Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39257753

RESUMEN

TMPRSS13, a member of the Type II Transmembrane Serine Proteases (TTSP) family, is involved in cancer progression and in cell entry of respiratory viruses. To date, no inhibitors have been specifically developed toward this protease. In this study, a chemical library of 65 ketobenzothiazole-based peptidomimetic molecules was screened against a proteolytically active form of recombinant TMPRSS13 to identify novel inhibitors. Following an initial round of screening, subsequent synthesis of additional derivatives supported by molecular modelling, uncovered important molecular determinants involved in TMPRSS13 inhibition. One inhibitor, N-0430, achieved low nanomolar affinity towards TMPRSS13 activity in a cellular context. Using a SARS-CoV-2 pseudovirus cell entry model, we further show the ability of N-0430 to block TMPRSS13-dependent entry of the pseudovirus. The identified peptidomimetic inhibitors and the molecular insights of their potency gained from this study will aid in the development of specific TMPRSS13 inhibitors.

2.
Biol Chem ; 403(10): 969-982, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-35796294

RESUMEN

TMPRSS13 is a member of the type II transmembrane serine protease (TTSP) family. Here we characterize a novel post-translational mechanism important for TMPRSS13 function: proteolytic cleavage within the extracellular TMPRSS13 stem region located between the transmembrane domain and the first site of N-linked glycosylation at asparagine (N)-250 in the scavenger receptor cysteine rich (SRCR) domain. Importantly, the catalytic competence of TMPRSS13 is essential for stem region cleavage, suggesting an autonomous mechanism of action. Site-directed mutagenesis of the 10 basic amino acids (four arginine and six lysine residues) in this region abrogated zymogen activation and catalytic activity of TMPRSS13, as well as phosphorylation, cell surface expression, and shedding. Mutation analysis of individual arginine residues identified R223, a residue located between the low-density lipoprotein receptor class A domain and the SRCR domain, as important for stem region cleavage. Mutation of R223 causes a reduction in the aforementioned functional processing steps of TMPRSS13. These data provide further insight into the roles of different post-translational modifications as regulators of the function and localization of TMPRSS13. Additionally, the data suggest the presence of complex interconnected regulatory mechanisms that may serve to ensure the proper levels of cell-surface and pericellular TMPRSS13-mediated proteolysis under homeostatic conditions.


Asunto(s)
Proteínas de la Membrana , Procesamiento Proteico-Postraduccional , Arginina/metabolismo , Precursores Enzimáticos/metabolismo , Proteínas de la Membrana/metabolismo , Proteolisis
3.
J Biol Chem ; 297(4): 101227, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34562451

RESUMEN

TMPRSS13, a member of the type II transmembrane serine protease (TTSP) family, harbors four N-linked glycosylation sites in its extracellular domain. Two of the glycosylated residues are located in the scavenger receptor cysteine-rich (SRCR) protein domain, while the remaining two sites are in the catalytic serine protease (SP) domain. In this study, we examined the role of N-linked glycosylation in the proteolytic activity, autoactivation, and cellular localization of TMPRSS13. Individual and combinatory site-directed mutagenesis of the glycosylated asparagine residues indicated that glycosylation of the SP domain is critical for TMPRSS13 autoactivation and catalytic activity toward one of its protein substrates, the prostasin zymogen. Additionally, SP domain glycosylation-deficient TMPRSS13 displayed impaired trafficking of TMPRSS13 to the cell surface, which correlated with increased retention in the endoplasmic reticulum. Importantly, we showed that N-linked glycosylation was a critical determinant for subsequent phosphorylation of endogenous TMPRSS13. Taken together, we conclude that glycosylation plays an important role in regulating TMPRSS13 activation and activity, phosphorylation, and cell surface localization.


Asunto(s)
Membrana Celular/enzimología , Precursores Enzimáticos/metabolismo , Proteínas de la Membrana/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , Serina Endopeptidasas/metabolismo , Animales , Células COS , Membrana Celular/genética , Chlorocebus aethiops , Precursores Enzimáticos/genética , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Dominios Proteicos , Transporte de Proteínas/genética , Serina Endopeptidasas/genética
4.
Oncogene ; 39(41): 6421-6436, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32868877

RESUMEN

Breast cancer progression is accompanied by increased expression of extracellular and cell-surface proteases capable of degrading the extracellular matrix as well as cleaving and activating downstream targets. The type II transmembrane serine proteases (TTSPs) are a family of cell-surface proteases that play critical roles in numerous types of cancers. Therefore, the aim of this study was to identify novel and uncharacterized TTSPs with differential expression in breast cancer and to determine their potential roles in progression. Systematic in silico data analysis followed by immunohistochemical validation identified increased expression of the TTSP family member, TMPRSS13 (transmembrane protease, serine 13), in invasive ductal carcinoma patient tissue samples compared to normal breast tissue. To test whether loss of TMPRSS13 impacts tumor progression, TMPRSS13 was genetically ablated in the oncogene-induced transgenic MMTV-PymT tumor model. TMPRSS13 deficiency resulted in a significant decrease in overall tumor burden and growth rate, as well as a delayed formation of detectable mammary tumors, thus suggesting a causal relationship between TMPRSS13 expression and the progression of breast cancer. Complementary studies using human breast cancer cell culture models revealed that siRNA-mediated silencing of TMPRSS13 expression decreases proliferation, induces apoptosis, and attenuates invasion. Importantly, targeting TMPRSS13 expression renders aggressive triple-negative breast cancer cell lines highly responsive to chemotherapy. At the molecular level, knockdown of TMPRSS13 in breast cancer cells led to increased protein levels of the tumor-suppressive protease prostasin. TMPRSS13/prostasin co-immunoprecipitation and prostasin zymogen activation experiments identified prostasin as a potential novel target for TMPRSS13. Regulation of prostasin levels may be a mechanism that contributes to the pro-oncogenic properties of TMPRSS13 in breast cancer. TMPRSS13 represents a novel candidate for targeted therapy in combination with standard of care chemotherapy agents in patients with hormone receptor-negative breast cancer or in patients with tumors refractory to endocrine therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Ductal de Mama/patología , Neoplasias Mamarias Experimentales/patología , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Mama/patología , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Serina Endopeptidasas/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética
5.
Sci Rep ; 10(1): 13896, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807808

RESUMEN

Cancer progression is often accompanied by increased levels of extracellular proteases capable of remodeling the extracellular matrix and promoting pro-cancerous signaling pathways by activating growth factors and receptors. The type II transmembrane serine protease (TTSP) family encompasses several proteases that play critical roles in cancer progression; however, the expression or function of the TTSP TMPRSS13 in carcinogenesis has not been examined. In the present study, we found TMPRSS13 to be differentially expressed at both the transcript and protein levels in human colorectal cancer (CRC). Immunohistochemical analyses revealed consistent high expression of TMPRSS13 protein on the cancer cell surface in CRC patient samples; in contrast, the majority of normal colon samples displayed no detectable expression. On a functional level, TMPRSS13 silencing in CRC cell lines increased apoptosis and impaired invasive potential. Importantly, transgenic overexpression of TMPRSS13 in CRC cell lines increased tolerance to apoptosis-inducing agents, including paclitaxel and HA14-1. Conversely, TMPRSS13 silencing rendered CRC cells more sensitive to these agents. Together, our findings suggest that TMPRSS13 plays an important role in CRC cell survival and in promoting resistance to drug-induced apoptosis; we also identify TMPRSS13 as a potential new target for monotherapy or combination therapy with established chemotherapeutics to improve treatment outcomes in CRC patients.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Proteínas de la Membrana/genética , Invasividad Neoplásica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Serina Endopeptidasas/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
6.
Cancer Metastasis Rev ; 38(3): 357-387, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31529338

RESUMEN

Over the last two decades, a novel subgroup of serine proteases, the cell surface-anchored serine proteases, has emerged as an important component of the human degradome, and several members have garnered significant attention for their roles in cancer progression and metastasis. A large body of literature describes that cell surface-anchored serine proteases are deregulated in cancer and that they contribute to both tumor formation and metastasis through diverse molecular mechanisms. The loss of precise regulation of cell surface-anchored serine protease expression and/or catalytic activity may be contributing to the etiology of several cancer types. There is therefore a strong impetus to understand the events that lead to deregulation at the gene and protein levels, how these precipitate in various stages of tumorigenesis, and whether targeting of selected proteases can lead to novel cancer intervention strategies. This review summarizes current knowledge about cell surface-anchored serine proteases and their role in cancer based on biochemical characterization, cell culture-based studies, expression studies, and in vivo experiments. Efforts to develop inhibitors to target cell surface-anchored serine proteases in cancer therapy will also be summarized.


Asunto(s)
Glicosilfosfatidilinositoles/metabolismo , Neoplasias/enzimología , Neoplasias/patología , Serina Proteasas/metabolismo , Animales , Membrana Celular/enzimología , Progresión de la Enfermedad , Humanos , Metástasis de la Neoplasia
7.
J Biol Chem ; 292(36): 14867-14884, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28710277

RESUMEN

TMPRSS13 is a member of the type II transmembrane serine protease (TTSP) family. Although various TTSPs have been characterized in detail biochemically and functionally, the basic properties of TMPRSS13 remain unclear. Here, we investigate the activation, inhibition, post-translational modification, and localization of TMPRSS13. We show that TMPRSS13 is a glycosylated, active protease and that its own proteolytic activity mediates zymogen cleavage. Full-length, active TMPRSS13 exhibits impaired cell-surface expression in the absence of the cognate Kunitz-type serine protease inhibitors, hepatocyte growth factor activator inhibitor (HAI)-1 or HAI-2. Concomitant presence of TMPRSS13 with either HAI-1 or -2 mediates phosphorylation of residues in the intracellular domain of the protease, and it coincides with efficient transport of the protease to the cell surface and its subsequent shedding. Cell-surface labeling experiments indicate that the dominant form of TMPRSS13 on the cell surface is phosphorylated, whereas intracellular TMPRSS13 is predominantly non-phosphorylated. These data provide novel insight into the cellular properties of TMPRSS13 and highlight phosphorylation of TMPRSS13 as a novel post-translational modification of this TTSP family member and potentially other members of this family of proteases.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas Inhibidoras de Proteinasas Secretoras/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Fosforilación , Serina Endopeptidasas/genética
8.
FEBS J ; 284(10): 1421-1436, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27870503

RESUMEN

Pericellular proteases have long been implicated in carcinogenesis. Previous research focused on these proteins, primarily as extracellular matrix (ECM) protein-degrading enzymes which allowed cancer cells to breach the basement membrane and invade surrounding tissue. However, recently, there has been a shift in the view of cell surface proteases, including serine proteases, as proteolytic modifiers of particular targets, including growth factors and protease-activated receptors, which are critical for the activation of oncogenic signaling pathways. Of the 176 human serine proteases currently identified, a subset of 17, known as type II transmembrane serine proteases (TTSPs). Many have been shown to be relevant to cancer progression since they were first identified as a family around the turn of the century. To this end, altered expression of TTSPs appeared as a trademark of several tumor types. However, the substrates and underlying signaling pathways remained unclear. Localization of these proteins to the cell surface places them in the unique position to mediate signal transduction between the cell and its surrounding environment. Many of the TTSPs have already been shown to play key roles in processes such as postnatal development, tissue homeostasis, and tumor progression, which share overlapping molecular mechanisms. In this review, we summarize the current knowledge regarding the role of the TTSP family in pro-oncogenic signaling.


Asunto(s)
Neoplasias/metabolismo , Serina Proteasas/metabolismo , Animales , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neoplasias/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Serina Proteasas/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
9.
Oncotarget ; 7(36): 58162-58173, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27528224

RESUMEN

The poor prognosis for patients with inflammatory breast cancer (IBC) compared to patients with other types of breast cancers emphasizes the need to better understand the molecular underpinnings of this disease with the goal of developing effective targeted therapeutics. Dysregulation of matriptase expression, an epithelial-specific member of the type II transmembrane serine protease family, has been demonstrated in many different cancer types. To date, no studies have assessed the expression and potential pro-oncogenic role of matriptase in IBC. We examined the functional relationship between matriptase and the HGF/c-MET signaling pathway in the IBC cell lines SUM149 and SUM190, and in IBC patient samples. Matriptase and c-Met proteins are localized on the surface membrane of IBC cells and their expression is strongly correlated in infiltrating cancer cells and in the cancer cells of lymphatic emboli in patient samples. Abrogation of matriptase expression by silencing with RNAi or inhibition of matriptase proteolytic activity with a synthetic inhibitor impairs the conversion of inactive pro-HGF to active HGF and subsequent c-Met-mediated signaling, leading to efficient impairment of proliferation and invasion of IBC cells. These data show the potential of matriptase inhibitors as a novel targeted therapy for IBC, and lay the groundwork for the development and testing of such drugs.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias Inflamatorias de la Mama/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Serina Endopeptidasas/metabolismo , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Femenino , Humanos , Inmunohistoquímica , Invasividad Neoplásica , Precursores de Proteínas/metabolismo , Proteolisis/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Serina Endopeptidasas/genética , Transducción de Señal
10.
Biol Chem ; 397(9): 815-26, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27078673

RESUMEN

Carcinogenesis is accompanied by increased protein and activity levels of extracellular cell-surface proteases that are capable of modifying the tumor microenvironment by directly cleaving the extracellular matrix, as well as activating growth factors and proinflammatory mediators involved in proliferation and invasion of cancer cells, and recruitment of inflammatory cells. These complex processes ultimately potentiate neoplastic progression leading to local tumor cell invasion, entry into the vasculature, and metastasis to distal sites. Several members of the type II transmembrane serine protease (TTSP) family have been shown to play critical roles in cancer progression. In this review the knowledge collected over the past two decades about the molecular mechanisms underlying the pro-cancerous properties of selected TTSPs will be summarized. Furthermore, we will discuss how these insights may facilitate the translation into clinical settings in the future by specifically targeting TTSPs as part of novel cancer treatment regimens.


Asunto(s)
Membrana Celular/enzimología , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Serina Proteasas/metabolismo , Animales , Diagnóstico por Imagen , Humanos , Neoplasias/diagnóstico por imagen , Neoplasias/patología
11.
J Cell Physiol ; 231(7): 1476-83, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26297835

RESUMEN

Cancer progression is accompanied by increased levels of extracellular proteases that are capable of remodeling the extracellular matrix, as well as cleaving and activating growth factors and receptors that are involved in pro-cancerous signaling pathways. Several members of the type II transmembrane serine protease (TTSP) family have been shown to play critical roles in cancer progression, however, the expression or function of the TTSP Human Airway Trypsin-like protease (HAT) in carcinogenesis has not been examined. In the present study we aimed to determine the expression of HAT during squamous cell carcinogenesis. HAT transcript is present in several tissues containing stratified squamous epithelium and decreased expression is observed in carcinomas. We determined that HAT protein is consistently expressed on the cell surface in suprabasal/apical layers of squamous cells in healthy cervical and esophageal epithelia. To assess whether HAT protein is differentially expressed in normal tissue versus tissue in different stages of carcinogenesis, we performed a comprehensive immunohistochemical analysis of HAT protein expression levels and localization in arrays of paraffin embedded human cervical and esophageal carcinomas compared to the corresponding normal tissue. We found that HAT protein is expressed in the non-proliferating, differentiated cellular strata and is lost during the dedifferentiation of epithelial cells, a hallmark of squamous cell carcinogenesis. Thus, HAT expression may potentially be useful as a marker for clinical grading and assessment of patient prognosis in squamous cell carcinomas.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinogénesis/genética , Carcinoma de Células Escamosas/genética , Serina Endopeptidasas/genética , Biomarcadores de Tumor/biosíntesis , Carcinoma de Células Escamosas/patología , Membrana Celular/genética , Membrana Celular/metabolismo , Epitelio/metabolismo , Epitelio/patología , Esófago/metabolismo , Esófago/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Serina Endopeptidasas/biosíntesis
12.
Nat Commun ; 6: 6776, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25873032

RESUMEN

Matriptase is an epithelia-specific membrane-anchored serine protease that has received considerable attention in recent years because of its consistent dysregulation in human epithelial tumours, including breast cancer. Mice with reduced levels of matriptase display a significant delay in oncogene-induced mammary tumour formation and blunted tumour growth. The abated tumour growth is associated with a decrease in cancer cell proliferation. Here we demonstrate by genetic deletion and silencing that the proliferation impairment in matriptase-deficient breast cancer cells is caused by their inability to initiate activation of the c-Met signalling pathway in response to fibroblast-secreted pro-HGF. Similarly, inhibition of matriptase catalytic activity using a selective small-molecule inhibitor abrogates the activation of c-Met, Gab1 and AKT, in response to pro-HGF, which functionally leads to attenuated proliferation in breast carcinoma cells. We conclude that matriptase is critically involved in breast cancer progression and represents a potential therapeutic target in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma/genética , Proliferación Celular/genética , Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias Mamarias Experimentales/genética , Proteínas de la Membrana/genética , Precursores de Proteínas/metabolismo , Serina Endopeptidasas/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Fosfoproteínas , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-met , Transducción de Señal
13.
PLoS One ; 9(2): e87675, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24498351

RESUMEN

Over the last two decades, cell surface proteases belonging to the type II transmembrane serine protease (TTSP) family have emerged as important enzymes in the mammalian degradome, playing critical roles in epithelial biology, regulation of metabolic homeostasis, and cancer. Human airway trypsin-like protease 5 (HATL5) is one of the few family members that remains uncharacterized. Here we demonstrate that HATL5 is a catalytically active serine protease that is inhibited by the two Kunitz type serine protease inhibitors, hepatocyte growth factor activator inhibitor (HAI)-1 and 2, as well as by serpinA1. Full-length HATL5 is localized on the cell surface of cultured mammalian cells as demonstrated by confocal microscopy. HATL5 displays a relatively restricted tissue expression profile, with both transcript and protein present in the cervix, esophagus, and oral cavity. Immunohistochemical analysis revealed an expression pattern where HATL5 is localized on the cell surface of differentiated epithelial cells in the stratified squamous epithelia of all three of these tissues. Interestingly, HATL5 is significantly decreased in cervical, esophageal, and head and neck carcinomas as compared to normal tissue. Analysis of cervical and esophageal cancer tissue arrays demonstrated that the squamous epithelial cells lose their expression of HATL5 protein upon malignant transformation.


Asunto(s)
Membrana Celular/metabolismo , Epitelio/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias/enzimología , Serina Endopeptidasas/metabolismo , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Secuencia de Aminoácidos , Animales , Células COS , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , Epitelio/patología , Neoplasias Esofágicas/enzimología , Neoplasias Esofágicas/genética , Femenino , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Inmunohistoquímica , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/genética , Neoplasias/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina Endopeptidasas/genética , Neoplasias de la Lengua/enzimología , Neoplasias de la Lengua/genética , Neoplasias del Cuello Uterino/enzimología , Neoplasias del Cuello Uterino/genética
14.
Cell Tissue Res ; 351(2): 245-53, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22350849

RESUMEN

The type II transmembrane serine protease matriptase has an essential role in the integrity and function of multiple epithelial tissues. In the epidermis, matriptase activates the glycosylphosphatidylinositol (GPI) anchored membrane serine protease prostasin to initiate a proteolytic cascade that is required for the development of the stratum corneum barrier function. Accordingly, mice deficient for matriptase phenocopy mice deficient for epidermal prostasin and present with impaired corneocyte differentiation, imparied lipid matrix formation, loss of profilaggrin processing and loss of tight junction formation and function. Together, these defects lead to a compromised epidermal barrier and result in fatal dehydration during the neonatal period. Proteolytic activity of the matriptase-prostasin cascade is regulated in the epidermis via inhibition by the Kunitz-type serine protease inhibitor hepatocyte growth factor activator inhibitor-1 (HAI-1). Importantly, targeted post-natal ablation of matriptase in mice perturbs the function of multiple adult tissues, indicating an ongoing requirement for matriptase proteolysis in the maintenance of diverse types of epithelia. Impaired matriptase proteolytic activity has been linked to human Autosomal Recessive Icthyosis with Hypotrichosis (ARIH), whereas aberrant matriptase activity has been implicated in Netherton's Syndrome. This review will summarize information pertaining to the role of matriptase in epithelial biology and will discuss recent advancements in our understanding of how matriptase activity is regulated and the down-stream effectors of matriptase proteolysis.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Epitelio/patología , Serina Endopeptidasas/metabolismo , Animales , Diferenciación Celular/fisiología , Células Epiteliales/enzimología , Células Epiteliales/patología , Epitelio/enzimología , Humanos
15.
Breast Cancer Res ; 14(4): R104, 2012 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-22788954

RESUMEN

INTRODUCTION: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have shown clinical efficacy in lung, colon, and pancreatic cancers. In lung cancer, resistance to EGFR TKIs correlates with amplification of the hepatocyte growth factor (HGF) receptor tyrosine kinase Met. Breast cancers do not respond to EGFR TKIs, even though EGFR is overexpressed. This intrinsic resistance to EGFR TKIs in breast cancer does not correlate with Met amplification. In several tissue monoculture models of human breast cancer, Met, although expressed, is not phosphorylated, suggesting a requirement for a paracrine-produced ligand. In fact, HGF, the ligand for Met, is not expressed in epithelial cells but is secreted by fibroblasts in the tumor stroma. We have identified a number of breast cancer cell lines that are sensitive to EGFR TKIs. This sensitivity is in conflict with the observed clinical resistance to EGFR TKIs in breast cancers. Here we demonstrate that fibroblast secretion of HGF activates Met and leads to EGFR/Met crosstalk and resistance to EGFR TKIs in triple-negative breast cancer (TNBC). METHODS: The SUM102 and SUM149 TNBC cell lines were used in this study. Recombinant HGF as well as conditioned media from fibroblasts expressing HGF were used as sources for Met activation. Furthermore, we co-cultured HGF-secreting fibroblasts with Met-expressing cancer cells to mimic the paracrine HGF/Met pathway, which is active in the tumor microenvironment. Cell growth, survival, and transformation were measured by cell counting, clonogenic and MTS assays, and soft agar colony formation, respectively. Student's t test was used for all statistical analysis. RESULTS: Here we demonstrate that treatment of breast cancer cells sensitive to EGFR TKIs with recombinant HGF confers a resistance to EGFR TKIs. Interestingly, knocking down EGFR abrogated HGF-mediated cell survival, suggesting a crosstalk between EGFR and Met. HGF is secreted as a single-chain pro-form, which has to be proteolytically cleaved in order to activate Met. To determine whether the proteases required to activate pro-HGF were present in the breast cancer cells, we utilized a fibroblast cell line expressing pro-HGF (RMF-HGF). Addition of pro-HGF-secreting conditioned fibroblast media to TNBC cells as well as co-culturing of TNBC cells with RMF-HGF fibroblasts resulted in robust phosphorylation of Met and stimulated proliferation in the presence of an EGFR TKI. CONCLUSIONS: Taken together, these data suggest a role for Met in clinical resistance to EGFR TKIs in breast cancer through EGFR/Met crosstalk mediated by tumor-stromal interactions.


Asunto(s)
Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Fibroblastos/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Comunicación Paracrina , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Receptores ErbB/genética , Gefitinib , Expresión Génica , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-met/metabolismo , Quinazolinas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
16.
J Cell Physiol ; 227(4): 1604-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21678412

RESUMEN

Breast cancer tumorigenesis is accompanied by increased levels of extracellular proteases that are capable of remodeling the extracellular matrix as well as cleaving and activating growth factors and signaling receptors that are critically involved in neoplastic progression. Multiple studies implicate the membrane anchored serine protease matriptase (also known as MT-SP1 and epithin) in breast cancer. The pro-form of the GPI-anchored serine protease prostasin has recently been identified as a physiological substrate of matriptase and the two proteases are co-expressed in multiple healthy tissues. In this study, the inter-relationship between the two membrane-anchored serine proteases in breast cancer was investigated using breast cancer cell lines and breast cancer patient samples to delineate the association between matriptase and prostasin. We used Western blotting to determine the expression of matriptase and prostasin proteins in a panel of breast cancer cell lines and immunohistochemistry to assess the expression in serial sections from breast cancer tissue arrays. We demonstrate that the expression of matriptase and prostasin is closely correlated in breast cancer cell lines as well as in breast cancer tissue samples. Furthermore, matriptase and prostasin display a near identical spatial expression pattern in the epithelial compartment of breast cancer tissue. These data suggest that the matriptase-prostasin cascade might play a critical role in breast cancer.


Asunto(s)
Neoplasias de la Mama/enzimología , Serina Endopeptidasas/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/enzimología , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Epitelio/enzimología , Epitelio/patología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , ARN Interferente Pequeño/genética , Serina Endopeptidasas/genética , Especificidad por Sustrato , Distribución Tisular
17.
Prostate ; 70(13): 1422-8, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20687215

RESUMEN

BACKGROUND: Prostate cancer progression is accompanied by increased levels of extracellular proteases that are capable of remodeling the extracellular matrix as well as cleaving and activating growth factors and their receptors that are critically involved in pro-cancerous signaling pathways. The membrane anchored serine protease matriptase (also known as MT-SP1, epithin, and TADG15) has been implicated in prostate cancer. Several studies have demonstrated that the expression of this protease, both on the RNA and protein level is significantly increased during prostate cancer progression. Hepatocyte activator growth factor inhibitor-2 (HAI-2) has recently been identified as a physiological inhibitor of matriptase. It has been proposed that the increase of matriptase with a concomitant loss of its inhibitors may play a critical role in cancer progression. METHODS: In this study, we used immunohistochemistry to determine the expression of HAI-2 protein in 136 prostate cancer samples, 20 prostate benign prostatic hyperplasia (BPH) samples, and 31 normal or tumor-adjacent prostate samples. Specificity of detection was ensured by using two unrelated HAI-2 antibodies and corresponding non-immune IgG antibodies. RESULTS: We demonstrate that HAI-2 protein is significantly decreased in malignant lesions as compared to normal and BPH lesions, and that the most poorly differentiated tumors (Gleason score 8-10) have the lowest level of HAI-2 expression. CONCLUSION: These data suggest that the loss of HAI-2 may be actively involved in prostate cancer progression by causing a reduced inhibitory capacity of proteolysis possibly of the physiological target for HAI-2 matriptase.


Asunto(s)
Carcinoma/metabolismo , Glicoproteínas de Membrana/metabolismo , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Neoplasias de la Próstata/metabolismo , Carcinoma/patología , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Masculino , Próstata/patología , Hiperplasia Prostática/patología , Neoplasias de la Próstata/patología , Estadísticas no Paramétricas , Análisis de Matrices Tisulares
18.
Nat Genet ; 42(8): 676-83, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20657595

RESUMEN

Deficiency in the serine protease inhibitor LEKTI is the etiological origin of Netherton syndrome, which causes detachment of the stratum corneum and chronic inflammation. Here we show that the membrane protease matriptase initiates Netherton syndrome in a LEKTI-deficient mouse model by premature activation of a pro-kallikrein cascade. Auto-activation of pro-inflammatory pro-kallikrein-related peptidases that are associated with stratum corneum detachment was either low or undetectable, but they were efficiently activated by matriptase. Ablation of matriptase from LEKTI-deficient mice dampened inflammation, eliminated aberrant protease activity, prevented detachment of the stratum corneum, and improved the barrier function of the epidermis. These results uncover a pathogenic matriptase-pro-kallikrein pathway that could operate in several human skin and inflammatory diseases.


Asunto(s)
Calicreínas/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Epidermis/metabolismo , Epidermis/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Ratones , Síndrome de Netherton , Péptido Hidrolasas/metabolismo , Inhibidores de Serina Proteinasa/metabolismo , Piel/metabolismo , Piel/patología
19.
Proc Natl Acad Sci U S A ; 107(9): 4200-5, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20142489

RESUMEN

The intestinal epithelium serves as a major protective barrier between the mammalian host and the external environment. Here we show that the transmembrane serine protease matriptase plays a pivotol role in the formation and integrity of the intestinal epithelial barrier. St14 hypomorphic mice, which have a 100-fold reduction in intestinal matriptase mRNA levels, display a 35% reduction in intestinal transepithelial electrical resistance (TEER). Matriptase is expressed during intestinal epithelial differentiation and colocalizes with E-cadherin to apical junctional complexes (AJC) in differentiated polarized Caco-2 monolayers. Inhibition of matriptase activity using a specific peptide inhibitor or by knockdown of matriptase by siRNA disrupts the development of TEER in barrier-forming Caco-2 monolayers and increases paracellular permeability to macromolecular FITC-dextran. Loss of matriptase was associated with enhanced expression and incorporation of the permeability-associated, "leaky" tight junction protein claudin-2 at intercellular junctions. Knockdown of claudin-2 enhanced the development of TEER in matriptase-silenced Caco-2 monolayers, suggesting that the reduced barrier integrity was caused, at least in part, by an inability to regulate claudin-2 expression and incorporation into junctions. We find that matriptase enhances the rate of claudin-2 protein turnover, and that this is mediated indirectly through an atypical PKCzeta-dependent signaling pathway. These results support a key role for matriptase in regulating intestinal epithelial barrier competence, and suggest an intriguing link between pericellular serine protease activity and tight junction assembly in polarized epithelia.


Asunto(s)
Mucosa Intestinal/metabolismo , Serina Endopeptidasas/metabolismo , Células CACO-2 , Membrana Celular/enzimología , Proliferación Celular , Claudinas , Silenciador del Gen , Humanos , Proteínas de la Membrana/metabolismo , Permeabilidad , Proteína Quinasa C/metabolismo , ARN Interferente Pequeño , Serina Endopeptidasas/genética , Transducción de Señal
20.
Am J Pathol ; 175(4): 1453-63, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19717635

RESUMEN

A pericellular proteolytic pathway initiated by the transmembrane serine protease matriptase plays a critical role in the terminal differentiation of epidermal tissues. Matriptase is constitutively expressed in multiple other epithelia, suggesting a putative role of this membrane serine protease in general epithelial homeostasis. Here we generated mice with conditional deletion of the St14 gene, encoding matriptase, and show that matriptase indeed is essential for the maintenance of multiple types of epithelia in the mouse. Thus, embryonic or postnatal ablation of St14 in epithelial tissues of diverse origin and function caused severe organ dysfunction, which was often associated with increased permeability, loss of tight junction function, mislocation of tight junction-associated proteins, and generalized epithelial demise. The study reveals that the homeostasis of multiple simple and stratified epithelia is matriptase-dependent, and provides an important animal model for the exploration of this membrane serine protease in a range of physiological and pathological processes.


Asunto(s)
Epitelio/enzimología , Procesamiento Proteico-Postraduccional , Serina Endopeptidasas/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Alelos , Animales , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/patología , Epitelio/efectos de los fármacos , Eliminación de Gen , Homeostasis/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/enzimología , Intestinos/patología , Megacolon/enzimología , Megacolon/patología , Proteínas de la Membrana/metabolismo , Ratones , Especificidad de Órganos/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Glándulas Salivales/efectos de los fármacos , Glándulas Salivales/enzimología , Glándulas Salivales/patología , Serina Endopeptidasas/deficiencia , Tamoxifeno/farmacología , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA