Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Immunol ; 358: 104237, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33137650

RESUMEN

We previously reported that protein tyrosine phosphatase non-receptor type 3 (PTPN3), which is upregulated in activated lymphocytes, acts as an immune checkpoint. However, the mechanism by which PTPN3 expression is enhanced in activated lymphocytes is unknown. In this study, we analyzed the mechanism of PTPN3 expression in activated lymphocytes with a view for developing a novel immune checkpoint inhibitor that suppresses PTPN3. Through the activation process, lymphocytes showed enhanced NFκB activation as well as increased PTPN3 expression. NFκB enhanced proliferation, migration, and cytotoxicity of lymphocytes. Furthermore, NFκB enhanced PTPN3 expression and tyrosine kinase activation. TGFß reduced PTPN3 expression and NFκB activation in the cancer microenvironment, and suppressed the biological activity of lymphocytes. The results of this study are expected to provide significant implications for improving existing immunotherapy and developing novel immunotherapy.


Asunto(s)
FN-kappa B/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Humanos , Activación de Linfocitos/inmunología , Linfocitos/metabolismo , FN-kappa B/fisiología , Fosforilación/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/fisiología
2.
Mol Cancer Res ; 18(6): 903-912, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32169891

RESUMEN

The underlying molecular mechanism driving clear cell renal cell carcinoma (ccRCC) progression is still to be explored. The significant downregulation of protein tyrosine phosphatase nonreceptor type 3 (PTPN3) expression in the tumor tissues suggested its protective role in ccRCC progression. IHC analysis of PTPN3 protein in 172 ccRCC tissue revealed that PTPN3 was an independently favorable prognostic factor for progression-free survival (P = 0.0166) and overall survival (P = 0.0343) of patients. The ccRCC cell lines SN12C, 1932, ACHN, and Caki-1 were used to evaluate, both in vitro and in vivo, the biological roles of PTPN3. We observed that overexpression of PTPN3 significantly inhibited the proliferation, migration, and invasion of ccRCC cells. In contrast, the knocking down of PTPN3 elicited opposite effects. Overexpressing PTPN3 inhibited xenograft tumor growth and lung metastasis displayed by the in vivo mice models. PTPN3 inhibited tumor cell motility by suppressing the phosphorylation of AKT, and subsequently inactivating the PI3K/AKT signaling pathway of renal cell carcinoma cells. Furthermore, the inhibition of phospho-AKTThr308 and phospho-AKTSer473 reversed PTPN3-induced silencing in tumor cell migration. Our work revealed that the overexpression of PTPN3 could suppress kidney cancer progression by negatively regulating the AKT signaling pathway, and served as a favorable prognostic factor in patients with ccRCC. Our findings provided insight that PTPN3 could be a potential target for therapy aiming to inhibit the malignant behaviors of ccRCC. IMPLICATIONS: PTPN3 is an independent favorable prognostic factor for patients with ccRCC and could be a potential target for therapy aiming to inhibit the malignant behaviors of ccRCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/prevención & control , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/prevención & control , Fosfatidilinositol 3-Quinasas/química , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Animales , Apoptosis , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/secundario , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Fosforilación , Pronóstico , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Gene ; 733: 144383, 2020 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-31972307

RESUMEN

We elucidate in this study that up-regulation of miR-574-5p in gastric cancer cells under hypoxic conditions contributed to angiogenesis. We found that miR-574-5p and HIF-1α were up-regulated in gastric cancer cells cultured under 2% O2 or in medium containing CoCl2, and in muscle tissues of mice injected with NaNO2, indicating up-regulation of miR-574-5p in vitro or in vivo in response to hypoxic conditions. We hypothesized that up-regulation of miR-574-5p could promote angiogenesis. Transfection of gastric cancer cells with miR-574-5p mimics or inhibitor resulted in increase or decrease in the expression of VEGFA. Viability, migration, invasion and tube formation of HUVECs cultured with conditioned medium from SGC/574 cells transfected with miR-574-5p inhibitor were reduced. Tube formation of HUVECs cultured with conditioned medium from SGC-7901 cells transfected with miR-574-5p mimics was increased. An in vivo study demonstrated that inhibition of miR-574-5p in the tumor xenografts of mice reduced the expression of CD31 one of the endothelial cell markers. We identified PTPN3 a tyrosine phosphatase as a target of miR-574-5p that bound to the 3'UTR of PTPN3 mRNA to inhibit the expression of PTPN3. Furthermore, the data in this study demonstrated that inhibition of PTPN3 in gastric cancer cells enhanced phosphorylation of p44/42 MAPKs and promoted angiogenesis. We conclude that miR-574-5p in gastric cancer cells promoted angiogenesis via enhancing phosphorylation of p44/42 MAPKs by miR-574-5p inhibition of PTPN3 expression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neovascularización Patológica , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Neoplasias Gástricas/irrigación sanguínea , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Biomed Pharmacother ; 121: 109583, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31706106

RESUMEN

BACKGROUND: Perihilar cholangiocarcinoma (PHCCA) is the most common type of human cholangiocarcinoma with a very dismal prognosis. Tumor markers and target drugs of PHCCA are desperately needed. Protein phosphatase N3 (PTPN3) has dual roles in the progression of human cancers, but its expression and functions in PHCCA have not been elucidated. MATERIALS AND METHODS: The expression of PTPN3 in PHCCA was detected with western blotting, qRT-PCR and immunohistochemistry. The clinical significance of PTPN3 was identified by analyzing the correlations between its expression and the clinicopathological variables, and the prognostic value was evaluated by univariate and multivariate analyses. The functions of PTPN3 in the progression of PHCCA were estimated with both in vitro and in vivo experiments. RESULTS: PTPN3 expression was down-regulated in PHCCA compared with normal bile duct. Low PTPN3 expression was markedly associated with large tumor size and unfavorable prognosis. After knocking down PTPN3, the percentages of G2/S phase of PHCCA cells were elevated, and the proliferation increased significantly. Moreover, we demonstrated that the phosphorylation of AKT was elevated by PTPN3 knockdown, and it was required in PTPN3-involved proliferation of PHCCA. Within vivo experiments, PTPN3 and AKT inhibitor MK-2206 were demonstrated to suppress tumor size of PHCCA. CONCLUSION: PTPN3 was a favorable prognostic biomarker of PHCCA. PTPN3 suppressed the proliferation of PHCCA by inhibiting AKT phosphorylation and arresting cell cycle. Our results suggested thatpost-operative detection of PTPN3 would be a helpful approach to stratify the PHCCA patients with high-risk.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Biomarcadores de Tumor/biosíntesis , Proliferación Celular/fisiología , Tumor de Klatskin/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Anciano , Animales , Neoplasias de los Conductos Biliares/diagnóstico , Neoplasias de los Conductos Biliares/genética , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Tumor de Klatskin/diagnóstico , Tumor de Klatskin/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Fosforilación/fisiología , Pronóstico , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , ARN Interferente Pequeño/administración & dosificación , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
EMBO J ; 38(14): e99945, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31304624

RESUMEN

TGF-ß controls a variety of cellular functions during development. Abnormal TGF-ß responses are commonly found in human diseases such as cancer, suggesting that TGF-ß signaling must be tightly regulated. Here, we report that protein tyrosine phosphatase non-receptor 3 (PTPN3) profoundly potentiates TGF-ß signaling independent of its phosphatase activity. PTPN3 stabilizes TGF-ß type I receptor (TßRI) through attenuating the interaction between Smurf2 and TßRI. Consequently, PTPN3 facilitates TGF-ß-induced R-Smad phosphorylation, transcriptional responses, and subsequent physiological responses. Importantly, the leucine-to-arginine substitution at amino acid residue 232 (L232R) of PTPN3, a frequent mutation found in intrahepatic cholangiocarcinoma (ICC), disables its role in enhancing TGF-ß signaling and abolishes its tumor-suppressive function. Our findings have revealed a vital role of PTPN3 in regulating TGF-ß signaling during normal physiology and pathogenesis.


Asunto(s)
Neoplasias Hepáticas/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Trasplante de Neoplasias , Fosforilación , Estabilidad Proteica , Receptor Tipo I de Factor de Crecimiento Transformador beta/química , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Proteínas Smad/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
6.
Biosci Rep ; 39(8)2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31350343

RESUMEN

To investigate the role of microRNA-497-5p (miR-497-5p) in the tumorigenesis of colorectal cancer (CRC), the present study applied qRT-PCR to detect the expression level of miR-497-5p in both clinical samples and CRC cell lines. Furthermore, to specifically evaluate the carcinogenic role of miR-497-5p in CRC, the expression of miR-497-5p was monitored by transfecting with the mimics or inhibitors of miR-497-5p. Transwell assay as well as CCK-8 assay were used to determine the functions of miR-497-5p on cell invasion, migration and proliferation, respectively. miR-497-5p expression was remarkably down-regulated in clinical samples with cancer development as well as in CRC cell lines. Additionally, low miR-497-5p expression was remarkably correlated with higher TNM stage and lymph node metastasis of CRC patients. Up-regulation of miR-497-5p significantly inhibited proliferation, migration, and invasion of LOVO CRC cell line. Conversely, antagonizing miR-497-5p significantly promoted cell proliferation, migration and invasion. Mechanistic analysis revealed that miR-497-5p directly bound to its downstream target, protein tyrosine phosphatase non-receptor type 3 (PTPN3), whose aberrant expression partially reversed inhibition of cell proliferation and migration. Taken together, the present study elucidated the inhibitory role of miR-497-5p in CRC via targeting PTPN3, which potentiated miR-497-5p as a potential therapeutic target for combating CRC.


Asunto(s)
Proliferación Celular/genética , Neoplasias Colorrectales/genética , MicroARNs/genética , Invasividad Neoplásica/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Regulación hacia Arriba/genética , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/genética , Neoplasias Colorrectales/patología , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Células HT29 , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/patología
7.
Sci Rep ; 9(1): 7438, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092861

RESUMEN

The human protein tyrosine phosphatase non-receptor type 3 (PTPN3) is a PDZ (PSD-95/Dlg/ZO-1) domain-containing phosphatase with a tumor-suppressive or a tumor-promoting role in many cancers. Interestingly, the high-risk genital human papillomavirus (HPV) types 16 and 18 target the PDZ domain of PTPN3. The presence of a PDZ binding motif (PBM) on E6 confers interaction with a number of different cellular PDZ domain-containing proteins and is a marker of high oncogenic potential. Here, we report the molecular basis of interaction between the PDZ domain of PTPN3 and the PBM of the HPV E6 protein. We combined biophysical, NMR and X-ray experiments to investigate the structural and functional properties of the PDZ domain of PTPN3. We showed that the C-terminal sequences from viral proteins encompassing a PBM interact with PTPN3-PDZ with similar affinities to the endogenous PTPN3 ligand MAP kinase p38γ. PBM binding stabilizes the PDZ domain of PTPN3. We solved the X-ray structure of the PDZ domain of PTPN3 in complex with the PBM of the HPV E6 protein. The crystal structure and the NMR chemical shift mapping of the PTPN3-PDZ/peptide complex allowed us to pinpoint the main structural determinants of recognition of the C-terminal sequence of the E6 protein and the long-range perturbations induced upon PBM binding.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Ligandos , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Dominios PDZ , Infecciones por Papillomavirus/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Unión Proteica , Estabilidad Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 3/química , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Relación Estructura-Actividad
8.
J Cell Physiol ; 234(2): 1071-1079, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30189107

RESUMEN

Protein Tyrosine Phosphatase H1/Protein Tyrosine Phosphatase Non receptor Type 3 (PTPH1/PTPN3) is upregulated and/or mutated in glioma, ovarian, gastric, and colorectal cancers. Previous studies have documented that PTPH1-associated breast cancers exhibit enhanced sensitivity to tamoxifen and tyrosine kinase inhibitors through dephosphorylation of ER and epidermal growth factor receptor, respectively. Owing to the key role that PTPH1 plays as a biomarker in predicting the response of chemotherapeutic drugs and lack of studies on Indian breast cancer patients, the present study investigated PTPH1 protein expression and its relationship to clinical features, ER/PR/HER2/neu statuses, and methylation of promoter in breast cancer tissues (n = 67) among Indian population by immunohistochemistry and methylation specific polymerase chain reaction. PTPH1 expression was upregulated in 58.21% (39/67) and downregulated in the rest of tumor specimens, and it correlated with ER, PR, and HER2/neu statuses with p values of <0.0001, 0.0113, and 0.0448, respectively. Additionally, we found that the 2 kb region upstream of PTPH1 gene harbored CpG sites within, and was ubiquitously methylated in breast cancer (n = 13), colon cancer tissue (n = 1), uterine cancer tissue (n = 1), normal breast tissue (n = 1) in addition to Hela and MCF7 cell lines. In conclusion, our data showed a strong correlation of the PTPH1 status with the ER and ubiquitous nature of PTPH1 promoter methylation at specific CpG sites irrespective of cancer types and protein expression. Our findings underscore the clinical relevance of PTPH1 expression in Indian patients and warrant additional studies to explore the importance of ubiquitously methylated promoter at specific CpG sites in upstream of the PTPH1 gene.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Islas de CpG , Metilación de ADN , Inmunohistoquímica , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/enzimología , Femenino , Células HeLa , Humanos , India , Células MCF-7 , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Estudios Retrospectivos
9.
Med Sci Monit ; 24: 7556-7562, 2018 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-30348936

RESUMEN

BACKGROUND PTPN3 was demonstrated to be involved in the progression of several types of cancers, such as gastric adenocarcinoma, lung cancer, and intrahepatic cholangiocarcinoma. However, its clinical significance in glioblastoma (GBM) has not been elucidated. MATERIAL AND METHODS We investigated the expression of PTPN3 in 95 cases of GBM with immunohistochemistry and in 8 pairs of fresh GBMs and their adjacent tissues with qualitative polymerase chain reaction. Moreover, the correlation between PTPN3 and clinicopathological factors was evaluated by chi-square test. The prognostic value of PTPN3 was investigated with univariate analysis and multivariate analysis. With MTT assay and Transwell assay, the oncogenic functions of PTPN3 in GBM proliferation and invasion were further investigated. RESULTS Expression of PTPN3 in GBM tissues was significantly higher than in their corresponding adjacent tissues. High expression of PTPN3 was significantly associated with unfavorable prognosis of GBM. Moreover, in GBM cell lines, PTPN3 promoted cell proliferation and invasion, and the PTP common inhibitor pervanadate suppressed GBM proliferation and invasion. CONCLUSIONS Our experiments show that PTPN3 is an independent prognostic factor in GBM and indicated that postoperative detection of PTPN3 can be used to identify high-risk patients and guide individual treatment.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/biosíntesis , Adulto , Anciano , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Femenino , Glioblastoma/genética , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Interferencia de ARN , Transcriptoma
10.
Structure ; 23(4): 653-64, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25728925

RESUMEN

Epidermal growth factor receptor (EGFR) pathway substrate 15 (Eps15) is a newly identified substrate for protein tyrosine phosphatase N3 (PTPN3), which belongs to the FERM-containing PTP subfamily comprising five members including PTPN3, N4, N13, N14, and N21. We solved the crystal structures of the PTPN3-Eps15 phosphopeptide complex and found that His812 of PTPN3 and Pro850 of Eps15 are responsible for the specific interaction between them. We defined the critical role of the additional residue Tyr676 of PTPN3, which is replaced by Ile939 in PTPN14, in recognition of tyrosine phosphorylated Eps15. The WPD loop necessary for catalysis is present in all members but not PTPN21. We identified that Glu instead of Asp in the WPE loop contributes to the catalytic incapability of PTPN21 due to an extended distance beyond protonation targeting a phosphotyrosine substrate. Together with in vivo validations, our results provide novel insights into the substrate specificity and plasticity of FERM-containing PTPs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Simulación del Acoplamiento Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 3/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Línea Celular Tumoral , Células HEK293 , Humanos , Datos de Secuencia Molecular , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Especificidad por Sustrato
11.
Oncogene ; 34(29): 3791-803, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25263444

RESUMEN

Epidermal growth factor receptor (EGFR) regulates multiple signaling cascades essential for cell proliferation, growth and differentiation. Using a genetic approach, we found that Drosophila FERM and PDZ domain-containing protein tyrosine phosphatase, dPtpmeg, negatively regulates border cell migration and inhibits the EGFR/Ras/mitogen-activated protein kinase signaling pathway during wing morphogenesis. We further identified EGFR pathway substrate 15 (Eps15) as a target of dPtpmeg and its human homolog PTPN3. Eps15 is a scaffolding adaptor protein known to be involved in EGFR endocytosis and trafficking. Interestingly, PTPN3-mediated tyrosine dephosphorylation of Eps15 promotes EGFR for lipid raft-mediated endocytosis and lysosomal degradation. PTPN3 and the Eps15 tyrosine phosphorylation-deficient mutant suppress non-small-cell lung cancer cell growth and migration in vitro and reduce lung tumor xenograft growth in vivo. Moreover, depletion of PTPN3 impairs the degradation of EGFR and enhances proliferation and tumorigenicity of lung cancer cells. Taken together, these results indicate that PTPN3 may act as a tumor suppressor in lung cancer through its modulation of EGFR signaling.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Movimiento Celular , Proliferación Celular , Receptores ErbB/metabolismo , Neoplasias Pulmonares/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Modificados Genéticamente , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular , Línea Celular Tumoral , Endocitosis , Femenino , Células HEK293 , Humanos , Immunoblotting , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Microdominios de Membrana/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Confocal , Mutación , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Interferencia de ARN , Trasplante Heterólogo
13.
Gastroenterology ; 146(5): 1397-407, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24503127

RESUMEN

BACKGROUND & AIMS: The pathogenesis of intrahepatic cholangiocarcinoma (ICC), the second most common hepatic cancer, is poorly understood, and the incidence of ICC is increasing worldwide. We searched for mutations in human ICC tumor samples and investigated how they affect ICC cell function. METHODS: We performed whole exome sequencing of 7 pairs of ICC tumors and their surrounding nontumor tissues to detect somatic alterations. We then screened 124 pairs of ICC and nontumor samples for these mutations, including 7 exomes. We compared mutations in PTPN3 with tumor recurrence in 124 patients and PTPN3 expression levels with recurrence in 322 patients (the combination of both in 86 patients). The functional effects of PTPN3 variations were determined by RNA interference and transgenic expression in cholangiocarcinoma cell lines (RBE, HCCC-9810, and Huh28). RESULTS: Based on exome sequencing, pathways that regulate protein phosphorylation were among the most frequently altered in ICC samples and genes encoding protein tyrosine phosphatases (PTPs) were among the most frequently mutated. We identified mutations in 9 genes encoding PTPs in 4 of 7 ICC exomes. In the prevalence screen of 124 paired samples, 51.6% of ICCs contained somatic mutations in at least 1 of 9 PTP genes; 41.1% had mutations in PTPN3. Transgenic expression of PTPN3 in cell lines increased cell proliferation, colony formation, and migration. PTPN3(L232R) and PTPN3(L384H), which were frequently detected in ICC samples, were found to be gain-of-function mutations; their expression in cell lines further increased cell proliferation, colony formation, and migration. ICC-associated variants of PTPN3 altered phosphatase activity. Patients whose tumors contained activating mutations or higher levels of PTPN3 protein than nontumor tissues had higher rates of disease recurrence than patients whose tumors did not have these characteristics. CONCLUSIONS: Using whole exome sequencing of ICC samples from patients, we found that more than 40% contain somatic mutations in PTPN3. Activating mutations in and high expression levels of PTPN3 were associated with tumor recurrence.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/enzimología , Movimiento Celular , Proliferación Celular , Colangiocarcinoma/genética , Neoplasias Hepáticas/genética , Mutación , Recurrencia Local de Neoplasia , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Neoplasias de los Conductos Biliares/enzimología , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/patología , Línea Celular Tumoral , Colangiocarcinoma/enzimología , Colangiocarcinoma/patología , Análisis Mutacional de ADN , Activación Enzimática , Exosomas , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Fenotipo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Interferencia de ARN , Factores de Tiempo , Transfección
14.
Mol Cancer Ther ; 13(1): 230-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24227889

RESUMEN

Estrogen receptor α (ERα or ER) is the only target of breast cancer therapy using antiestrogens. However, about 50% of ER-expressing breast cancer is intrinsically refractory to the antihormone therapy and strategies to improve the therapeutic response are urgently needed. Dynamic ER phosphorylation and dephosphorylation play an important role in ER activity and antihormone response. Although more than 10 kinases participate in phosphorylating ER protein, phosphatases involved remain mostly unidentified. Here, we tested the hypothesis that the protein-tyrosine phosphatase H1 (PTPH1) may regulate ER tyrosine phosphorylation and thereby impact breast cancer antihormone sensitivity. Our results showed that PTPH1 dephosphorylates ER at Tyr537 in vitro and in breast cancer cells. Moreover, PTPH1 stimulates ER nuclear accumulation and increases breast cancer sensitivity to tamoxifen (TAM) and/or fulvestrant in cell culture and in a xenograft model. Further analysis revealed that PTPH1 depends on its catalytic activity to stimulate ER nuclear accumulation and to enhance breast cancer antihormone sensitivity. These studies thus identified PTPH1 as a novel ER phosphatase and further demonstrate a therapeutic potential of enhancing breast cancer sensitivity to antiestrogens through dephosphorylating ER by PTPH1.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Moduladores de los Receptores de Estrógeno/administración & dosificación , Receptor alfa de Estrógeno/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Fosforilación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Tamoxifeno/administración & dosificación , Tirosina/genética
15.
Anim Genet ; 44(2): 217-22, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22812605

RESUMEN

Marek's disease (MD) is a neoplastic disease in chickens, caused by the Marek's disease virus (MDV). To investigate host genetic resistance to MD, we conducted a genome-wide association study (GWAS) on 67 MDV-infected chickens based on a case and control design, including 57 susceptible chickens in the case group and 10 resistant chickens as controls. After searching 38 655 valid genomic markers, two SNPs were found to be associated with host resistance to MD. One SNP, rs14527240, reaching chromosome-wide significance level (P < 0.01) was located in the SPARC-related modular calcium-binding 1 (SMOC1) gene on GGA5. The other one, GGaluGA156129, reaching genome-wide significance (P < 0.05), was located in the protein tyrosine phosphatase, non-receptor type 3 (PTPN3) gene on GGA2. In addition, expression patterns of these two genes in spleens were detected by qPCR. The expression of SMOC1 was significantly up-regulated (P < 0.05), whereas the expression of PTNP3 did not show significance when the case group was compared with the control group. Up-regulation of SMOC1 in susceptible spleens suggests its important roles in MD tumorigenesis. This is the first study to investigate MD-resistant loci, and it demonstrates the power of GWASs for mapping genes associated with MD resistance.


Asunto(s)
Pollos , Resistencia a la Enfermedad/genética , Enfermedad de Marek/genética , Polimorfismo de Nucleótido Simple/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Animales , Estudios de Casos y Controles , Mapeo Cromosómico , Cartilla de ADN/genética , Marcadores Genéticos/genética , Estudio de Asociación del Genoma Completo/veterinaria , Genotipo , Osteonectina/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Organismos Libres de Patógenos Específicos , Bazo/metabolismo
16.
J Biol Chem ; 287(33): 27895-905, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22730326

RESUMEN

Phosphatase plays a crucial role in determining cellular fate by inactivating its substrate kinase, but it is not known whether a kinase can vice versa phosphorylate its phosphatase to execute this function. Protein-tyrosine phosphatase H1 (PTPH1) is a specific phosphatase of p38γ mitogen-activated protein kinase (MAPK) through PDZ binding, and here, we show that p38γ is also a PTPH1 kinase through which it executes its oncogenic activity and regulates stress response. PTPH1 was identified as a substrate of p38γ by unbiased proteomic analysis, and its resultant phosphorylation at Ser-459 occurs in vitro and in vivo through their complex formation. Genetic and pharmacological analyses showed further that Ser-459 phosphorylation is directly regulated by Ras signaling and is important for Ras, p38γ, and PTPH1 oncogenic activity. Moreover, experiments with physiological stimuli revealed a novel stress pathway from p38γ to PTPH1/Ser-459 phosphorylation in regulating cell growth and cell death by a mechanism dependent on cellular environments but independent of canonical MAPK activities. These results thus reveal a new mechanism by which a MAPK regulates Ras oncogenesis and stress response through directly phosphorylating its phosphatase.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Estrés Fisiológico , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Humanos , Ratones , Ratones Noqueados , Proteína Quinasa 12 Activada por Mitógenos/genética , Fosforilación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Proteínas ras/genética
17.
Genes Chromosomes Cancer ; 51(6): 590-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22334442

RESUMEN

An increasing number of chromosomal aberrations is being identified in solid tumors providing novel biomarkers for various types of cancer and new insights into the mechanisms of carcinogenesis. We applied next generation sequencing technique to analyze the transcriptome of the non-small cell lung carcinoma (NSCLC) cell line H2228 and discovered a fusion transcript composed of multiple exons of ALK (anaplastic lymphoma receptor tyrosine kinase) and PTPN3 (protein tyrosine phosphatase, nonreceptor Type 3). Detailed analysis of the genomic structure revealed that a portion of genomic region encompassing Exons 10 and 11 of ALK has been translocated into the intronic region between Exons 2 and 3 of PTPN3. The key net result appears to be the null mutation of one allele of PTPN3, a gene with tumor suppressor activity. Consistently, ectopic expression of PTPN3 in NSCLC cell lines led to inhibition of colony formation. Our study confirms the utility of next generation sequencing as a tool for the discovery of somatic mutations and has led to the identification of a novel mutation in NSCLC that may be of diagnostic, prognostic, and therapeutic importance.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Proteínas de Fusión Oncogénica/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , ARN Neoplásico/genética , Proteínas Tirosina Quinasas Receptoras/genética , Quinasa de Linfoma Anaplásico , Secuencia de Bases , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/diagnóstico , Datos de Secuencia Molecular , ARN Neoplásico/química , Análisis de Secuencia de ADN
18.
Cancer Res ; 70(7): 2901-10, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20332238

RESUMEN

Protein phosphatases are believed to coordinate with kinases to execute biological functions, but examples of such integrated activities, however, are still missing. In this report, we have identified protein tyrosine phosphatase H1 (PTPH1) as a specific phosphatase for p38gamma mitogen-activated protein kinase (MAPK) and shown their cooperative oncogenic activity through direct binding. p38gamma, a Ras effector known to act independent of its phosphorylation, was first shown to require its unique PDZ-binding motif to increase Ras transformation. Yeast two-hybrid screening and in vitro and in vivo analyses further identified PTPH1 as a specific p38gamma phosphatase through PDZ-mediated binding. Additional experiments showed that PTPH1 itself plays a role in Ras-dependent malignant growth in vitro and/or in mice by a mechanism depending on its p38gamma-binding activity. Moreover, Ras increases both p38gamma and PTPH1 protein expression and there is a coupling of increased p38gamma and PTPH1 protein expression in primary colon cancer tissues. These results reveal a coordinative oncogenic activity of a MAPK with its specific phosphatase and suggest that PDZ-mediated p38gamma/PTPH1 complex may be a novel target for Ras-dependent malignancies.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias del Colon/enzimología , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Proteínas ras/metabolismo , Procesos de Crecimiento Celular/fisiología , Transformación Celular Neoplásica/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Genes ras , Células HCT116 , Humanos , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 12 Activada por Mitógenos/biosíntesis , Proteína Quinasa 12 Activada por Mitógenos/genética , Dominios PDZ , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Proteína Tirosina Fosfatasa no Receptora Tipo 3/biosíntesis , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , ARN Interferente Pequeño/genética , Proteínas ras/genética
19.
PLoS One ; 3(12): e4014, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19107198

RESUMEN

PTPN3 and PTPN4 are two closely-related non-receptor protein tyrosine phosphatases (PTP) that, in addition to a PTP domain, contain FERM (Band 4.1, Ezrin, Radixin, and Moesin) and PDZ (PSD-95, Dlg, ZO-1) domains. Both PTP have been implicated as negative-regulators of early signal transduction through the T cell antigen receptor (TCR), acting to dephosphorylate the TCRzeta chain, a component of the TCR complex. Previously, we reported upon the production and characterization of PTPN3-deficient mice which show normal TCR signal transduction and T cell function. To address if the lack of a T cell phenotype in PTPN3-deficient mice can be explained by functional redundancy of PTPN3 with PTPN4, we generated PTPN4-deficient and PTPN4/PTPN3 double-deficient mice. As in PTPN3 mutants, T cell development and homeostasis and TCR-induced cytokine synthesis and proliferation were found to be normal in PTPN4-deficient and PTPN4/PTPN3 double-deficient mice. PTPN13 is another FERM and PDZ domain-containing non-receptor PTP that is distantly-related to PTPN3 and PTPN4 and which has been shown to function as a negative-regulator of T helper-1 (Th1) and Th2 differentiation. Therefore, to determine if PTPN13 might compensate for the loss of PTPN3 and PTPN4 in T cells, we generated mice that lack functional forms of all three PTP. T cells from triple-mutant mice developed normally and showed normal cytokine secretion and proliferative responses to TCR stimulation. Furthermore, T cell differentiation along the Th1, Th2 and Th17 lineages was largely unaffected in triple-mutants. We conclude that PTPN3 and PTPN4 are dispensable for TCR signal transduction.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 3/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 4/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 4/genética , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/fisiología
20.
J Biomed Sci ; 14(6): 731-44, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17588219

RESUMEN

Protein-tyrosine phosphatase PTPN3 is a membrane-associated non-receptor protein-tyrosine phosphatase. PTPN3 contains a N-terminal FERM domain, a middle PDZ domain, and a C-terminal phosphatase domain. Upon co-expression of PTPN3, the level of human hepatitis B viral (HBV) RNAs, 3.5 kb, 2.4/2.1 kb, and 0.7 kb transcribed from a replicating HBV expression plasmid is significantly reduced in human hepatoma HuH-7 cells. When the expression of endogenous PTPN3 protein is diminished by specific small interfering RNA, the expression of HBV genes is enhanced, indicating that the endogenous PTPN3 indeed plays a suppressive role on HBV gene expression. PTPN3 can interact with HBV core protein. The interaction is mediated via the PDZ domain of PTPN3 and the carboxyl-terminal last four amino acids of core. Either deletion of PDZ domain of PTPN3 or substitution of PDZ ligand in core has no effect on PTPN3-mediated suppression. These results clearly show that the interaction of PTPN3 with core is not required for PTPN3 suppressive effect. Mutation of (359)serine and (835)serine of 14-3-3beta binding sites to alanine, which slightly reduces the interaction with 14-3-3beta, does not influence the PTPN3 effect. In contrast, mutation of the invariant (842)cysteine residue in phosphatase domain to serine, which makes the phosphatase activity inactive, does not change its subcellular localization and interaction with core or 14-3-3beta, but completely abolishes PTPN3-mediated suppression. Furthermore, deletion of FERM domain does not affect the phosphatase activity or interaction with 14-3-3beta, but changes the subcellular localization from cytoskeleton-membrane interface to cytoplasm and nucleus, abolishes binding to core, and diminishes the PTPN3 effect on HBV gene expression. Taken together, these results demonstrate that the phosphatase activity and FERM domain of PTPN3 are essential for its suppression of HBV gene expression.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Carcinoma Hepatocelular/metabolismo , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Silenciador del Gen , Genes Virales , Virus de la Hepatitis B/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 3/análisis , Proteína Tirosina Fosfatasa no Receptora Tipo 3/genética , Supresión Genética , Transfección , Células Tumorales Cultivadas , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA