Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 144(5): 703-18, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21376233

RESUMEN

Among breast cancers, triple-negative breast cancer (TNBC) is the most poorly understood and is refractory to current targeted therapies. Using a genetic screen, we identify the PTPN12 tyrosine phosphatase as a tumor suppressor in TNBC. PTPN12 potently suppresses mammary epithelial cell proliferation and transformation. PTPN12 is frequently compromised in human TNBCs, and we identify an upstream tumor-suppressor network that posttranscriptionally controls PTPN12. PTPN12 suppresses transformation by interacting with and inhibiting multiple oncogenic tyrosine kinases, including HER2 and EGFR. The tumorigenic and metastatic potential of PTPN12-deficient TNBC cells is severely impaired upon restoration of PTPN12 function or combined inhibition of PTPN12-regulated tyrosine kinases, suggesting that TNBCs are dependent on the proto-oncogenic tyrosine kinases constrained by PTPN12. Collectively, these data identify PTPN12 as a commonly inactivated tumor suppressor and provide a rationale for combinatorially targeting proto-oncogenic tyrosine kinases in TNBC and other cancers based on their profile of tyrosine-phosphatase activity.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Transformación Celular Neoplásica , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , MicroARNs/metabolismo , Mutación , Metástasis de la Neoplasia , Procesamiento Proteico-Postraduccional
2.
PLoS Pathog ; 18(10): e1010640, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36191034

RESUMEN

Colonization of host phagocytic cells by Leishmania metacyclic promastigotes involves several parasite effectors, including the zinc-dependent metalloprotease GP63. The major mode of action of this virulence factor entails the cleavage/degradation of host cell proteins. Given the potent proteolytic activity of GP63, identification of its substrates requires the adequate preparation of cell lysates to prevent artefactual degradation during cell processing. In the present study, we re-examined the cleavage/degradation of reported GP63 substrates when GP63 activity was efficiently neutralized during the preparation of cell lysates. To this end, we infected bone marrow-derived macrophages with either wild type, Δgp63, and Δgp63+GP63 L. major metacyclic promastigotes for various time points. We prepared cell lysates in the absence or presence of the zinc-metalloprotease inhibitor 1,10-phenanthroline and examined the levels and integrity of ten previously reported host cell GP63 substrates. Inhibition of GP63 activity with 1,10-phenanthroline during the processing of macrophages prevented the cleavage/degradation of several previously described GP63 targets, including PTP-PEST, mTOR, p65RelA, c-Jun, VAMP3, and NLRP3. Conversely, we confirmed that SHP-1, Synaptotagmin XI, VAMP8, and Syntaxin-5 are bona fide GP63 substrates. These results point to the importance of efficiently inhibiting GP63 activity during the preparation of Leishmania-infected host cell lysates. In addition, our results indicate that the role of GP63 in Leishmania pathogenesis must be re-evaluated.


Asunto(s)
Leishmania , Proteína Tirosina Fosfatasa no Receptora Tipo 12 , Leishmania/metabolismo , Metaloendopeptidasas/metabolismo , Metaloproteasas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteínas Qa-SNARE/metabolismo , Sinaptotagminas , Serina-Treonina Quinasas TOR/metabolismo , Proteína 3 de Membrana Asociada a Vesículas/metabolismo , Factores de Virulencia , Zinc/metabolismo
3.
Proteins ; 91(6): 831-846, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36645312

RESUMEN

The AMP-activated protein kinase (AMPK) is known to be activated by the protein tyrosine phosphatase non-receptor type 12 (PTP-PEST) under hypoxic conditions. This activation is mediated by tyrosine dephosphorylation of the AMPKα subunit. However, the identity of the phosphotyrosine residues that PTP-PEST dephosphorylates remains unknown. In this study, we first predicted the structure of the complex of the AMPKα2 subunit and PTP-PEST catalytic domain using bioinformatics tools and further confirmed the stability of the complex using molecular dynamics simulations. Evaluation of the protein-protein interfaces indicated that residue Tyr232 is the most likely dephosphorylation site on AMPKα2. In addition, we explored the effect of phosphorylation of PTP-PEST residue Tyr64 on the stability of the complex. Phosphorylation of the highly conserved Tyr64, an interface residue, enhances the stability of the complex via the rearrangement of a network of electrostatic interactions in conjunction with conformational changes in the catalytic WPD loop. We generated a phosphomimetic (PTP-PEST-Y64D) mutant and used co-immunoprecipitation to study the effect of PTP-PEST phosphorylation on AMPKα2 binding. The mutant exhibited an increased affinity for AMPKα2 and corroborated the in-silico predictions. Together, our findings present a plausible structural basis of AMPK regulation by PTP-PEST and show how phosphorylation of PTP-PEST affects its interaction with AMPKα2.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Proteína Tirosina Fosfatasa no Receptora Tipo 12 , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Tirosina Fosfatasas/química , Fosforilación , Dominio Catalítico
4.
Exp Cell Res ; 416(2): 113130, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35364057

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor worldwide. Long noncoding RNAs (lncRNAs) exhibit a regulatory role in the progression of ESCC. Our research was performed to investigate the potential molecular mechanism of lncRNA GATA2-AS1 in ESCC. METHODS: The expression of GATA2-AS1 was identified by qRT-PCR. Cell function assays explored the potential effect of GATA2-AS1 on ESCC progression. The subcellular hierarchical localization method was executed to identify the subcellular localization of GATA2-AS1 in ESCC cells. A prediction website was utilized to discover the relationships among GATA2-AS1, miR-940 and PTPN12. Dual luciferase reporter gene, pull-down assays and RIP assays were executed to verify the binding activity among GATA2-AS1, miR-940 and PTPN12. Xenograft tumor experiments were performed to evaluate ESCC cell growth in vivo. RESULTS: The expression of GATA2-AS1 and PTPN12 was reduced, while miR-940 expression was enhanced in ESCC tissues and cell lines. In vivo experiments showed that GATA2-AS1 inhibited the progression of ESCC cells toward malignancy. Bioinformatics analysis, dual luciferase and RIP assays revealed that GATA2-AS1 upregulated PTPN12 expression by competitively targeting miR-940. miR-940 reversed the inhibitory effect of GATA2-AS1 on the biological behavior of ESCC cells. CONCLUSION: Our findings suggested that GATA2-AS1, expressed at low levels in ESCC, plays a crucial role in the progression of ESCC by targeting the miR-940/PTPN12 axis and could be a potential drug target to treat ESCC patients.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , MicroARNs , Proteína Tirosina Fosfatasa no Receptora Tipo 12 , ARN Largo no Codificante , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
5.
J Orthop Sci ; 28(2): 468-475, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35063332

RESUMEN

BACKGROUND: Protein tyrosine phosphatase non-receptor 12 (PTPN12) plays a prominent role in various cancers as a tumor suppressor. However, the expression of PTPN12 and its biological functions in osteosarcoma (OS) remains to be determined. METHODS: PTPN12 expression in OS was explored in public databases and detected by immunohistochemistry and Western blot. The cell viability was determined by Cell Counting Kit-8 (CCK-8) assay and colony formation. The cell migration and invasion were assessed by the Transwell assay. Flow cytometry analysis was applied to detect cell apoptosis and cell cycle distribution. To investigate the related mechanism, the levels of EGFR and downstream proteins were detected by Western blot. RESULTS: PTPN12 expression was significantly decreased in OS samples in GEO database and our hospital. OS cell lines in Cancer Cell Line Encyclopedia (CCLE) database and our cultured OS cells also demonstrated low PTPN12 expression. Lentivirus-induced overexpression of PTPN12 significantly inhibited the cell viability, migration and invasion of 143B and U2OS cells. The results of flow cytometry found that PTPN12 overexpression promoted cell apoptosis and induced cell cycle arrest at G1 phase in 143B and U2OS cells. The phosphorylation levels of EGFR and subsequent proteins of the PI3K/AKT and ERK pathways were inactivated as a result of PTPN12 overexpression in OS. CONCLUSION: PTPN12 plays a tumor suppressive role in OS cells. Restoring of PTPN12 activity may provide new insights for the treatment of this disease.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Apoptosis , Osteosarcoma/patología , Neoplasias Óseas/genética , Receptores ErbB/metabolismo , Proliferación Celular , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo
6.
Biophys J ; 117(8): 1467-1475, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31542223

RESUMEN

Mitral valve diseases affect ∼3% of the population and are the most common reasons for valvular surgery because no drug-based treatments exist. Inheritable genetic mutations have now been established as the cause of mitral valve insufficiency, and four different missense mutations in the filamin A gene (FLNA) have been found in patients suffering from nonsyndromic mitral valve dysplasia (MVD). The filamin A (FLNA) protein is expressed, in particular, in endocardial endothelia during fetal valve morphogenesis and is key in cardiac development. The FLNA-MVD-causing mutations are clustered in the N-terminal region of FLNA. How the mutations in FLNA modify its structure and function has mostly remained elusive. In this study, using NMR spectroscopy and interaction assays, we investigated FLNA-MVD-causing V711D and H743P mutations. Our results clearly indicated that both mutations almost completely destroyed the folding of the FLNA5 domain, where the mutation is located, and also affect the folding of the neighboring FLNA4 domain. The structure of the neighboring FLNA6 domain was not affected by the mutations. These mutations also completely abolish FLNA's interactions with protein tyrosine phosphatase nonreceptor type 12, which has been suggested to contribute to the pathogenesis of FLNA-MVD. Taken together, our results provide an essential structural and molecular framework for understanding the molecular bases of FLNA-MVD, which is crucial for the development of new therapies to replace surgery.


Asunto(s)
Filaminas/química , Prolapso de la Válvula Mitral/genética , Mutación Missense , Pliegue de Proteína , Sitios de Unión , Filaminas/genética , Filaminas/metabolismo , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo
7.
BMC Cancer ; 19(1): 944, 2019 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-31606028

RESUMEN

BACKGROUND: Protein tyrosine phosphatase non-receptor 12 (PTPN12) is ubiquitously tyrosine phosphatase with tumor suppressive properties. METHODS: PTPN12 expression was analyzed by immunohistochemistry on a tissue microarray with 13,660 clinical prostate cancer specimens. RESULTS: PTPN12 staining was typically absent or weak in normal prostatic epithelium but seen in the majority of cancers, where staining was considered weak in 26.5%, moderate in 39.9%, and strong in 4.7%. High PTPN12 staining was associated with high pT category, high classical and quantitative Gleason grade, lymph node metastasis, positive surgical margin, high Ki67 labeling index and early prostate specific antigen recurrence (p < 0.0001 each). PTPN12 staining was seen in 86.4% of TMPRSS2:ERG fusion positive but in only 58.4% of ERG negative cancers. Subset analyses discovered that all associations with unfavorable phenotype and prognosis were markedly stronger in ERG positive than in ERG negative cancers but still retained in the latter group. Multivariate analyses revealed an independent prognostic impact of high PTPN12 expression in all cancers and in the ERG negative subgroup and to a lesser extent also in ERG positive cancers. Comparison with 12 previously analyzed chromosomal deletions revealed that high PTPN12 expression was significantly associated with 10 of 12 deletions in ERG negative and with 7 of 12 deletions in ERG positive cancers (p < 0.05 each) indicating that PTPN12 overexpression parallels increased genomic instability in prostate cancer. CONCLUSIONS: These data identify PTPN12 as an independent prognostic marker in prostate cancer. PTPN12 analysis, either alone or in combination with other biomarkers might be of clinical utility in assessing prostate cancer aggressiveness.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Anciano , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Deleción Cromosómica , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Análisis Multivariante , Clasificación del Tumor , Proteínas de Fusión Oncogénica/metabolismo , Células PC-3 , Pronóstico , Prostatectomía , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Receptor ErbB-2/metabolismo , Análisis de Matrices Tisulares , Regulador Transcripcional ERG/metabolismo
8.
BMC Cancer ; 19(1): 451, 2019 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-31088422

RESUMEN

BACKGROUND: In a time of increasing concerns over personalized and precision treatment in breast cancer (BC), filtering prognostic factors attracts more attention. Apoptosis-Inducing Factor Mitochondrion-associated 3 (AIFM3) is widely expressed in various tissues and aberrantly expressed in several cancers. However, clinical implication of AIFM3 has not been reported in BC. The aim of the study is to investigate the crystal structure, clinical and prognostic implications of AIFM3 in BC. METHODS: AIFM3 expression in 151 BC samples were assessed by immunohistochemistry (IHC). The Cancer Genome Atlas (TCGA) and Kaplan-Meier survival analysis were used to demonstrate expression and survival of AIFM3 signature. Gene Set Enrichment Analysis (GSEA) was performed to investigate the mechanisms related to AIFM3 expression in BC. RESULTS: AIFM3 was significantly more expressed in breast cancer tissues than in normal tissues. AIFM3 expression had a significant association with tumor size, lymph node metastasis, TNM stage and molecular typing. Higher AIFM3 expression was related to a shorter overall survival (OS) and disease-free survival (DFS). Lymph node metastasis and TNM stage were independent factors of AIFM3 expression. The study presented the crystal structure of AIFM3 successfully and predicted several binding sites when AIFM3 bonded to PTPN12 by Molecular Operating Environment software (MOE). CONCLUSIONS: AIFM3 might be a potential biomarker for predicting prognosis in BC, adding to growing evidence that AIFM3 might interact with PTPN12.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , Neoplasias de la Mama/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Regulación hacia Arriba , Adulto , Anciano , Anciano de 80 o más Años , Factor Inductor de la Apoptosis/química , Sitios de Unión , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática , Persona de Mediana Edad , Proteínas Mitocondriales/química , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estadificación de Neoplasias , Pronóstico , Carga Tumoral
9.
FASEB J ; 32(1): 73-82, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28842430

RESUMEN

The protein tyrosine phosphatase nonreceptor type 12 (PTPN12) is a multifunctional protein and has elicited much research attention because its decreased protein level has been associated with poor prognosis of several types of cancers. Recently, we have solved the crystal structure of the phosphatase domain of PTPN12, which disclosed a specific PTPN12-insert-loop harboring a cyclin-dependent kinase 2 (CDK2) phosphorylation site. However, the functional significance of this phosphorylation is undefined. In the present study, we found that S19 site phosphorylation of PTPN12 by CDK2 discharged its antitumor activity by down-regulation of its inhibitory role in cell migration, but not affecting its other regulatory functions. Phosphorylation of PTPN12 at the S19 site changed its substrate interface, and by doing so, selectively decreased its activity toward the human epidermal growth factor receptor 2 (HER2)- pY1196 site, but not other HER2 phosphorylation sites or other known PTPN12 substrates. A further in-depth mechanism study revealed that the phosphorylation of PTPN12 by CDK2 impaired recruitment of the serine/threonine-protein kinase 1 (PAK1) to HER2, resulted in the blockade of the HER2-pY1196-PAK1-T423 signaling pathway, thus increased tumor cell motility. Taken together, our results identified a new phosphorylation-based substrate recognition mechanism of PTPN12 by CDK2, which orchestrated signaling crosstalk between the oncogenic CDK2 and HER2 pathways. The newly identified governing mechanism of the substrate selectivity of a particular phosphatase was previously unappreciated and exemplifies how a phospho-network is precisely controlled in different cellular contexts.-Li, H., Yang, D., Ning, S., Xu, Y., Yang, F., Yin, R., Feng, T., Han, S., Guo, L., Zhang, P., Qu, W., Guo, R., Song, C., Xiao, P., Zhou, C., Xu, Z., Sun, J.-P., Yu, X. Switching of the substrate specificity of protein tyrosine phosphatase N12 by cyclin-dependent kinase 2 phosphorylation orchestrating 2 oncogenic pathways.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Sitios de Unión , Neoplasias de la Mama/metabolismo , Carcinogénesis/metabolismo , Línea Celular Tumoral , Movimiento Celular , Factor de Crecimiento Epidérmico , Femenino , Humanos , Modelos Biológicos , Modelos Moleculares , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Proteína Tirosina Fosfatasa no Receptora Tipo 12/química , Receptor ErbB-2/metabolismo , Transducción de Señal , Especificidad por Sustrato , Quinasas p21 Activadas/metabolismo
10.
Immunity ; 33(2): 167-80, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20727793

RESUMEN

PTP-PEST (encoded by Ptpn12) is an intracellular protein tyrosine phosphatase belonging to the same family as LYP. LYP inhibits secondary T cell responses by suppressing Src family protein tyrosine kinases and is implicated in human autoimmunity. To determine the function of PTP-PEST in T cells, we generated mice with a conditionally deleted allele of Ptpn12. By removing PTP-PEST in T cells, we determined that PTP-PEST was not necessary for T cell development or primary responses. However, PTP-PEST was required for secondary T cell responses, anergy prevention, and autoimmunity induction. PTP-PEST specifically regulated the phosphorylation of Pyk2, a substrate of the Src family kinase Fyn. It also promoted the formation of T cell homoaggregates, which are known to enhance T cell activation. Thus, PTP-PEST controls Pyk2 activity and is a positive regulator of secondary T cell activation. These data illustrate the critical role of protein tyrosine phosphatases in T cell regulation.


Asunto(s)
Quinasa 2 de Adhesión Focal/inmunología , Quinasa 2 de Adhesión Focal/metabolismo , Activación de Linfocitos , Proteína Tirosina Fosfatasa no Receptora Tipo 12/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Linfocitos T/inmunología , Animales , Agregación Celular , Proliferación Celular , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 12/deficiencia , Linfocitos T/citología , Linfocitos T/enzimología , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
11.
Protein Expr Purif ; 142: 45-52, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28965803

RESUMEN

Protein tyrosine phosphatase non-receptor type 12 (PTPN12), also known as PTP-PEST, was broadly expressed in hemopoietic cells. Recent research has shown that this enzyme is involved in tumorigenesis, as well as in tumor progression and transfer, as it can suppress multiple oncogenic tyrosine kinases. However, the difficulty of soluble expression of PTP-PEST in prokaryotic cells has resulted in great limitations in investigating its structure and functions. In this study, we successfully carried out soluble expression of the catalytic domain of PTP-PEST (ΔPTP-PEST) in Escherichia coli and performed an enzymatic characterization and kinetics. To confirm expression efficiency, we also induced the expression of the chaperon, FKBP_C. FKBP_C expression indicated efficacious prokaryotic expression of ΔPTP-PEST. In conclusion, our work yielded a practical expression system and two-step chromatography purification method that may serve as a valuable tool for the structural and functional analysis of proteins that are difficult to express in the soluble form in prokaryotic cells.


Asunto(s)
Proteínas Arqueales/genética , Chaperonas Moleculares/genética , Isomerasa de Peptidilprolil/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Proteínas de Unión a Tacrolimus/genética , Thermococcus/química , Proteínas Arqueales/metabolismo , Dominio Catalítico , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Cinética , Chaperonas Moleculares/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/aislamiento & purificación , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Thermococcus/metabolismo
12.
Mol Cell ; 35(1): 11-25, 2009 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-19595712

RESUMEN

Activated Ras has been found in many types of cancer. However, the mechanism underlying Ras-promoted tumor metastasis remains unclear. We demonstrate here that activated Ras induces tyrosine dephosphorylation and inhibition of FAK mediated by the Ras downstream Fgd1-Cdc42-PAK1-MEK-ERK signaling cascade. ERK phosphorylates FAK S910 and recruits PIN1 and PTP-PEST, which colocalize with FAK at the lamellipodia of migrating cells. PIN1 binding and prolyl isomerization of FAK cause PTP-PEST to interact with and dephosphorylate FAK Y397. Inhibition of FAK mediated by this signal relay promotes Ras-induced cell migration, invasion, and metastasis. These findings uncover the importance of sequential modification of FAK-by serine phosphorylation, isomerization, and tyrosine dephosphorylation--in the regulation of FAK activity and, thereby, in Ras-related tumor metastasis.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteínas ras/metabolismo , Animales , Sitios de Unión , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Immunoblotting , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/genética , Células 3T3 NIH , Peptidilprolil Isomerasa de Interacción con NIMA , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Isomerasa de Peptidilprolil/genética , Fosforilación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Serina/metabolismo , Transfección , Tirosina/metabolismo , Proteínas ras/genética
13.
Int J Mol Sci ; 19(1)2017 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-29278368

RESUMEN

Protein tyrosine phosphatase non-receptor 12 (PTPN12) is an important protein tyrosine phosphatase involved in regulating cell adhesion and migration as well as tumorigenesis. Here, we solved a crystal structure of the native PTPN12 catalytic domain with the catalytic cysteine (residue 231) in dual conformation (phosphorylated and unphosphorylated). Combined with molecular dynamics simulation data, we concluded that those two conformations represent different states of the protein which are realized during the dephosphorylation reaction. Together with docking and mutagenesis data, our results provide a molecular basis for understanding the catalytic mechanism of PTPN12 and its role in tumorigenesis.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 12/química , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Simulación de Dinámica Molecular , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Conformación Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Especificidad por Sustrato
14.
BMC Cell Biol ; 17(1): 31, 2016 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-27542844

RESUMEN

BACKGROUND: The serine/threonine kinase PAK1 is an important regulator of cell motility. Both PAK1 and the hormone/cytokine prolactin (PRL) have been implicated in breast cancer cell motility, however, the exact mechanisms guiding PRL/PAK1 signaling in breast cancer cells have not been fully elucidated. Our lab has previously demonstrated that PRL-activated tyrosine kinase JAK2 phosphorylates PAK1 on tyrosines 153, 201, and 285, and that tyrosyl phosphorylated PAK1 (pTyr-PAK1) augments migration and invasion of breast cancer cells. RESULTS: Here we further investigate the mechanisms by which pTyr-PAK1 enhances breast cancer cell motility in response to PRL. We demonstrate a distinct reduction in PRL-induced FAK auto-phosphorylation in T47D and TMX2-28 breast cancer cells overexpressing wild-type PAK1 (PAK1 WT) when compared to cells overexpressing either GFP or phospho-tyrosine-deficient mutant PAK1 (PAK1 Y3F). Furthermore, pTyr-PAK1 phosphorylates MEK1 on Ser298 resulting in subsequent ERK1/2 activation. PRL-induced FAK auto-phosphorylation is rescued in PAK1 WT cells by inhibiting tyrosine phosphatases and tyrosine phosphatase inhibition abrogates cell motility and invasion in response to PRL. siRNA-mediated knockdown of the tyrosine phosphatase PTP-PEST rescues FAK auto-phosphorylation in PAK1 WT cells and reduces both cell motility and invasion. Finally, we provide evidence that PRL-induced pTyr-PAK1 stimulates tumor cell metastasis in vivo. CONCLUSION: These data provide insight into the mechanisms guiding PRL-mediated breast cancer cell motility and invasion and highlight a significant role for pTyr-PAK1 in breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Fosfotirosina/metabolismo , Prolactina/farmacología , Quinasas p21 Activadas/metabolismo , Animales , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Silenciador del Gen/efectos de los fármacos , Humanos , Ratones , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 12/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo
15.
J Cell Sci ; 127(Pt 3): 497-508, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24284071

RESUMEN

Tyrosine phosphorylation is implicated in regulating the adherens junction protein, p120 catenin (p120), however, the mechanisms are not well defined. Here, we show, using substrate trapping, that p120 is a direct target of the protein tyrosine phosphatase, PTP-PEST, in epithelial cells. Stable shRNA knockdown of PTP-PEST in colon carcinoma cells results in an increased cytosolic pool of p120 concomitant with its enhanced tyrosine phosphorylation and decreased association with E-cadherin. Consistent with this, PTP-PEST knockdown cells exhibit increased motility, enhanced Rac1 and decreased RhoA activity on a collagen substrate. Furthermore, p120 localization is enhanced at actin-rich protrusions and lamellipodia and has an increased association with the guanine nucleotide exchange factor, VAV2, and cortactin. Exchange factor activity of VAV2 is enhanced by PTP-PEST knockdown whereas overexpression of a VAV2 C-terminal domain or DH domain mutant blocks cell motility. Analysis of point mutations identified tyrosine 335 in the N-terminal domain of p120 as the site of PTP-PEST dephosphorylation. A Y335F mutant of p120 failed to induce the 'p120 phenotype', interact with VAV2, stimulate cell motility or activate Rac1. Together, these data suggest that PTP-PEST affects epithelial cell motility by controlling the distribution and phosphorylation of p120 and its availability to control Rho GTPase activity.


Asunto(s)
Movimiento Celular/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteína Activadora de GTPasa p120/genética , Proteínas de Unión al GTP rho/genética , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Células Epiteliales , Humanos , Mutación , Fosforilación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Tirosina/genética , Proteína Activadora de GTPasa p120/metabolismo , Proteínas de Unión al GTP rho/metabolismo
16.
J Biol Chem ; 288(36): 25739-25748, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-23897807

RESUMEN

PTP-PEST is a cytosolic ubiquitous protein tyrosine phosphatase (PTP) that contains, in addition to its catalytic domain, several protein-protein interaction domains that allow it to interface with several signaling pathways. Among others, PTP-PEST is a key regulator of cellular motility and cytoskeleton dynamics. The complexity of the PTP-PEST interactome underscores the necessity to identify its interacting partners and physiological substrates in order to further understand its role in focal adhesion complex turnover and actin organization. Using a modified yeast substrate trapping two-hybrid system, we identified a cytosolic adaptor protein named Src kinase-associated phosphoprotein 55 homologue (SKAP-Hom) as a novel substrate of PTP-PEST. To confirm PTP-PEST interaction with SKAP-Hom, in vitro pull down assays were performed demonstrating that the PTP catalytic domain and Proline-rich 1 (P1) domain are respectively binding to the SKAP-Hom Y260 and Y297 residues and its SH3 domain. Subsequently, we generated and rescued SKAP-Hom-deficient mouse embryonic fibroblasts (MEFs) with WT SKAP-Hom, SKAP-Hom tyrosine mutants (Y260F, Y260F/Y297F), or SKAP-Hom SH3 domain mutant (W335K). Given the role of PTP-PEST, wound-healing and trans-well migration assays were performed using the generated lines. Indeed, SKAP-Hom-deficient MEFs showed a defect in migration compared with WT-rescued MEFs. Interestingly, the SH3 domain mutant-rescued MEFs showed an enhanced cell migration corresponding potentially with higher tyrosine phosphorylation levels of SKAP-Hom. These findings suggest a novel role of SKAP-Hom and its phosphorylation in the regulation of cellular motility. Moreover, these results open new avenues by which PTP-PEST regulates cellular migration, a hallmark of metastasis.


Asunto(s)
Movimiento Celular/fisiología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Sustitución de Aminoácidos , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones Noqueados , Mutación Missense , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Técnicas del Sistema de Dos Híbridos , Dominios Homologos src
17.
J Cell Sci ; 125(Pt 22): 5338-52, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22946047

RESUMEN

Cell migration and invasion require regulated turnover of integrin-dependent adhesion complexes. Rap1-GTP-interacting adaptor molecule (RIAM) is an adaptor protein that mediates talin recruitment to the cell membrane, and whose depletion leads to defective melanoma cell migration and invasion. In this study, we investigated the potential involvement of RIAM in focal adhesion (FA) dynamics. RIAM-depleted melanoma and breast carcinoma cells displayed an increased number, size and stability of FAs, which accumulated centrally at the ventral cell surface, a phenotype caused by defective FA disassembly. Impairment in FA disassembly resulting from RIAM knockdown correlated with deficient integrin-dependent mitogen-activated protein kinase kinase (MEK)-Erk1/2 activation and, importantly, overexpression of constitutively active MEK resulted in rescue of FA disassembly and recovery of cell invasion. Furthermore, RIAM-promoted Ras homologue gene family, member A (RhoA) activation following integrin engagement was needed for subsequent Erk1/2 activation. In addition, RhoA overexpression partially rescued the FA phenotype in RIAM-depleted cells, also suggesting a functional role for RhoA downstream of RIAM, but upstream of Erk1/2. RIAM knockdown also led to enhanced phosphorylation of paxillin Tyr118 and Tyr31. However, expression of phosphomimetic and nonphosphorylatable mutants at these paxillin residues indicated that paxillin hyperphosphorylation is a subsequent consequence of the blockade of FA disassembly, but does not cause the FA phenotype. RIAM depletion also weakened the association between FA proteins, suggesting that it has important adaptor roles in the correct assembly of adhesion complexes. Our data suggest that integrin-triggered, RIAM-dependent MEK activation represents a key feedback event required for efficient FA disassembly, which could help explain the role of RIAM in cell migration and invasion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adhesiones Focales/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Animales , Línea Celular Tumoral , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Melanoma/enzimología , Melanoma/patología , Ratones , Modelos Biológicos , Paxillin/metabolismo , Fosforilación , Fosfotirosina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Regulación hacia Arriba , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas/metabolismo
18.
Biochemistry ; 52(33): 5645-55, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23848432

RESUMEN

A robust, high-throughput method has been developed to screen one-bead-one-compound peptide libraries to systematically profile the sequence specificity of protein kinases. Its ability to provide individual sequences of the preferred substrates permits the identification of sequence contextual effects and nonpermissive residues. Application of the library method to kinases Pim1, MKK6, and Csk revealed that Pim1 and Csk are highly active toward peptide substrates and recognize specific sequence motifs, whereas MKK6 has little activity or sequence selectivity against peptide substrates. Pim1 recognizes peptide substrates of the consensus RXR(H/R)X(S/T); it accepts essentially any amino acid at the S/T-2 and S/T+1 positions, but strongly disfavors acidic residues (Asp or Glu) at the S/T-2 position and a proline residue at the S/T+1 position. The selected Csk substrates show strong sequence covariance and fall into two classes with the consensus sequences of (D/E)EPIYϕXϕ and (D/E)(E/D)S(E/D/I)YϕXϕ (where X is any amino acid and ϕ is a hydrophobic amino acid). Database searches and in vitro kinase assays identified phosphatase PTP-PEST as a Pim1 substrate and phosphatase SHP-1 as a potential Csk substrate. Our results demonstrate that the sequence specificity of protein kinases is defined not only by favorable interactions between permissive residue(s) on the substrate and their cognate binding site(s) on the kinase but also by repulsive interactions between the kinase and nonpermissive residue(s).


Asunto(s)
Biblioteca de Péptidos , Péptidos/metabolismo , Proteínas Quinasas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Western Blotting , Proteína Tirosina Quinasa CSK , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Proteínas Fúngicas , MAP Quinasa Quinasa 6/química , MAP Quinasa Quinasa 6/genética , MAP Quinasa Quinasa 6/metabolismo , Proteínas Quinasas Activadas por Mitógenos/química , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estructura Molecular , Péptidos/química , Péptidos/genética , Unión Proteica , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/química , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/química , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Rodaminas/química , Rodaminas/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Familia-src Quinasas/química , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
19.
J Biol Chem ; 287(51): 43180-90, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23105101

RESUMEN

Protein-tyrosine phosphatase (PTP)-PEST (PTPN12) is ubiquitously expressed. It is essential for normal embryonic development and embryonic viability in mice. Herein we addressed the involvement of PTP-PEST in endothelial cell functions using a combination of genetic and biochemical approaches. By generating primary endothelial cells from an inducible PTP-PEST-deficient mouse, we found that PTP-PEST is not needed for endothelial cell differentiation and proliferation or for the control of endothelial cell permeability. Nevertheless, it is required for integrin-mediated adhesion and migration of endothelial cells. PTP-PEST-deficient endothelial cells displayed increased tyrosine phosphorylation of Cas, paxillin, and Pyk2, which were previously also implicated in integrin functions. By eliminating PTP-PEST in endothelial cells in vivo, we obtained evidence that expression of PTP-PEST in endothelial cells is required for normal vascular development and embryonic viability. Therefore, PTP-PEST is a key regulator of integrin-mediated functions in endothelial cells seemingly through its capacity to control Cas, paxillin, and Pyk2. This function explains at least in part the essential role of PTP-PEST in embryonic development and viability.


Asunto(s)
Vasos Sanguíneos/embriología , Permeabilidad de la Membrana Celular , Movimiento Celular , Embrión de Mamíferos/enzimología , Células Endoteliales/citología , Células Endoteliales/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Animales , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Embrión de Mamíferos/citología , Femenino , Quinasa 2 de Adhesión Focal/metabolismo , Genotipo , Integrinas/metabolismo , Uniones Intercelulares/metabolismo , Ratones , Ratones Endogámicos C57BL , Paxillin/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 12/deficiencia
20.
J Cell Sci ; 124(Pt 16): 2816-25, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21807946

RESUMEN

Cdc25A, which is one of the three mammalian CDK-activating Cdc25 protein phosphatases (Cdc25A, B and C), is degraded through SCF(ßTrCP)-mediated ubiquitylation following genomic insult; however, the regulation of the stability of the other two Cdc25 proteins is not well understood. Previously, we showed that Cdc25B is primarily degraded by cellular stresses that activate stress-activated MAPKs, such as Jun NH(2)-terminal kinase (JNK) and p38. Here, we report that Cdc25B was ubiquitylated by SCF(ßTrCP) E3 ligase upon phosphorylation at two Ser residues in the ßTrCP-binding-motif-like sequence D(94)AGLCMDSPSP(104). Point mutation of these Ser residues to alanine (Ala) abolished the JNK-induced ubiquitylation by SCF(ßTrCP), and point mutation of DAG to AAG or DAA eradicated both ßTrCP binding and ubiquitylation. Further analysis of the mode of ßTrCP binding to this region revealed that the PEST-like sequence from E(82)SS to D(94)AG is crucially involved in both the ßTrCP binding and ubiquitylation of Cdc25B. Furthermore, the phospho-mimetic replacement of all 10 Ser residues in the E(82)SS to SPSP(104) region with Asp resulted in ßTrCP binding. Collectively, these results indicate that stress-induced Cdc25B ubiquitylation by SCF(ßTrCP) requires the phosphorylation of S(101)PS(103)P in the ßTrCP-binding-motif-like and adjacent PEST-like sequences.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteolisis , Proteínas Ligasas SKP Cullina F-box/metabolismo , Fosfatasas cdc25/metabolismo , Animales , Daño del ADN , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Mutación/genética , Fosforilación/genética , Unión Proteica/genética , Ingeniería de Proteínas , Dominios y Motivos de Interacción de Proteínas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Serina/genética , Serina/metabolismo , Ubiquitinación/genética , Fosfatasas cdc25/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA