Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Biol Chem ; 295(20): 7168-7177, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32241917

RESUMEN

Serine protease 14 (Prss14)/epithin is a transmembrane serine protease that plays essential roles in tumor progression and metastasis and therefore is a promising target for managing cancer. Prss14/epithin shedding may underlie its activity in cancer and worsen outcomes; accordingly, a detailed understanding of the molecular mechanisms in Prss14/epithin shedding may inform the design of future cancer therapies. On the basis of our previous observation that an activator of PKC, phorbol 12-myristate 13-acetate (PMA), induces Prss14/epithin shedding, here we further investigated the intracellular signaling pathway involved in this process. While using mitogen-activated protein kinase inhibitors to investigate possible effectors of downstream PKC signaling, we unexpectedly found that an inhibitor of c-Jun N-terminal kinase (JNK), SP600125, induces Prss14/epithin shedding even in the absence of PMA. SP600125-induced shedding, like that stimulated by PMA, was mediated by tumor necrosis factor-α-converting enzyme. In contrast, a JNK activator, anisomycin, partially abolished the effects of SP600125 on Prss14/epithin shedding. Moreover, the results from loss-of-function experiments with specific inhibitors, short hairpin RNA-mediated knockdown, and overexpression of dominant-negative PKCßII variants indicated that PKCßII is a major player in JNK inhibition- and PMA-mediated Prss14/epithin shedding. SP600125 increased phosphorylation of PKCßII and tumor necrosis factor-α-converting enzyme and induced their translocation into the plasma membrane. Finally, in vitro cell invasion experiments and bioinformatics analysis of data in The Cancer Genome Atlas breast cancer database revealed that JNK and PKCßII are important for Prss14/epithin-mediated cancer progression. These results provide important information regarding strategies against tumor metastasis.


Asunto(s)
Antracenos/farmacología , MAP Quinasa Quinasa 4/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteína Quinasa C beta/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Serina Endopeptidasas/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Humanos , MAP Quinasa Quinasa 4/metabolismo , Metástasis de la Neoplasia , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Proteína Quinasa C beta/genética , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Serina Endopeptidasas/genética , Acetato de Tetradecanoilforbol/farmacología
2.
BMC Biol ; 18(1): 60, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493324

RESUMEN

BACKGROUND: Epithin/PRSS14, a type II transmembrane serine protease, is an emerging target of cancer therapy because of its critical roles in tumor progression and metastasis. In many circumstances, the protease, through its ectodomain shedding, exists as a soluble form and performs its proteolytic functions in extracellular environments increasing cellular invasiveness. The seemingly functional integrity of the soluble form raises the question of why the protease is initially made as a membrane-associated protein. RESULTS: In this report, we show that the epithin/PRSS14 intracellular domain (EICD) can be released from the membrane by the action of signal peptide peptidase-like 2b (SPPL2b) after ectodomain shedding. The EICD preferentially localizes in the nucleus and can enhance migration, invasion, and metastasis of epithelial cancer when heterologously expressed. Unbiased RNA-seq analysis and subsequent antibody arrays showed that EICD could control the gene expression of chemokines involved in cell motility, by increasing their promoter activities. Finally, bioinformatics analysis provided evidence for the clinical significance of the intramembrane proteolysis of epithin/PRSS14 by revealing that the poor survival of estrogen receptor (ER)-negative breast cancer patients with high epithin/PRSS14 expression is further worsened by high levels of SPPL2b. CONCLUSIONS: These results show that ectodomain shedding of epithin/PRSS14 can initiate a unique and synchronized bidirectional signal for cancer metastasis: extracellularly broadening proteolytic modification of the surrounding environment and intracellularly reprogramming the transcriptome for metastatic conversion. Clinically, this study also suggests that the intracellular function of epithin/PRSS14 should be considered for targeting this protease for anti-cancer treatment.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de la Membrana/genética , Proteolisis , Serina Endopeptidasas/genética , Animales , Neoplasias de la Mama/fisiopatología , Movimiento Celular , Núcleo Celular/metabolismo , Células Cultivadas , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Serina Endopeptidasas/metabolismo
3.
J Cell Sci ; 129(10): 2030-42, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27044755

RESUMEN

Vimentin, an intermediate filament protein induced during epithelial-to-mesenchymal transition, is known to regulate cell migration and invasion. However, it is still unclear how vimentin controls such behaviors. In this study, we aimed to find a new integrin regulator by investigating the H-Ras-mediated integrin suppression mechanism. Through a proteomic screen using the integrin ß3 cytoplasmic tail protein, we found that vimentin might work as an effector of H-Ras signaling. H-Ras converted filamentous vimentin into aggregates near the nucleus, where no integrin binding can occur. In addition, an increase in the amount of vimentin filaments accessible to the integrin ß3 tail enhanced talin-induced integrin binding to its ligands by inducing integrin clustering. In contrast, the vimentin head domain, which was found to bind directly to the integrin ß3 tail and compete with endogenous vimentin filaments for integrin binding, induced nuclear accumulation of vimentin filaments and reduced the amount of integrin-ligand binding. Finally, we found that expression of the vimentin head domain can reduce cell migration and metastasis. From these data, we suggest that filamentous vimentin underneath the plasma membrane is involved in increasing integrin adhesiveness, and thus regulation of the vimentin-integrin interaction might control cell adhesion.


Asunto(s)
Adhesión Celular/genética , Citoesqueleto/metabolismo , Integrina beta3/genética , Vimentina/genética , Animales , Células CHO , Membrana Celular/genética , Membrana Celular/metabolismo , Movimiento Celular/genética , Cricetinae , Cricetulus , Citoesqueleto/genética , Transición Epitelial-Mesenquimal/genética , Humanos , Integrina beta3/metabolismo , Ligandos , Unión Proteica , Mapas de Interacción de Proteínas , Proteómica , Vimentina/metabolismo
4.
Biochem Biophys Res Commun ; 452(4): 1084-90, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25245289

RESUMEN

Epithin/PRSS14, a type II transmembrane serine protease, plays critical roles in cancer metastasis. Previously, we have reported that epithin/PRSS14 undergoes ectodomain shedding in response to phorbol myristate acetate (PMA) stimulation. In this study, we show that transforming growth factor-ß (TGF-ß) induces rapid epithin/PRSS14 shedding through receptor mediated pathway in 427.1.86 thymoma cells. Tumor necrosis factor-α converting enzyme (TACE) is responsible for this shedding. Amino acid sequence encompassing the putative shedding cleavage site of epithin/PRSS14 exhibit strong homology to the cleavage site of l-selectin, a known TACE substrate. TACE inhibitor, TAPI-0 and TACE siRNA greatly reduced TGF-ß-induced epithin/PRSS14 shedding. TGF-ß treatment induces translocation of intracellular pool of TACE to the membrane where epithin/PRSS14 resides. These findings suggest that TGF-ß induces epithin/PRSS14 shedding by mediating translocation of epithin/PRSS14 sheddase, TACE, to the membrane.


Asunto(s)
Proteínas ADAM/metabolismo , Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína ADAM17 , Animales , Línea Celular , Ratones , Transporte de Proteínas/fisiología
5.
Blood ; 117(4): 1415-24, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21097670

RESUMEN

Epithin/PRSS14, a type II transmembrane serine protease, is involved in normal epithelial development and tumor progression. Here we report, as an interacting substrate of epithin, a receptor tyrosine kinase Tie2 that is well known for important roles in the vessel stability. Epithin interacts with and degrades the Tie2 extracellular portion that contains the ligand-binding domain. Epithin is located in the neighbor of Tie2-expressing vessels in normal tissue. Furthermore, epithin can cleave and degrade Tie2 not only in the same cell but also from neighboring cells nearby, resulting in the degradation of the Tie2 ectodomain. The remaining Tie2 fragment was highly phosphorylated and was able to recruit a downstream effector, phosphatidylinositol 3-kinase. Knocking down epithin expression using short hairpin RNA in thymoma cell severely impaired the migration through endothelial cells that show the actin rearrangement during the process. The diminution of epithin protein expression in 4T1 breast cancer cells caused the significant decrease in the number of transendothelial migrating cells in vitro as well as in those of metastasizing tumor nodules in vivo, Therefore, we propose that epithin, which regulates endothelial Tie2 functions, plays a critical role in the fine tuning of transendothelial migration for normal and cancer cells.


Asunto(s)
Procesamiento Proteico-Postraduccional , Receptor TIE-2/metabolismo , Serina Endopeptidasas/fisiología , Migración Transendotelial y Transepitelial/fisiología , Animales , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , Serina Endopeptidasas/química , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/genética , Transfección
6.
Mol Cells ; 45(8): 564-574, 2022 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-35950457

RESUMEN

Epithin/PRSS14 is a membrane serine protease that plays a key role in tumor progression. The protease exists on the cell surface until its ectodomain shedding, which releases most of the extracellular domain. Previously, we showed that the remaining portion on the membrane undergoes intramembrane proteolysis, which results in the liberation of the intracellular domain and the intracellular domainmediated gene expression. In this study, we investigated how the intramembrane proteolysis for the nuclear function is initiated. We observed that ectodomain shedding of epithin/PRSS14 in mouse breast cancer 4T1 cells increased depending on environmental conditions and was positively correlated with invasiveness of the cells and their proinvasive cytokine production. We identified selenite as an environmental factor that can induce ectodomain shedding of the protease and increase C-C motif chemokine ligand 2 (CCL2) secretion in an epithin/PRSS14-dependent manner. Additionally, by demonstrating that the expression of the intracellular domain of epithin/PRSS14 is sufficient to induce CCL2 secretion, we established that epithin/PRSS14- dependent shedding and its subsequent intramembrane proteolysis are responsible for the metastatic conversion of 4T1 cells under these conditions. Consequently, we propose that epithin/PRSS14 can act as an environment-sensing receptor that promotes cancer metastasis by liberating the intracellular domain bearing transcriptional activity under conditions promoting ectodomain shedding.


Asunto(s)
Proteínas de la Membrana/metabolismo , Neoplasias , Serina Endopeptidasas/metabolismo , Animales , Membrana Celular/metabolismo , Quimiocinas/metabolismo , Ligandos , Ratones , Neoplasias/patología
7.
Biochem Biophys Res Commun ; 405(4): 644-50, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21295011

RESUMEN

PRSS14/Epithin (also known as matriptase and ST14), a member of the type II transmembrane serine proteases, is primarily found in a subpopulation of normal epithelial cells and in epithelial cancers. Its known functions include maintaining the epithelial barrier, thymic development, and cancer progression. In this study, we show that several macrophage cell lines and activated bone marrow-derived macrophages also express PRSS14/Epithin. Surface expression, as well as cytoplasmic expression, was detectable upon activation by IFN-γ, but not TNF-α or TGF-ß. Induction of the protein appeared to be restricted to macrophages. IFN-γ showed a biphasic regulation in RAW264.7 cells, and upregulated expression was sustained for several days. This induction by IFN-γ was partially through the increase of PRSS14/Epithin mRNA production, which is downstream of the JAK pathway, shown by the inhibition by tyrphostin AG490. Using chromatin immunoprecipitation, we verified that two sites among six putative STAT1 binding sites in the PRSS14/Epithin promoter were occupied by STAT1 upon activation. Treatment with IFN-γ enhanced the serum-triggered transendothelial migration of RAW264.7 cells, but not that of PRSS14/Epithin knock-down RAW264.7 cells, although they express multiple markers such as ICAM1, CD80, and CD40 at normal levels. These data strongly suggest that PRSS14/Epithin plays an important role in the transendothelial migration of activated macrophages in the inflammatory microenvironment, and the mode of action is similar to the events in cancer metastasis.


Asunto(s)
Movimiento Celular , Interferón gamma/metabolismo , Quinasas Janus/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Factor de Transcripción STAT1/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Línea Celular , Células Endoteliales/inmunología , Matriz Extracelular/inmunología , Interferón gamma/farmacología , Macrófagos/efectos de los fármacos , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Serina Endopeptidasas/genética
8.
PLoS Biol ; 6(8): e191, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18684012

RESUMEN

Although much effort has been directed at dissecting the mechanisms of central tolerance, the role of thymic stromal cells remains elusive. In order to further characterize this event, we developed a mouse model restricting LacZ to thymic stromal cotransporter (TSCOT)-expressing thymic stromal cells (TDLacZ). The thymus of this mouse contains approximately 4,300 TSCOT+ cells, each expressing several thousand molecules of the LacZ antigen. TSCOT+ cells express the cortical marker CDR1, CD40, CD80, CD54, and major histocompatibility complex class II (MHCII). When examining endogenous responses directed against LacZ, we observed significant tolerance. This was evidenced in a diverse T cell repertoire as measured by both a CD4 T cell proliferation assay and an antigen-specific antibody isotype analysis. This tolerance process was at least partially independent of Autoimmune Regulatory Element gene expression. When TDLacZ mice were crossed to a novel CD4 T cell receptor (TCR) transgenic reactive against LacZ (BgII), there was a complete deletion of double-positive thymocytes. Fetal thymic reaggregate culture of CD45- and UEA-depleted thymic stromal cells from TDLacZ and sorted TCR-bearing thymocytes excluded the possibility of cross presentation by thymic dendritic cells and medullary epithelial cells for the deletion. Overall, these results demonstrate that the introduction of a neoantigen into TSCOT-expressing cells can efficiently establish complete tolerance and suggest a possible application for the deletion of antigen-specific T cells by antigen introduction into TSCOT+ cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Epiteliales/inmunología , Tolerancia Inmunológica , Operón Lac/inmunología , Simportadores/inmunología , Timo/citología , Animales , Células Presentadoras de Antígenos/inmunología , Diferenciación Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Activación de Linfocitos , Ratones , Ratones Transgénicos , Células del Estroma/citología , Células del Estroma/inmunología , Simportadores/genética , Timo/inmunología
9.
Biochem Biophys Res Commun ; 395(4): 553-9, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20398629

RESUMEN

The epithelial-derived type II transmembrane serine protease epithin has been shown to be upregulated in a variety of cancer cell lines and tumor tissues, and its upregulation correlates well with tumor progression in many cases. However, little is known regarding the regulation of its expression and the mechanism of its roles in tumor progression. Here, we show that transforming growth factor-beta (TGF-beta), a potent inducer of epithelial-mesenchymal transition (EMT) in tumor progression, upregulates epithin, and that epithin plays a critical role in TGF-beta-induced EMT. Forced overexpression of epithin induced EMT to exhibit characteristic morphological changes, alternations in EMT-related proteins and enhanced cell motility. Conversely, shRNA-mediated knockdown of endogenous epithin inhibited TGF-beta-induced expression of mesenchymal markers and morphological changes. Furthermore, TGF-beta-induced cell migration and invasion were significantly impaired by epithin knockdown. In addition, we demonstrate that TGF-beta upregulates epithin transcriptionally via the Smad2/Smad4-mediated pathway. These results suggest that epithin is a key mediator of TGF-beta-induced EMT in tumor progression.


Asunto(s)
Transdiferenciación Celular/genética , Epitelio/patología , Regulación Neoplásica de la Expresión Génica , Mesodermo/patología , Neoplasias/patología , Serina Endopeptidasas/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Perros , Epitelio/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas de la Membrana , Mesodermo/metabolismo , Ratones , Neoplasias/genética , ARN Interferente Pequeño/genética , Transducción de Señal , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Activación Transcripcional , Regulación hacia Arriba
10.
Immune Netw ; 20(4): e33, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32895620

RESUMEN

We tested how adjuvants effect in a cancer vaccine model using an epitope derived from an autoactivation loop of membrane-type protease serine protease 14 (PRSS14; loop metavaccine) in mouse mammary tumor virus (MMTV)-polyoma middle tumor-antigen (PyMT) system and in 2 other orthotopic mouse systems. Earlier, we reported that loop metavaccine effectively prevented progression and metastasis regardless of adjuvant types and TH types of hosts in tail-vein injection systems. However, the loop metavaccine with Freund's complete adjuvant (CFA) reduced cancer progression and metastasis while that with alum, to our surprise, were adversely affected in 3 tumor bearing mouse models. The amounts of loop peptide specific antibodies inversely correlated with tumor burden and metastasis, meanwhile both TH1 and TH2 isotypes were present regardless of host type and adjuvant. Tumor infiltrating myeloid cells such as eosinophil, monocyte, and neutrophil were asymmetrically distributed among 2 adjuvant groups with loop metavaccine. Systemic expression profiling using the lymph nodes of the differentially immunized MMTV-PyMT mouse revealed that adjuvant types, as well as loop metavaccine can change the immune signatures. Specifically, loop metavaccine itself induces TH2 and TH17 responses but reduces TH1 and Treg responses regardless of adjuvant type, whereas CFA but not alum increased follicular TH response. Among the myeloid signatures, eosinophil was most distinct between CFA and alum. Survival analysis of breast cancer patients showed that eosinophil chemokines can be useful prognostic factors in PRSS14 positive patients. Based on these observations, we concluded that multiple immune parameters are to be considered when applying a vaccine strategy to cancer patients.

11.
J Exp Clin Cancer Res ; 38(1): 363, 2019 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-31426843

RESUMEN

BACKGROUND: In order to develop a new immunotherapeutic agent targeting metastatic breast cancers, we chose to utilize autocatalytic feature of the membrane serine protease Prss14/ST14, a specific prognosis marker for ER negative breast cancer as a target molecule. METHODS: The study was conducted using three mouse breast cancer models, 4 T1 and E0771 mouse breast cancer cells into their syngeneic hosts, and an MMTV-PyMT transgenic mouse strain was used. Prss14/ST14 knockdown cells were used to test function in tumor growth and metastasis, peptides derived from the autocatalytic loop for activation were tested as preventive metastasis vaccine, and monoclonal and humanized antibodies to the same epitope were tested as new therapeutic candidates. ELISA, immunoprecipitation, Immunofluorescent staining, and flow cytometry were used to examine antigen binding. The functions of antibodies were tested in vitro for cell migration and in vivo for tumor growth and metastasis. RESULTS: Prss14/ST14 is critically involved in the metastasis of breast cancer and poor survival rather than primary tumor growth in two mouse models. The epitopes derived from the specific autocatalytic loop region of Prss14/ST14, based on structural modeling acted as efficient preventive metastasis vaccines in mice. A new specific monoclonal antibody mAb3F3 generated against the engineered loop structure could reduce cell migration, eliminate metastasis in PyMT mice, and can detect the Prss14/ST14 protein expressed in various human cancer cells. Humanized antibody huAb3F3 maintained the specificity and reduced the migration of human breast cancer cells in vitro. CONCLUSION: Our study demonstrates that Prss14/ST14 is an important target for modulating metastasis. Our newly developed hybridoma mAbs and humanized antibody can be further developed as new promising candidates for the use in diagnosis and in immunotherapy of human metastatic breast cancer.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales/farmacología , Neoplasias de la Mama/prevención & control , Epítopos/inmunología , Neoplasias Pulmonares/prevención & control , Fragmentos de Péptidos/inmunología , Serina Endopeptidasas/inmunología , Animales , Antígenos Transformadores de Poliomavirus/genética , Apoptosis , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Nat Commun ; 9(1): 225, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29335408

RESUMEN

Whether hematopoietic cell-restricted distribution of antigens affects the degree of thymic negative selection has not been investigated in detail. Here, we show that T cells specific for hematopoietic cell-restricted antigens (HRA) are not completely deleted in the thymus, using the mouse minor histocompatibility antigen H60, the expression of which is restricted to hematopoietic cells. As a result, low avidity T cells escape from thymic deletion. This incomplete thymic deletion occurs to the T cells developing de novo in the thymus of H60-positive recipients in H60-mismatched bone marrow transplantation (BMT). H60-specific thymic deletion escapee CD8+ T cells exhibit effector differentiation potentials in the periphery and contribute to graft-versus-leukemia effects in the recipients of H60-mismatched BMT, regressing H60+ hematological tumors. These results provide information essential for understanding thymic negative selection and developing a strategy to treat hematological tumors.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Madre Hematopoyéticas/inmunología , Antígenos de Histocompatibilidad Menor/inmunología , Timo/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Trasplante de Médula Ósea/métodos , Linfocitos T CD8-positivos/metabolismo , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Ratones Noqueados , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Timocitos/inmunología , Timocitos/metabolismo , Timo/metabolismo , Inmunología del Trasplante/inmunología
13.
Oncotarget ; 7(23): 34643-63, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27167193

RESUMEN

To elucidate the role of a type II transmembrane serine protease, ST14/Prss14, during breast cancer progression, we utilized publically accessible databases including TCGA, GEO, NCI-60, and CCLE. Survival of breast cancer patients with high ST14/Prss14 expression is significantly poor in estrogen receptor (ER) negative populations regardless of the ratios of ST14/Prss14 to its inhibitors, SPINT1 or SPINT2. In a clustering of 1085 selected EMT signature genes, ST14/Prss14 is located in the same cluster with CDH3, and closer to post-EMT markers, CDH2, VIM, and FN1 than to the pre-EMT marker, CDH1. Coexpression analyses of known ST14/Prss14 substrates and transcription factors revealed context dependent action. In cell lines, paradoxically, ST14/Prss14 expression is higher in the ER positive group and located closer to CDH1 in clustering. This apparent contradiction is not likely due to ST14/Prss14 expression in a cancer microenvironment, nor due to negative regulation by ER. Genes consistently coexpressed with ST14/Prss14 include transcription factors, ELF5, GRHL1, VGLL1, suggesting currently unknown mechanisms for regulation. Here, we report that ST14/Prss14 is an emerging therapeutic target for breast cancer where HER2 is not applicable. In addition we suggest that careful conclusions should be drawn not exclusively from the cell line studies for target development.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Receptores de Estrógenos/metabolismo , Serina Endopeptidasas/metabolismo , Biomarcadores de Tumor/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Pronóstico , Proteínas Inhibidoras de Proteinasas Secretoras/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Represoras/metabolismo , Serina Endopeptidasas/genética , Factores de Transcripción/metabolismo
14.
J Immunol Methods ; 297(1-2): 265-70, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15777949

RESUMEN

We generated monoclonal antibodies reacting to a mouse thymic epithelial cell specific membrane protein, Thymic Stromal Co-transporter (TSCOT)/Ly110. These antibodies showed specificity to the peptide sequences derived from TSCOT/Ly110 determined by specific peptide inhibition in flow cytometric analyses with cells expressing the protein on the surface. TSCOT/Ly110 expressing subpopulation can be identified among the CDR1(+) or 6C3(+) cortical epithelial cells. Furthermore, CDR1 positive cortical thymic epithelial cells can be separated into further distinguishable populations; CDR1(+)6C3(+)Ly110(+), CDR1(+)6C3(-)/(low)Ly110(+), CDR1(+)Ly110(-). Some of TSCOT/Ly110 expressing cells negative for both CDR1 and 6C3 markers were found at the earlier stages of development, while most of the cells are positive for both at 1-week-old stage. After then, downregulation in 6C3 and/or CDR1 expression was noticed until 16 weeks of age. These results suggest that TSCOT/Ly110 is a new marker for the subpopulation of CDR1(+) or 6C3(+) epithelial cells in the neonatal and adult thymus and is useful for the studies on the epithelial cell differentiation process.


Asunto(s)
Células Epiteliales/citología , Citometría de Flujo , Simportadores/análisis , Timo/citología , Animales , Anticuerpos Monoclonales/inmunología , Biomarcadores/análisis , Diferenciación Celular , Células Epiteliales/clasificación , Células Epiteliales/inmunología , Ratones , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/inmunología , Simportadores/inmunología , Timo/inmunología
15.
Mol Cells ; 38(6): 548-61, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26013383

RESUMEN

By combining conventional single cell analysis with flow cytometry and public database searches with bioinformatics tools, we extended the expression profiling of thymic stromal cotransporter (TSCOT), Slc46A2/Ly110, that was shown to be expressed in bipotent precursor and cortical thymic epithelial cells. Genome scale analysis verified TSCOT expression in thymic tissue- and cell type- specific fashion and is also expressed in some other epithelial tissues including skin and lung. Coexpression profiling with genes, Foxn1 and Hoxa3, revealed the role of TSCOT during the organogenesis. TSCOT expression was detected in all thymic epithelial cells (TECs), but not in the CD31(+) endothelial cell lineage in fetal thymus. In addition, ABC transporter-dependent side population and Sca-1(+) fetal TEC populations both contain TSCOT-expressing cells, indicating TEC stem cells express TSCOT. TSCOT expression was identified as early as in differentiating embryonic stem cells. TSCOT expression is not under the control of Foxn1 since TSCOT is present in the thymic rudiment of nude mice. By searching variations in the expression levels, TSCOT is positively associated with Grhl3 and Irf6. Cytokines such as IL1b, IL22 and IL24 are the potential regulators of the TSCOT expression. Surprisingly, we found TSCOT expression in the lung is diminished in lung cancers, suggesting TSCOT may be involved in the suppression of lung tumor development. Based on these results, a model for TEC differentiation from the stem cells was proposed in context of multiple epithelial organ formation.


Asunto(s)
Células Epiteliales/metabolismo , Células Madre/metabolismo , Simportadores/biosíntesis , Timo/metabolismo , Animales , Diferenciación Celular/fisiología , Células Epiteliales/citología , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Ratones , Ratones Desnudos , Regiones Promotoras Genéticas , Simportadores/genética , Timo/patología
16.
Virchows Arch ; 464(1): 19-27, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24248283

RESUMEN

Matriptase is one of the type II transmembrane serine proteases and is known to be involved in cancer progression. Increased matriptase expression has been reported in a variety of human cancers, and its association with poor prognosis has been highlighted in some cancer types. However, its exact role in cancer progression and its effect on patient survival in esophageal squamous cell carcinoma (ESCC) are still unclear. We performed immunohistochemical staining of matriptase in 171 ESCC samples after antibody validation and evaluated the association of its expression with clinicopathological parameters and prognosis. High matriptase expression was observed in 38.6 % (66/171) of ESCC samples and more frequently in N3 stage and in poorly differentiated tumors. Both overall survival (OS) and disease-free survival (DFS) were significantly lower for patients with high expression of matriptase than for patients with low expression (5-year OS rate, 38.6 vs 55.3 %; p=0.034 and 5-year DFS rate, 30.5 vs 49.4 %; p=0.007). High matriptase expression was an independent prognostic factor for OS [hazard ratio (HR), 1.65 (95 % confidence interval (CI), 1.01-2.68); p=0.045] and for DFS [HR, 1.79 (95 % CI, 1.14-2.81); p=0.012]. In conclusion, higher expression of matriptase is an independent prognostic factor involved in the progression of ESCC, which suggests that matriptase is a factor in ESCC tumor progression and also a potential molecular therapeutic target.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/mortalidad , Neoplasias Esofágicas/enzimología , Neoplasias Esofágicas/mortalidad , Serina Endopeptidasas/análisis , Adulto , Anciano , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Femenino , Células HEK293 , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pronóstico , Serina Endopeptidasas/fisiología
17.
Mol Cells ; 34(5): 481-93, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23178972

RESUMEN

In order to investigate the role of thymic epithelial cell (TEC) subsets during T-cell development, we established a new transgenic system, enabling inducible cell-specific ablation as well as marking the TEC subsets using bicistronic bacterial nitroreductase and EGFP genes. Two different lengths of the TSCOT promoter in transgenic mice, named 3.1T-NE and 9.1T-NE, drive EGFP expression into TECs. In adult life, EGFP expression was located in the medulla with a smaller 3.1 kb TSCOT promoter, while it was maintained in the cortex with a 9.1 kb promoter, suggesting putative TEC specific as well as compartment specific cis elements within two promoters. Nitroreductase induced cell death was specific without bystander killing upon the treatment of prodrugs such as nitrofurantoin and metronidazol. The degree of cell death was dependent on the dose of the prodrug in the cell and the fetal thymic organ cultures (FTOCs). Fetal thymic stromal populations were analyzed based on the expression levels of EpCAM, MHCII, CDR1 and/or UEA-1. EGFP expression patterns varied among subsets indicating the differential TSCOT promoter activity in each TEC subset. Prodrug treatment in FTOCs reduced the numbers of total and subsets of thymocytes. A CD4(+)CD8(+) double positive cell population was highly susceptible in both transgenic lines. Surprisingly, there was a distinct reduction in γδ T cell population only in the 9.1T-NE thymus, indicating that they require a NTREGFP expressing TEC population. Therefore, these results support a division of labor within TEC subsets for the αß and γδ lineage specification.


Asunto(s)
Células Epiteliales/citología , Regiones Promotoras Genéticas , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Simportadores/genética , Subgrupos de Linfocitos T/citología , Timo/metabolismo , Células 3T3 , Animales , Diferenciación Celular , Células Epiteliales/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Subgrupos de Linfocitos T/metabolismo
18.
Mol Cells ; 29(6): 617-23, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20652801

RESUMEN

Epithin (PRSS14/matriptase/ST14), a type II membrane protein, is involved in progression of epithelial cancers and metastasis as well as in the normal epidermal barrier function. When activated, it translocates into the cell-cell contacts and sheds into media. In order to understand the specific mechanism during tumor progression, we tested the angiogenic potential of secreted form of epithin. Epithin produced from the cancer cells shed more in hypoxia and induced motility of endothelial cells. Epithin enhanced the migration and invasion of mouse and bovine endothelial cells without cell proliferation. Furthermore, soluble epithin induced endothelial differentiation in the assay of the human endothelial microvessel-like tube formation and in that of the chicken chorioallantoic membrane. The knock-down of epithin in the 427 thymoma cell line abolished the protease activity of secreted epithin fraction, reduced the invasion of endothelial cells through matrigel, and tube formation activity. Only specific antibodies abolished the migration of endothelial cell and the vessel morphogenesis, suggesting that epithin specifically functions in these systems. Therefore, we propose that the secreted epithin in the hypoxic cancer microenvironment plays a role as a proangiogenic factor, and can be modulated with specific antibodies.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Carcinoma/metabolismo , Células Endoteliales/metabolismo , Neoplasias/metabolismo , Serina Endopeptidasas/metabolismo , Inductores de la Angiogénesis/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Vasos Sanguíneos/crecimiento & desarrollo , Carcinoma/irrigación sanguínea , Carcinoma/genética , Carcinoma/patología , Carcinoma/fisiopatología , Bovinos , Diferenciación Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Pollos , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/crecimiento & desarrollo , Progresión de la Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Humanos , Melanoma Experimental , Proteínas de la Membrana , Ratones , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Células 3T3 NIH , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/genética , Neoplasias/patología , Neoplasias/fisiopatología , Neovascularización Patológica , ARN Interferente Pequeño/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/inmunología , Transgenes/genética
19.
EMBO Rep ; 6(11): 1045-51, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16170303

RESUMEN

Epithin is a type II transmembrane serine protease that exists in a soluble and membrane-bound form. Shedding is thought to be important in regulating its action, but little is known regarding the intracellular events that trigger such shedding. Here, we show that phorbol myristate acetate (PMA) causes the release of epithin. It also causes accumulation of the protein at the site of cell-cell contacts, and this accumulation is dependent on the formation of cortical actin. In addition, we have identified the actin-binding protein, filamin, as the linker between epithin and the actin cytoskeleton. The interaction of epithin and filamin was enhanced by PMA, and epithin was not released from filamin-deficient M2 cells. We also show that the release of epithin does not require its own activity and is blocked by a metalloprotease inhibitor, GM6001. These results show that filamin has an essential role in shedding by linking epithin to the as yet unidentified metalloprotease-shedding enzyme(s).


Asunto(s)
Proteínas Contráctiles/metabolismo , Proteínas de Microfilamentos/metabolismo , Procesamiento Proteico-Postraduccional , Serina Endopeptidasas/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Proteínas Contráctiles/genética , Citocalasina D/farmacología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Dipéptidos/farmacología , Filaminas , Humanos , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/metabolismo , Metaloendopeptidasas/antagonistas & inhibidores , Proteínas de Microfilamentos/genética , Datos de Secuencia Molecular , Transporte de Proteínas/efectos de los fármacos , Serina Endopeptidasas/genética , Acetato de Tetradecanoilforbol/antagonistas & inhibidores , Transfección , Técnicas del Sistema de Dos Híbridos
20.
Immunity ; 23(4): 375-86, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16226503

RESUMEN

Conventional understanding of CD4 T cell development is that the MHC class II molecules on cortical thymic epithelial cell are necessary for positive selection, as demonstrated in mouse models. Clinical data, however, show that hematopoietic stem cells reconstitute CD4 T cells in patients devoid of MHC class II. Additionally, CD4 T cells generated from human stem cells in immunocompromised mice were restricted to human, but not mouse, MHC class II. These studies suggest an alternative pathway for CD4 T cell development that does not normally exist in mice. MHC class II is expressed on developing human thymocytes, indicating a possible role of MHC II on thymocytes for CD4 T cell generation. Therefore, we created mice in which MHC class II is expressed only on T lineage cells. Remarkably, the CD4 compartment in such mice is efficiently reconstituted with unique specificity, demonstrating a novel thymocyte-driven pathway of CD4 T cell selection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Genes MHC Clase II/inmunología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Timo/inmunología , Animales , Antígenos de Diferenciación de Linfocitos T/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Células Epiteliales/inmunología , Humanos , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Timo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA