Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Immunother ; 47(3): 89-97, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38297883

RESUMEN

In the tumor microenvironment, wherein cytotoxic lymphocytes interact with cancer cells, lymphocyte exhaustion, an immune checkpoint inhibitor target, is promoted. However, the efficacy of these inhibitors is limited, and improving response rates remains challenging. We previously reported that protein tyrosine phosphatase nonreceptor type (PTPN) 3 is a potential immune checkpoint molecule for activated lymphocytes and that PTPN3 inhibition should be a focus area for cancer immunotherapy development. Therefore, in this study, we focused on PTPN3-suppressive therapy in terms of lymphocyte exhaustion under hypoxic conditions, which are a cancer microenvironment, and investigated measures for improving the response to anti-programmed death receptor (PD)-1 antibody drugs. We found that PTPN3 expression was upregulated in activated lymphocytes under hypoxic conditions, similar to the findings for other immune checkpoint molecules, such as PD-1, T cell immunoglobulin mucin-3, and lymphocyte-activation gene-3; furthermore, it functioned as a lymphocyte exhaustion marker. In addition, PTPN3-suppressed activated lymphocytes promoted the mammalian target of rapamycin (mTOR)-Akt signaling pathway activation and enhanced proliferation, migration, and cytotoxic activities under hypoxic conditions. Furthermore, PTPN3 suppression in activated lymphocytes increased PD-1 expression and enhanced the antitumor effects of anti-PD-1 antibody drugs against head and neck cancer in vitro and in vivo. These results suggest that the suppression of PTPN3 expression in activated lymphocytes enhances the therapeutic effect of anti-PD-1 antibody drugs in head and neck cancer, especially under hypoxic conditions that cause lymphocyte exhaustion.


Asunto(s)
Antineoplásicos , Neoplasias de Cabeza y Cuello , Humanos , Receptor de Muerte Celular Programada 1 , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Linfocitos/metabolismo , Inmunoterapia , Microambiente Tumoral , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo
2.
J Cancer Res Clin Oncol ; 149(16): 14619-14630, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37584709

RESUMEN

PURPOSE: In a previous study, protein tyrosine phosphatase non-receptor type (PTPN) 3 was identified as an immune checkpoint molecule in lymphocytes, and its potential as a novel target for cancer immunotherapy was anticipated. However, evaluation of dendritic cell (DC) function as antigen-presenting cells is critical for the development of immunotherapy. In this study, we aimed to analyze the biological effect of PTPN3 on DCs induced from human peripheral blood monocytes obtained from healthy individuals. METHODS: We used short-interfering RNA to knock down PTP3 in DCs. For DC maturation, we added cancer cell lysate and tumor necrosis factor-α/interferon-α to immature DCs. In the cytotoxic assay, the target cancer cells were SBC5, unmatched with DCs from healthy human leukocyte antigen (HLA)-A24, or Sq-1, matched with DCs. Enzyme-linked immunosorbent assay was used to determine the amount of cytokines. To examine the intracellular signaling system, intracellular staining was used. RESULTS: PTPN3 knockdown significantly increased the number of DCs, expression of CD80 and chemokine receptor (CCR)7, and production of interleukin-12p40/p70 in mature DCs. In the HLA-A24-restricted DC and human lung squamous cell carcinoma cell cytotoxic assay, inhibition of PTPN3 expression in mature DCs induced cytotoxic T lymphocytes with increased production of INF-γ and granzyme B, and enhanced toxicity against cancer cells and migration to cancer. Furthermore, inhibition of PTPN3 expression activated the mitogen-activated protein kinase pathway in DCs. CONCLUSION: Based on our findings, inhibition of PTPN3 expression could contribute to the development of novel cancer immunotherapies that activate not only lymphocytes but also DCs.


Asunto(s)
Células Dendríticas , Neoplasias , Humanos , Citocinas/metabolismo , Linfocitos T Citotóxicos , Interleucinas , Neoplasias/metabolismo , Inmunoterapia , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo
3.
Anticancer Res ; 42(6): 2869-2874, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35641270

RESUMEN

BACKGROUND/AIM: Recently, protein tyrosine phosphatase non-receptor type 3 (PTPN3) has gained attention. However, the role of PTPN3 in cancer has not been fully elucidated. In the present study, we analyzed the role of PTPN3 in pancreatic cancer and investigated whether PTPN3 could be a new therapeutic target for pancreatic cancer. MATERIALS AND METHODS: Two pancreatic ductal adenocarcinoma (PDAC) cell lines were used as target cells. Cell proliferation was investigated using cell counting and a xenograft mouse model. Migration and invasion were analyzed using Transwell inserts. Activation-related signaling molecules were examined by western blotting. RESULTS: PTPN3 contributes to the proliferation, migration, and invasion of PDAC cells in vitro. PTPN3 promotes tumor growth in a mouse xenograft model, an action mediated partially through the MAPK pathway. CONCLUSION: PTPN3 could be a new therapeutic target for pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Proteína Tirosina Fosfatasa no Receptora Tipo 3 , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Humanos , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Neoplasias Pancreáticas
4.
Sci Rep ; 11(1): 944, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441627

RESUMEN

Interactions between the hepatitis B virus core protein (HBc) and host cell proteins are poorly understood, although they may be essential for the propagation of the virus and its pathogenicity. HBc has a C-terminal PDZ (PSD-95, Dlg1, ZO-1)-binding motif (PBM) that is responsible for interactions with host PDZ domain-containing proteins. In this work, we focused on the human protein tyrosine phosphatase non-receptor type 3 (PTPN3) and its interaction with HBc. We solved the crystal structure of the PDZ domain of PTPN3 in complex with the PBM of HBc, revealing a network of interactions specific to class I PDZ domains despite the presence of a C-terminal cysteine in this atypical PBM. We further showed that PTPN3 binds the HBc protein within capsids or as a homodimer. We demonstrate that overexpression of PTPN3 significantly affects HBV infection in HepG2 NTCP cells. Finally, we performed proteomics studies on both sides by pull-down assays and screening of a human PDZ domain library. We identified a pool of human PBM-containing proteins that might interact with PTPN3 in cells and that could be in competition with the HBc PBM during infection, and we also identified potential cellular partners of HBc through PDZ-PBM interactions. This study opens up many avenues of future investigations into the pathophysiology of HBV.


Asunto(s)
Antígenos del Núcleo de la Hepatitis B/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/ultraestructura , Cápside/metabolismo , Hepatitis B/metabolismo , Hepatitis B/virología , Antígenos del Núcleo de la Hepatitis B/ultraestructura , Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis B/patogenicidad , Virus de la Hepatitis B/fisiología , Humanos , Dominios PDZ/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 3/química , Proteína Tirosina Fosfatasa no Receptora Tipo 3/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Tirosina/metabolismo , Proteínas del Núcleo Viral/metabolismo
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Cells ; 8(3)2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30875834

RESUMEN

Human papillomaviruses (HPV) replicate their DNA in the suprabasal layer of the infected mucosa or skin. In order to create a suitable environment for vegetative viral DNA replication HPV delay differentiation and sustain keratinocyte proliferation that can lead to hyperplasia. The mechanism underlying cell growth stimulation is not well characterized. Here, we show that the E6 oncoprotein of the ßHPV type 8 (HPV8), which infects the cutaneous skin and is associated with skin cancer in Epidermodysplasia verruciformis patients and immunosuppressed organ transplant recipients, binds to the protein tyrosine phosphatase H1 (PTPH1), which resulted in increased protein expression and phosphatase activity of PTPH1. Suppression of PTPH1 in immortalized keratinocytes reduced cell proliferation as well as the level of epidermal growth factor receptor (EGFR). Furthermore, we report that HPV8E6 expressing keratinocytes have increased level of active, GTP-bound Ras. This effect was independent of PTPH1. Therefore, HPV8E6-mediated targeting of PTPH1 might result in higher level of EGFR and enhanced keratinocyte proliferation. The HPV8E6-mediated stimulation of Ras may be an additional step to induce cell growth. Our results provide novel insights into the mechanism how ßHPVE6 proteins support proliferation of infected keratinocytes, thus creating an environment with increased risk of development of skin cancer particularly upon UV-induced DNA mutations.


Asunto(s)
Queratinocitos/citología , Queratinocitos/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Oncogenes , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Proliferación Celular , Receptores ErbB/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Rayos Ultravioleta , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas ras/metabolismo
11.
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
12.
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
13.
Oncotarget ; 6(15): 13320-33, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-26079946

RESUMEN

Protein-protein interactions can increase or decrease its therapeutic target activity and the determining factors involved, however, are largely unknown. Here, we report that tyrosine-dephosphorylation of epidermal growth factor receptor (EGFR) increases its therapeutic target activity by disrupting its interaction with estrogen receptor (ER). Protein tyrosine phosphatase H1 (PTPH1) dephosphorylates the tyrosine kinase EGFR, disrupts its interaction with the nuclear receptor ER, and increases breast cancer sensitivity to small molecule tyrosine kinase inhibitors (TKIs). These effects require PTPH1 catalytic activity and its interaction with EGFR, suggesting that the phosphatase may increase the sensitivity by dephosphorylating EGFR leading to its dissociation with ER. Consistent with this notion, a nuclear-localization defective ER has a higher EGFR-binding activity and confers the resistance to TKI-induced growth inhibition. Additional analysis show that PTPH1 stabilizes EGFR, stimulates the membranous EGFR accumulation, and enhances the growth-inhibitory activity of a combination therapy of TKIs with an anti-estrogen. Since EGFR and ER both are substrates for PTPH1 in vitro and in intact cells, these results indicate that an inhibitory EGFR-ER protein complex can be switched off through a competitive enzyme-substrate binding. Our results would have important implications for the treatment of breast cancer with targeted therapeutics.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores ErbB/metabolismo , Receptores de Estrógenos/metabolismo , Tirosina/metabolismo , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Humanos , Fosforilación , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
J Gen Virol ; 88(Pt 11): 2956-2965, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17947517

RESUMEN

The E6 proteins of high-risk genital human papillomaviruses (HPV), such as HPV types 16 and 18, possess a conserved C-terminal PDZ-binding motif, which mediates interaction with some cellular PDZ domain proteins. The binding of E6 usually results in their ubiquitin-mediated degradation. The ability of E6 to bind to PDZ domain proteins correlates with the oncogenic potential. Using a yeast two-hybrid system, GST pull-down experiments and coimmunoprecipitations, we identified the protein tyrosine phosphatase H1 (PTPH1/PTPN3) as a novel target of the PDZ-binding motif of E6 of HPV16 and 18. PTPH1 has been suggested to function as tumour suppressor protein, since mutational analysis revealed somatic mutations in PTPH1 in a minor fraction of various human tumours. We show here that HPV16 E6 accelerated the proteasome-mediated degradation of PTPH1, which required the binding of E6 to the cellular ubiquitin ligase E6-AP and to PTPH1. The endogenous levels of PTPH1 were particularly low in HPV-positive cervical carcinoma cell lines. The reintroduction of the E2 protein into the HPV16-positive cervical carcinoma cell line SiHa, known to lead to a sharp repression of E6 expression and to induce growth suppression, resulted in an increase of the amount of PTPH1. Our data suggest that reducing the level of PTPH1 may contribute to the oncogenic activity of high-risk genital E6 proteins.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Papillomavirus Humano 16/fisiología , Papillomavirus Humano 18/fisiología , Proteínas Oncogénicas Virales/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 3/metabolismo , Proteínas Represoras/metabolismo , Línea Celular , Inmunoprecipitación de Cromatina , Humanos , Unión Proteica , Técnicas del Sistema de Dos Híbridos , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...