Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 37(15-16): 743-759, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37669874

RESUMEN

Protein tyrosine phosphatases (PTPs) are critical regulators of signal transduction but have yet to be exploited fully for drug development. Receptor protein tyrosine phosphatase δ (RPTPδ/PTPRD) has been shown to elicit tumor-promoting functions, including elevating SRC activity and promoting metastasis in certain cell contexts. Dimerization has been implicated in the inhibition of receptor protein tyrosine phosphatases (RPTPs). We have generated antibodies targeting PTPRD ectodomains with the goal of manipulating their dimerization status ectopically, thereby regulating intracellular signaling. We have validated antibody binding to endogenous PTPRD in a metastatic breast cancer cell line, CAL51, and demonstrated that a monoclonal antibody, RD-43, inhibited phosphatase activity and induced the degradation of PTPRD. Similar effects were observed following chemically induced dimerization of its phosphatase domain. Mechanistically, RD-43 triggered the formation of PTPRD dimers in which the phosphatase activity was impaired. Subsequently, the mAb-PTPRD dimer complex was degraded through lysosomal and proteasomal pathways, independently of secretase cleavage. Consequently, treatment with RD-43 inhibited SRC signaling and suppressed PTPRD-dependent cell invasion. Together, these findings demonstrate that manipulating RPTP function via antibodies to the extracellular segments has therapeutic potential.


Asunto(s)
Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores , Transducción de Señal , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Dimerización , Línea Celular , Monoéster Fosfórico Hidrolasas
2.
Immunity ; 46(4): 635-648, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28410990

RESUMEN

Mice carrying a hypomorphic point mutation in the Ptpn6 gene (Ptpn6spin mice) develop an inflammatory skin disease that resembles neutrophilic dermatosis in humans. Here, we demonstrated that interleukin-1α (IL-1α) signaling through IL-1R and MyD88 in both stromal and immune cells drive inflammation in Ptpn6spin mice. We further identified SYK as a critical kinase that phosphorylates MyD88, promoted MyD88-dependent signaling and mediates dermatosis in Ptpn6spin mice. Our studies further demonstrated that SHP1 encoded by Ptpn6 binds and suppresses SYK activation to inhibit MyD88 phosphorylation. Downstream of SHP1 and SYK-dependent counterregulation of MyD88 tyrosine phosphorylation, we have demonstrated that the scaffolding function of receptor interacting protein kinase 1 (RIPK1) and tumor growth factor-ß activated kinase 1 (TAK1)-mediating signaling were required to spur inflammatory disease. Overall, these studies identify SHP1 and SYK crosstalk as a critical regulator of MyD88 post-translational modifications and IL-1-driven inflammation.


Asunto(s)
Inflamación/inmunología , Interleucina-1alfa/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Enfermedades de la Piel/inmunología , Quinasa Syk/inmunología , Animales , Citometría de Flujo , Células HEK293 , Humanos , Immunoblotting , Inflamación/genética , Inflamación/metabolismo , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Noqueados , Modelos Inmunológicos , Mutación , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Interleucina-1/inmunología , Receptores de Interleucina-1/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Enfermedades de la Piel/genética , Enfermedades de la Piel/metabolismo , Quinasa Syk/genética , Quinasa Syk/metabolismo
3.
Cell ; 147(1): 185-98, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21962515

RESUMEN

Protein tyrosine phosphatase 1B (PTP1B) plays important roles in downregulation of insulin and leptin signaling and is an established therapeutic target for diabetes and obesity. PTP1B is regulated by reactive oxygen species (ROS) produced in response to various stimuli, including insulin. The reversibly oxidized form of the enzyme (PTP1B-OX) is inactive and undergoes profound conformational changes at the active site. We generated conformation-sensor antibodies, in the form of single-chain variable fragments (scFvs), that stabilize PTP1B-OX and thereby inhibit its phosphatase function. Expression of conformation-sensor scFvs as intracellular antibodies (intrabodies) enhanced insulin-induced tyrosyl phosphorylation of the ß subunit of the insulin receptor and its substrate IRS-1 and increased insulin-induced phosphorylation of PKB/AKT. Our data suggest that stabilization of the oxidized, inactive form of PTP1B with appropriate therapeutic molecules may offer a paradigm for phosphatase drug development.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 1/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Anticuerpos , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Oxidación-Reducción , Biblioteca de Péptidos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Anticuerpos de Cadena Única/química
4.
Genes Dev ; 32(13-14): 944-952, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29945887

RESUMEN

The levels of copper, which is an essential element in living organisms, are under tight homeostatic control. Inactivating mutations in ATP7B, a P-type Cu-ATPase that functions in copper excretion, promote aberrant accumulation of the metal, primarily the in liver and brain. This condition underlies Wilson's disease, a severe autosomal recessive disorder characterized by profound hepatic and neurological deficits. Current treatment regimens rely on the use of broad specificity metal chelators as "decoppering" agents; however, there are side effects that limit their effectiveness. Here, we present the characterization of DPM-1001 {methyl 4-[7-hydroxy-10,13-dimethyl-3-({4-[(pyridin-2-ylmethyl)amino]butyl}amino)hexadecahydro-1H-cyclopenta[a]phenanthren-17-yl] pentanoate} as a potent and highly selective chelator of copper that is orally bioavailable. Treatment of cell models, including fibroblasts derived from Wilson's disease patients, eliminated adverse effects associated with copper accumulation. Furthermore, treatment of the toxic milk mouse model of Wilson's disease with DPM-1001 lowered the levels of copper in the liver and brain, removing excess copper by excretion in the feces while ameliorating symptoms associated with the disease. These data suggest that it may be worthwhile to investigate DPM-1001 further as a new therapeutic agent for the treatment of Wilson's disease, with potential for application in other indications associated with elevated copper, including cancer and neurodegenerative diseases.


Asunto(s)
Quelantes/farmacología , Cobre/metabolismo , Degeneración Hepatolenticular/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Línea Celular , Quelantes/uso terapéutico , Cobre/toxicidad , ATPasas Transportadoras de Cobre/genética , ATPasas Transportadoras de Cobre/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Degeneración Hepatolenticular/fisiopatología , Hígado/efectos de los fármacos , Hígado/patología , Ratones
5.
Genes Dev ; 31(19): 1939-1957, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29066500

RESUMEN

Disruption of the balanced modulation of reversible tyrosine phosphorylation has been implicated in the etiology of various human cancers, including breast cancer. Protein Tyrosine Phosphatase N23 (PTPN23) resides in chromosomal region 3p21.3, which is hemizygously or homozygously lost in some breast cancer patients. In a loss-of-function PTPome screen, our laboratory identified PTPN23 as a suppressor of cell motility and invasion in mammary epithelial and breast cancer cells. Now, our TCGA (The Cancer Genome Atlas) database analyses illustrate a correlation between low PTPN23 expression and poor survival in breast cancers of various subtypes. Therefore, we investigated the tumor-suppressive function of PTPN23 in an orthotopic transplantation mouse model. Suppression of PTPN23 in Comma 1Dß cells induced breast tumors within 56 wk. In PTPN23-depleted tumors, we detected hyperphosphorylation of the autophosphorylation site tyrosine in the SRC family kinase (SFK) FYN as well as Tyr142 in ß-catenin. We validated the underlying mechanism of PTPN23 function in breast tumorigenesis as that of a key phosphatase that normally suppresses the activity of FYN in two different models. We demonstrated that tumor outgrowth from PTPN23-deficient BT474 cells was suppressed in a xenograft model in vivo upon treatment with AZD0530, an SFK inhibitor. Furthermore, double knockout of FYN and PTPN23 via CRISPR/CAS9 also attenuated tumor outgrowth from PTPN23 knockout Cal51 cells. Overall, this mechanistic analysis of the tumor-suppressive function of PTPN23 in breast cancer supports the identification of FYN as a therapeutic target for breast tumors with heterozygous or homozygous loss of PTPN23.


Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Tirosina Fosfatasas no Receptoras/genética , Animales , Antineoplásicos/farmacología , Benzodioxoles/farmacología , Neoplasias de la Mama/enzimología , Sistemas CRISPR-Cas , Carcinogénesis/efectos de los fármacos , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Fosforilación/genética , Quinazolinas/farmacología , Tasa de Supervivencia , beta Catenina/metabolismo
6.
J Biol Chem ; 299(5): 104582, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36871762

RESUMEN

The ability to define functional interactions between enzymes and their substrates is crucial for understanding biological control mechanisms; however, such methods face challenges in the transient nature and low stoichiometry of enzyme-substrate interactions. Now, we have developed an optimized strategy that couples substrate-trapping mutagenesis to proximity-labeling mass spectrometry for quantitative analysis of protein complexes involving the protein tyrosine phosphatase PTP1B. This methodology represents a significant shift from classical schemes; it is capable of being performed at near-endogenous expression levels and increasing stoichiometry of target enrichment without a requirement for stimulation of supraphysiological tyrosine phosphorylation levels or maintenance of substrate complexes during lysis and enrichment procedures. Advantages of this new approach are illustrated through application to PTP1B interaction networks in models of HER2-positive and Herceptin-resistant breast cancer. We have demonstrated that inhibitors of PTP1B significantly reduced proliferation and viability in cell-based models of acquired and de novo Herceptin resistance in HER2-positive breast cancer. Using differential analysis, comparing substrate-trapping to wild-type PTP1B, we have identified multiple unreported protein targets of PTP1B with established links to HER2-induced signaling and provided internal validation of method specificity through overlap with previously identified substrate candidates. Overall, this versatile approach can be readily integrated with evolving proximity-labeling platforms (TurboID, BioID2, etc.), and is broadly applicable across all PTP family members for the identification of conditional substrate specificities and signaling nodes in models of human disease.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Transducción de Señal , Femenino , Humanos , Neoplasias de la Mama/genética , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas/metabolismo , Trastuzumab/farmacología , Mapeo de Interacción de Proteínas
7.
Cell ; 139(3): 464-5, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19879835

RESUMEN

Catalytically inactive pseudophosphatases are able to signal in the absence of enzymatic activity. Analyzing the oocyte-to-zygote transition in the worm, Cheng et al. (2009) and Parry et al. (2009) now show how two pseudophosphatases, EGG-4 and EGG-5, can regulate signaling by the DYRK family kinase MBK-2.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Transducción de Señal
8.
Genes Dev ; 30(13): 1542-57, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27401557

RESUMEN

Ovarian cancer cells disseminate readily within the peritoneal cavity, which promotes metastasis, and are often resistant to chemotherapy. Ovarian cancer patients tend to present with advanced disease, which also limits treatment options; consequently, new therapies are required. The oncoprotein tyrosine kinase MET, which is the receptor for hepatocyte growth factor (HGF), has been implicated in ovarian tumorigenesis and has been the subject of extensive drug development efforts. Here, we report a novel ligand- and autophosphorylation-independent activation of MET through the nonreceptor tyrosine kinase feline sarcoma-related (FER). We demonstrated that the levels of FER were elevated in ovarian cancer cell lines relative to those in immortalized normal surface epithelial cells and that suppression of FER attenuated the motility and invasive properties of these cancer cells. Furthermore, loss of FER impaired the metastasis of ovarian cancer cells in vivo. Mechanistically, we demonstrated that FER phosphorylated a signaling site in MET: Tyr1349. This enhanced activation of RAC1/PAK1 and promoted a kinase-independent scaffolding function that led to recruitment and phosphorylation of GAB1 and the specific activation of the SHP2-ERK signaling pathway. Overall, this analysis provides new insights into signaling events that underlie metastasis in ovarian cancer cells, consistent with a prometastatic role of FER and highlighting its potential as a novel therapeutic target for metastatic ovarian cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/fisiopatología , Fosfoproteínas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular Tumoral , Movimiento Celular , Activación Enzimática , Femenino , Factor de Crecimiento de Hepatocito , Humanos , Ratones SCID , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Neoplasias Ováricas/mortalidad , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/genética , Células Tumorales Cultivadas
9.
IUBMB Life ; 75(4): 337-352, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36971473

RESUMEN

In October 2020, we were finally able to gather for a celebration of Eddy Fischer's 100th birthday. As with many other events, COVID had disrupted and restricted preparations for the gathering, which ultimately was held via ZOOM. Nevertheless, it was a wonderful opportunity to share a day with Eddy, an exceptional scientist and true renaissance man, and to appreciate his stellar contributions to science. Eddy Fischer, together with Ed Krebs, was responsible for the discovery of reversible protein phosphorylation, which launched the entire field of signal transduction. The importance of this seminal work is now being felt throughout the biotechnology industry with the development of drugs that target protein kinases, which have transformed the treatment of a wide array of cancers. I was privileged to have worked with Eddy both as a postdoc and a junior faculty member, during which time we laid the foundations for our current understanding of the protein tyrosine phosphatase (PTP) family of enzymes and their importance as critical regulators of signal transduction. This tribute to Eddy is based upon the talk I presented at the event, giving a personal perspective on Eddy's influence on my career, our early research efforts together in this area, and how the field has developed since then.


Asunto(s)
COVID-19 , Quercus , Humanos , Quercus/metabolismo , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal , Fosforilación
10.
Nat Chem Biol ; 16(2): 122-125, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31873221

RESUMEN

We have identified a molecular interaction between the reversibly oxidized form of protein tyrosine phosphatase 1B (PTP1B) and 14-3-3ζ that regulates PTP1B activity. Destabilizing the transient interaction between 14-3-3ζ and PTP1B prevented PTP1B inactivation by reactive oxygen species and decreased epidermal growth factor receptor phosphorylation. Our data suggest that destabilizing the interaction between 14-3-3ζ and the reversibly oxidized and inactive form of PTP1B may establish a path to PTP1B activation in cells.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteínas 14-3-3/metabolismo , Biotinilación , Activación Enzimática , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Oxidación-Reducción , Fosforilación , Mapas de Interacción de Proteínas , Proteína Tirosina Fosfatasa no Receptora Tipo 1/química , Especies Reactivas de Oxígeno/metabolismo , Serina/metabolismo , Tirosina/metabolismo
11.
Mol Cell ; 55(5): 782-90, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25175024

RESUMEN

Oncogenic RAS (H-RAS(V12)) induces premature senescence in primary cells by triggering production of reactive oxygen species (ROS), but the molecular role of ROS in senescence remains elusive. We investigated whether inhibition of protein tyrosine phosphatases by ROS contributed to H-RAS(V12)-induced senescence. We identified protein tyrosine phosphatase 1B (PTP1B) as a major target of H-RAS(V12)-induced ROS. Inactivation of PTP1B was necessary and sufficient to induce premature senescence in H-RAS(V12)-expressing IMR90 fibroblasts. We identified phospho-Tyr 393 of argonaute 2 (AGO2) as a direct substrate of PTP1B. Phosphorylation of AGO2 at Tyr 393 inhibited loading with microRNAs (miRNAs) and thus miRNA-mediated gene silencing, which counteracted the function of H-RAS(V12)-induced oncogenic miRNAs. Overall, our data illustrate that premature senescence in H-RAS(V12)-transformed primary cells is a consequence of oxidative inactivation of PTP1B and inhibition of miRNA-mediated gene silencing.


Asunto(s)
Proteínas Argonautas/metabolismo , Silenciador del Gen , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , Tirosina/metabolismo , Proteínas ras/fisiología , Proteínas Argonautas/química , Línea Celular , Senescencia Celular/genética , Humanos , MicroARNs/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tirosina/química , Proteínas ras/genética , Proteínas ras/metabolismo
12.
J Biol Chem ; 295(22): 7669-7685, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32327488

RESUMEN

Increased permeability of vascular lung tissues is a hallmark of acute lung injury and is often caused by edemagenic insults resulting in inflammation. Vascular endothelial (VE)-cadherin undergoes internalization in response to inflammatory stimuli and is recycled at cell adhesion junctions during endothelial barrier re-establishment. Here, we hypothesized that phospholipase D (PLD)-generated phosphatidic acid (PA) signaling regulates VE-cadherin recycling and promotes endothelial barrier recovery by dephosphorylating VE-cadherin. Genetic deletion of PLD2 impaired recovery from protease-activated receptor-1-activating peptide (PAR-1-AP)-induced lung vascular permeability and potentiated inflammation in vivo In human lung microvascular endothelial cells (HLMVECs), inhibition or deletion of PLD2, but not of PLD1, delayed endothelial barrier recovery after thrombin stimulation. Thrombin stimulation of HLMVECs increased co-localization of PLD2-generated PA and VE-cadherin at cell-cell adhesion junctions. Inhibition of PLD2 activity resulted in prolonged phosphorylation of Tyr-658 in VE-cadherin during the recovery phase 3 h post-thrombin challenge. Immunoprecipitation experiments revealed that after HLMVECs are thrombin stimulated, PLD2, VE-cadherin, and protein-tyrosine phosphatase nonreceptor type 14 (PTPN14), a PLD2-dependent protein-tyrosine phosphatase, strongly associate with each other. PTPN14 depletion delayed VE-cadherin dephosphorylation, reannealing of adherens junctions, and barrier function recovery. PLD2 inhibition attenuated PTPN14 activity and reversed PTPN14-dependent VE-cadherin dephosphorylation after thrombin stimulation. Our findings indicate that PLD2 promotes PTPN14-mediated dephosphorylation of VE-cadherin and that redistribution of VE-cadherin at adherens junctions is essential for recovery of endothelial barrier function after an edemagenic insult.


Asunto(s)
Antígenos CD/metabolismo , Barrera Alveolocapilar/metabolismo , Cadherinas/metabolismo , Células Endoteliales/metabolismo , Fosfolipasa D/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Uniones Adherentes/metabolismo , Animales , Barrera Alveolocapilar/citología , Células Endoteliales/citología , Femenino , Humanos , Masculino , Ratones , Fosforilación/efectos de los fármacos , Trombina/farmacología
13.
J Biol Chem ; 293(5): 1517-1525, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29217773

RESUMEN

The protein-tyrosine phosphatase PTP1B is a negative regulator of insulin and leptin signaling and a highly validated therapeutic target for diabetes and obesity. Conventional approaches to drug development have produced potent and specific PTP1B inhibitors, but these inhibitors lack oral bioavailability, which limits their potential for drug development. Here, we report that DPM-1001, an analog of the specific PTP1B inhibitor trodusquemine (MSI-1436), is a potent, specific, and orally bioavailable inhibitor of PTP1B. DPM-1001 also chelates copper, which enhanced its potency as a PTP1B inhibitor. DPM-1001 displayed anti-diabetic properties that were associated with enhanced signaling through insulin and leptin receptors in animal models of diet-induced obesity. Therefore, DPM-1001 represents a proof of concept for a new approach to therapeutic intervention in diabetes and obesity. Although the PTPs have been considered undruggable, the findings of this study suggest that allosteric PTP inhibitors may help reinvigorate drug development efforts that focus on this important family of signal-transducing enzymes.


Asunto(s)
Cobre/metabolismo , Inhibidores Enzimáticos , Insulina/metabolismo , Leptina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Administración Oral , Animales , Quelantes/farmacocinética , Quelantes/farmacología , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Células Hep G2 , Degeneración Hepatolenticular/tratamiento farmacológico , Degeneración Hepatolenticular/genética , Degeneración Hepatolenticular/metabolismo , Humanos , Insulina/genética , Leptina/genética , Ratones , Ratones Transgénicos , Modelos Biológicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo
14.
Genes Dev ; 25(13): 1412-25, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21724833

RESUMEN

We used an RNAi-mediated loss-of-function screen to study systematically the role of the protein tyrosine phosphatase (PTP) superfamily of enzymes in mammary epithelial cell motility in the absence or presence of the oncoprotein tyrosine kinase ERBB2. We report that although shRNAs directed against most of the PTP family were without effect, suppression of three PTPs-PRPN23, PTPRG, and PTPRR-enhanced cell motility. Furthermore, we found that suppression of PTPN23, but not PTPRG or PTPRR, induced cell invasion. Suppression of PTPN23 increased E-cadherin internalization, impaired early endosome trafficking of E-cadherin, induced the expression of mesenchymal proteins, and caused cell scattering. The activity of SRC and ß-catenin was elevated when PTPN23 was suppressed. Moreover, we identified SRC, E-cadherin, and ß-catenin as direct substrates of PTPN23. Inhibition of SRC with the small molecular inhibitor SU6656 blocked the effects of PTPN23 depletion. These findings suggest that loss of PTPN23 may increase the activity of SRC and the phosphorylation status of the E-cadherin/ß-catenin signaling complex to promote tumor growth and invasive behavior in breast cancer. In addition, our studies highlight functional specificity among PTPs and reveal new roles for PTPs in mammary epithelial cell biology.


Asunto(s)
Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Cadherinas/metabolismo , Caveolina 1/metabolismo , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular , Endocitosis , Endosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Invasividad Neoplásica , Transporte de Proteínas , Proteínas Tirosina Fosfatasas no Receptoras/genética , Interferencia de ARN , Receptor ErbB-2/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Proteínas Tirosina Fosfatasas Clase 7 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 7 Similares a Receptores/metabolismo , Transducción de Señal
15.
J Biol Chem ; 290(15): 9646-59, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25681440

RESUMEN

We used a loss-of-function screen to investigate the role of classical protein-tyrosine phosphatases (PTPs) in three-dimensional mammary epithelial cell morphogenesis and ERBB2 signaling. The study revealed a novel role for PTPD2 as a positive regulator of ERBB2 signaling. Suppression of PTPD2 attenuated the ERBB2-induced multiacinar phenotype in three-dimensional cultures specifically by inhibiting ERBB2-mediated loss of polarity and lumen filling. In contrast, overexpression of PTPD2 enhanced the ERBB2 phenotype. We also found that a lipid second messenger, phosphatidic acid, bound PTPD2 in vitro and enhanced its catalytic activity. Small molecule inhibitors of phospholipase D (PLD), an enzyme that produces phosphatidic acid in cells, also attenuated the ERBB2 phenotype. Exogenously added phosphatidic acid rescued the PLD-inhibition phenotype, but only when PTPD2 was present. These findings illustrate a novel pathway involving PTPD2 and the lipid second messenger phosphatidic acid that promotes ERBB2 function.


Asunto(s)
Células Epiteliales/metabolismo , Ácidos Fosfatidicos/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Apoptosis/efectos de los fármacos , Apoptosis/genética , Técnicas de Cultivo de Célula , Línea Celular , Colágeno , Combinación de Medicamentos , Células Epiteliales/efectos de los fármacos , Humanos , Immunoblotting , Indoles/farmacología , Laminina , Glándulas Mamarias Humanas/citología , Microscopía Fluorescente , Microscopía de Contraste de Fase , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/metabolismo , Unión Proteica , Proteínas Tirosina Fosfatasas no Receptoras/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteoglicanos , Interferencia de ARN , Receptor ErbB-2/genética , Sulfonamidas/farmacología , Tacrolimus/análogos & derivados , Tacrolimus/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
16.
J Biol Chem ; 290(26): 15934-47, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25897081

RESUMEN

Despite significant evidence to the contrary, the view that phosphatases are "nonspecific" still pervades the field. Systems biology approaches to defining how signal transduction pathways are integrated at the level of whole organisms also often downplay the contribution of phosphatases, defining them as "erasers" that serve merely to restore the system to its basal state. Here, we present a study that counteracts the idea of "nonspecific phosphatases." We have characterized two structurally similar and functionally related kinases, BRK and SRC, which are regulated by combinations of activating autophosphorylation and inhibitory C-terminal sites of tyrosine phosphorylation. We demonstrated specificity at the level of the kinases in that SRMS phosphorylated the C terminus of BRK, but not SRC; in contrast, CSK is the kinase responsible for C-terminal phosphorylation of SRC, but not BRK. For the phosphatases, we observed that RNAi-mediated suppression of PTP1B resulted in opposing effects on the activity of BRK and SRC and have defined the mechanisms underlying this specificity. PTP1B inhibited BRK by directly dephosphorylating the Tyr-342 autophosphorylation site. In contrast, PTP1B potentiated SRC activity, but not by dephosphorylating SRC itself directly; instead, PTP1B regulated the interaction between CBP/PAG and CSK. SRC associated with, and phosphorylated, the transmembrane protein CBP/PAG at Tyr-317, resulting in CSK recruitment. We identified PAG as a substrate of PTP1B, and dephosphorylation abolished recruitment of the inhibitory kinase CSK. Overall, these findings illustrate how the combinatorial effects of PTKs and PTPs may be integrated to regulate signaling, with both classes of enzymes displaying exquisite specificity.


Asunto(s)
Neoplasias de la Mama/enzimología , Proteínas de Neoplasias/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencias de Aminoácidos , Animales , Neoplasias de la Mama/genética , Proteína Tirosina Quinasa CSK , Femenino , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/genética , Familia-src Quinasas/química , Familia-src Quinasas/genética
17.
Nat Chem Biol ; 10(7): 558-66, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24845231

RESUMEN

PTP1B, a validated therapeutic target for diabetes and obesity, has a critical positive role in HER2 signaling in breast tumorigenesis. Efforts to develop therapeutic inhibitors of PTP1B have been frustrated by the chemical properties of the active site. We define a new mechanism of allosteric inhibition that targets the C-terminal, noncatalytic segment of PTP1B. We present what is to our knowledge the first ensemble structure of PTP1B containing this intrinsically disordered segment, within which we identified a binding site for the small-molecule inhibitor MSI-1436. We demonstrate binding to a second site close to the catalytic domain, with cooperative effects between the two sites locking PTP1B in an inactive state. MSI-1436 antagonized HER2 signaling, inhibited tumorigenesis in xenografts and abrogated metastasis in the NDL2 mouse model of breast cancer, validating inhibition of PTP1B as a therapeutic strategy in breast cancer. This new approach to inhibition of PTP1B emphasizes the potential of disordered segments of proteins as specific binding sites for therapeutic small molecules.


Asunto(s)
Sitio Alostérico/efectos de los fármacos , Antineoplásicos/farmacología , Colestanos/farmacología , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Espermina/análogos & derivados , Regulación Alostérica/efectos de los fármacos , Animales , Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Dominio Catalítico , Colestanos/química , Femenino , Humanos , Cinética , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Modelos Moleculares , Terapia Molecular Dirigida , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transducción de Señal , Espermina/química , Espermina/farmacología
18.
Biochem J ; 465(1): 89-101, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25287652

RESUMEN

MIM (Missing-in-Metastasis), also known as MTSS1 (metastasis suppressor 1), is a scaffold protein that is down-regulated in multiple metastatic cancer cell lines compared with non-metastatic counterparts. MIM regulates cytoskeletal dynamics and actin polymerization, and has been implicated in the control of cell motility and invasion. MIM has also been shown to bind to a receptor PTP (protein tyrosine phosphatase), PTPδ, an interaction that may provide a link between tyrosine-phosphorylation-dependent signalling and metastasis. We used shRNA-mediated gene silencing to investigate the consequences of loss of MIM on the migration and invasion of the MCF10A mammary epithelial cell model of breast cancer. We observed that suppression of MIM by RNAi enhanced migration and invasion of MCF10A cells, effects that were associated with increased levels of PTPδ. Furthermore, analysis of human clinical data indicated that PTPδ was elevated in breast cancer samples when compared with normal tissue. We demonstrated that the SRC protein tyrosine kinase is a direct substrate of PTPδ and, upon suppression of MIM, we observed changes in the phosphorylation status of SRC; in particular, the inhibitory site (Tyr527) was hypophosphorylated, whereas the activating autophosphorylation site (Tyr416) was hyperphosphorylated. Thus the absence of MIM led to PTPδ-mediated activation of SRC. Finally, the SRC inhibitor SU6656 counteracted the effects of MIM suppression on cell motility and invasion. The present study illustrates that both SRC and PTPδ have the potential to be therapeutic targets for metastatic tumours associated with loss of MIM.


Asunto(s)
Movimiento Celular , Proteínas de Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Familia-src Quinasas/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Activación Enzimática , Células Epiteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Humanas/patología , Proteínas de Microfilamentos/deficiencia , Invasividad Neoplásica , Proteínas de Neoplasias/deficiencia , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Especificidad por Sustrato
19.
Biochem J ; 465(3): 433-42, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25406946

RESUMEN

Although DNA encodes the molecular instructions that underlie the control of cell function, it is the proteins that are primarily responsible for implementing those instructions. Therefore quantitative analyses of the proteome would be expected to yield insights into important candidates for the detection and treatment of disease. We present an iTRAQ (isobaric tag for relative and absolute quantification)-based proteomic analysis of ten ovarian cancer cell lines and two normal ovarian surface epithelial cell lines. We profiled the abundance of 2659 cellular proteins of which 1273 were common to all 12 cell lines. Of the 1273, 75 proteins exhibited elevated expression and 164 proteins had diminished expression in the cancerous cells compared with the normal cell lines. The iTRAQ expression profiles allowed us to segregate cell lines based upon sensitivity and resistance to carboplatin. Importantly, we observed no substantial correlation between protein abundance and RNA expression or epigenetic DNA methylation data. Furthermore, we could not discriminate between sensitivity and resistance to carboplatin on the basis of RNA expression and DNA methylation data alone. The present study illustrates the importance of proteomics-based discovery for defining the basis for the carboplatin response in ovarian cancer and highlights candidate proteins, particularly involved in cellular redox regulation, homologous recombination and DNA damage repair, which otherwise could not have been predicted from whole genome and expression data sources alone.


Asunto(s)
Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Platino (Metal)/farmacología , Proteómica/métodos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Daño del ADN/fisiología , Femenino , Humanos
20.
Proc Natl Acad Sci U S A ; 110(33): 13398-403, 2013 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-23901112

RESUMEN

The inhibitory reversible oxidation of protein tyrosine phosphatases (PTPs) is an important regulatory mechanism in growth factor signaling. Studies on PTP oxidation have focused on pathways that increase or decrease reactive oxygen species levels and thereby affect PTP oxidation. The processes involved in reactivation of oxidized PTPs remain largely unknown. Here the role of the thioredoxin (Trx) system in reactivation of oxidized PTPs was analyzed using a combination of in vitro and cell-based assays. Cells lacking the major Trx reductase TrxR1 (Txnrd1(-/-)) displayed increased oxidation of PTP1B, whereas SHP2 oxidation was unchanged. Furthermore, in vivo-oxidized PTP1B was reduced by exogenously added Trx system components, whereas SHP2 oxidation remained unchanged. Trx1 reduced oxidized PTP1B in vitro but failed to reactivate oxidized SHP2. Interestingly, the alternative TrxR1 substrate TRP14 also reactivated oxidized PTP1B, but not SHP2. Txnrd1-depleted cells displayed increased phosphorylation of PDGF-ß receptor, and an enhanced mitogenic response, after PDGF-BB stimulation. The TrxR inhibitor auranofin also increased PDGF-ß receptor phosphorylation. This effect was not observed in cells specifically lacking PTP1B. Together these results demonstrate that the Trx system, including both Trx1 and TRP14, impacts differentially on the oxidation of individual PTPs, with a preference of PTP1B over SHP2 activation. The studies demonstrate a previously unrecognized pathway for selective redox-regulated control of receptor tyrosine kinase signaling.


Asunto(s)
Activación Enzimática/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Tiorredoxinas/farmacología , Animales , Activación Enzimática/efectos de los fármacos , Fibroblastos , Técnicas de Inactivación de Genes , Violeta de Genciana , Ratones , Oxidación-Reducción , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxina Reductasa 1/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA