, reduction of oligodendrocyte morphological maturation, and downregulation of myelin basic protein mRNA. High dose TSA treatment was also associated with reduction in OPC proliferation. HDACi treatment prevented downregulation of SOX2, ID4, and TCF7L2 mRNAs but did not regulate HES5, suggesting that targets of HDAC repression may differ between species. These results predict that HDACi treatment would impair proliferation and differentiation by parenchymal oligodendrocyte progenitors, and thereby degrade their potential for endogenous repair in human demyelinating disease. © 2012 Wiley Periodicals, Inc.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/enzimología , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Oligodendroglía/enzimología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Feto/citología , Feto/efectos de los fármacos , Feto/enzimología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/fisiología , Humanos , Oligodendroglía/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Células Madre/efectos de los fármacos , Células Madre/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...