Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 319(2): L294-L311, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32491951

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive, often fatal, fibrosing lung disease for which treatment remains suboptimal. Fibrogenic cytokines, including transforming growth factor-ß (TGF-ß), are central to its pathogenesis. Protein tyrosine phosphatase-α (PTPα) has emerged as a key regulator of fibrogenic signaling in fibroblasts. We have reported that mice globally deficient in PTPα (Ptpra-/-) were protected from experimental pulmonary fibrosis, in part via alterations in TGF-ß signaling. The goal of this study was to determine the lung cell types and mechanisms by which PTPα controls fibrogenic pathways and whether these pathways are relevant to human disease. Immunohistochemical analysis of lungs from patients with IPF revealed that PTPα was highly expressed by mesenchymal cells in fibroblastic foci and by airway and alveolar epithelial cells. To determine whether PTPα promotes profibrotic signaling pathways in lung fibroblasts and/or epithelial cells, we generated mice with conditional (floxed) Ptpra alleles (Ptpraf/f). These mice were crossed with Dermo1-Cre or with Sftpc-CreERT2 mice to delete Ptpra in mesenchymal cells and alveolar type II cells, respectively. Dermo1-Cre/Ptpraf/f mice were protected from bleomycin-induced pulmonary fibrosis, whereas Sftpc-CreERT2/Ptpraf/f mice developed pulmonary fibrosis equivalent to controls. Both canonical and noncanonical TGF-ß signaling and downstream TGF-ß-induced fibrogenic responses were attenuated in isolated Ptpra-/- compared with wild-type fibroblasts. Furthermore, TGF-ß-induced tyrosine phosphorylation of TGF-ß type II receptor and of PTPα were attenuated in Ptpra-/- compared with wild-type fibroblasts. The phenotype of cells genetically deficient in PTPα was recapitulated with the use of a Src inhibitor. These findings suggest that PTPα amplifies profibrotic TGF-ß-dependent pathway signaling in lung fibroblasts.


Asunto(s)
Fibroblastos/metabolismo , Pulmón/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Bleomicina/farmacología , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Transducción de Señal/efectos de los fármacos
2.
Semin Cell Dev Biol ; 37: 82-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25256403

RESUMEN

This review surveys the contributions of protein tyrosine phosphatases (PTPs) to maintenance and differentiation of stem and progenitor cells. A diverse family of PTPs acts at multiple steps of signaling cascades and cellular locales. Their activities as signaling modifiers provide another layer of integration of signaling pathways, which functionally link to the cellular activity and ultimately converge into the regulation of stem and progenitor cell fates. The development of agents targeting PTPs is a growing endeavor that will benefit stem cell biology and offer promising therapeutic strategies.


Asunto(s)
Proteínas Tirosina Fosfatasas/metabolismo , Células Madre/citología , Células Madre/metabolismo , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Células Madre Pluripotentes/metabolismo , Transducción de Señal
3.
J Cell Sci ; 127(Pt 11): 2420-32, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24652832

RESUMEN

Epithelial junctions are fundamental determinants of tissue organization, subject to regulation by tyrosine phosphorylation. Homophilic binding of E-cadherin activates tyrosine kinases, such as Src, that control junctional integrity. Protein tyrosine phosphatases (PTPs) also contribute to cadherin-based adhesion and signaling, but little is known about their specific identity or functions at epithelial junctions. Here, we report that the receptor PTP RPTPα (human gene name PTPRA) is recruited to epithelial adherens junctions at the time of cell-cell contact, where it is in molecular proximity to E-cadherin. RPTPα is required for appropriate cadherin-dependent adhesion and for cyst architecture in three-dimensional culture. Loss of RPTPα impairs adherens junction integrity, as manifested by defective E-cadherin accumulation and peri-junctional F-actin density. These effects correlate with a role for RPTPα in cellular (c)-Src activation at sites of E-cadherin engagement. Mechanistically, RPTPα is required for appropriate tyrosine phosphorylation of cortactin, a major Src substrate and a cytoskeletal actin organizer. Expression of a phosphomimetic cortactin mutant in RPTPα-depleted cells partially rescues F-actin and E-cadherin accumulation at intercellular contacts. These findings indicate that RPTPα controls cadherin-mediated signaling by linking homophilic E-cadherin engagement to cortactin tyrosine phosphorylation through c-Src.


Asunto(s)
Uniones Adherentes/metabolismo , Cadherinas/metabolismo , Células Epiteliales/fisiología , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Familia-src Quinasas/metabolismo , Actinas/metabolismo , Uniones Adherentes/genética , Proteína Tirosina Quinasa CSK , Células CACO-2 , Adhesión Celular/genética , Cortactina/genética , Cortactina/metabolismo , Células HEK293 , Humanos , Mutación/genética , Técnicas de Cultivo de Órganos , Organogénesis/genética , Fosforilación/genética , Transporte de Proteínas , ARN Interferente Pequeño/genética , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/genética , Transducción de Señal/genética
4.
Am J Pathol ; 184(5): 1489-502, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24650563

RESUMEN

Fibrotic lung diseases represent a diverse group of progressive and often fatal disorders with limited treatment options. Although the pathogenesis of these conditions remains incompletely understood, receptor type protein tyrosine phosphatase α (PTP-α encoded by PTPRA) has emerged as a key regulator of fibroblast signaling. We previously reported that PTP-α regulates cellular responses to cytokines and growth factors through integrin-mediated signaling and that PTP-α promotes fibroblast expression of matrix metalloproteinase 3, a matrix-degrading proteinase linked to pulmonary fibrosis. Here, we sought to determine more directly the role of PTP-α in pulmonary fibrosis. Mice genetically deficient in PTP-α (Ptpra(-/-)) were protected from pulmonary fibrosis induced by intratracheal bleomycin, with minimal alterations in the early inflammatory response or production of TGF-ß. Ptpra(-/-) mice were also protected from pulmonary fibrosis induced by adenoviral-mediated expression of active TGF-ß1. In reciprocal bone marrow chimera experiments, the protective phenotype tracked with lung parenchymal cells but not bone marrow-derived cells. Because fibroblasts are key contributors to tissue fibrosis, we compared profibrotic responses in wild-type and Ptpra(-/-) mouse embryonic and lung fibroblasts. Ptpra(-/-) fibroblasts exhibited hyporesponsiveness to TGF-ß, manifested by diminished expression of αSMA, EDA-fibronectin, collagen 1A, and CTGF. Ptpra(-/-) fibroblasts exhibited markedly attenuated TGF-ß-induced Smad2/3 transcriptional activity. We conclude that PTP-α promotes profibrotic signaling pathways in fibroblasts through control of cellular responsiveness to TGF-ß.


Asunto(s)
Fibroblastos/patología , Pulmón/patología , Fibrosis Pulmonar/patología , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Adenoviridae , Animales , Bleomicina , Citocinas/biosíntesis , Eliminación de Gen , Genes Reporteros , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Neumonía/complicaciones , Neumonía/patología , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/prevención & control , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Transcripción Genética
5.
J Biol Chem ; 287(37): 31104-15, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22822085

RESUMEN

In Helicobacter pylori infection, vacuolating cytotoxin (VacA)-induced mitochondrial damage leading to apoptosis is believed to be a major cause of cell death. It has also been proposed that VacA-induced autophagy serves as a host mechanism to limit toxin-induced cellular damage. Apoptosis and autophagy are two dynamic and opposing processes that must be balanced to regulate cell death and survival. Here we identify the low-density lipoprotein receptor-related protein-1 (LRP1) as the VacA receptor for toxin-induced autophagy in the gastric epithelial cell line AZ-521, and show that VacA internalization through binding to LRP1 regulates the autophagic process including generation of LC3-II from LC3-I, which is involved in formation of autophagosomes and autolysosomes. Knockdown of LRP1 and Atg5 inhibited generation of LC3-II as well as cleavage of PARP, a marker of apoptosis, in response to VacA, whereas caspase inhibitor, benzyloxycarbonyl-VAD-fluoromethylketone (Z-VAD-fmk), and necroptosis inhibitor, Necrostatin-1, did not inhibit VacA-induced autophagy, suggesting that VacA-induced autophagy via LRP1 binding precedes apoptosis. Other VacA receptors such as RPTPα, RPTPß, and fibronectin did not affect VacA-induced autophagy or apoptosis. Therefore, we propose that the cell surface receptor, LRP1, mediates VacA-induced autophagy and apoptosis.


Asunto(s)
Apoptosis , Autofagia , Proteínas Bacterianas/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Proteína 5 Relacionada con la Autofagia , Línea Celular , Inhibidores de Cisteína Proteinasa/farmacología , Epitelio/metabolismo , Epitelio/microbiología , Epitelio/patología , Fibronectinas/genética , Fibronectinas/metabolismo , Mucosa Gástrica/metabolismo , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/patología , Helicobacter pylori/genética , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Estómago/microbiología , Estómago/patología
6.
J Cell Biol ; 161(1): 143-53, 2003 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-12682088

RESUMEN

Cell motility on ECM critically depends on the cellular response to force from the matrix. We find that force-dependent reinforcement of alphav/beta3-integrin-mediated cell-matrix connections requires the receptor-like tyrosine phosphatase alpha (RPTPalpha). RPTPalpha colocalizes with alphav-integrins at the leading edge during early spreading, and coimmunoprecipitates with alphav-integrins during spreading on fibronectin and vitronectin. RPTPalpha-dependent activation of Src family kinases, in particular activation of Fyn, is required for the force-dependent formation of focal complexes and strengthening of alphav/beta3-integrin-cytoskeleton connections during the initial phase of ECM contact. These observations indicate that Src family kinases have distinct functions during adhesion site assembly, and that RPTPalpha is an early component in force-dependent signal transduction pathways leading to the assembly of focal complexes on both fibronectin and vitronectin.


Asunto(s)
Adhesión Celular/genética , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Integrina alfaVbeta3/metabolismo , Proteínas Tirosina Fosfatasas/deficiencia , Receptores de Superficie Celular , Animales , Membrana Celular/ultraestructura , Movimiento Celular/genética , Células Cultivadas , Matriz Extracelular/ultraestructura , Fibroblastos/ultraestructura , Fibronectinas/metabolismo , Técnica del Anticuerpo Fluorescente , Adhesiones Focales/metabolismo , Ratones , Ratones Noqueados , Proteínas Tirosina Fosfatasas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fyn , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Transducción de Señal/genética , Estrés Mecánico , Vimentina/metabolismo , Familia-src Quinasas/metabolismo
7.
Mol Biol Cell ; 17(10): 4330-42, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16870705

RESUMEN

Tyrosine phosphatases (PTPs) epsilon and alpha are closely related and share several molecular functions, such as regulation of Src family kinases and voltage-gated potassium (Kv) channels. Functional interrelationships between PTPepsilon and PTPalpha and the mechanisms by which they regulate K+ channels and Src were analyzed in vivo in mice lacking either or both PTPs. Lack of either PTP increases Kv channel activity and phosphorylation in Schwann cells, indicating these PTPs inhibit Kv current amplitude in vivo. Open probability and unitary conductance of Kv channels are unchanged, suggesting an effect on channel number or organization. PTPalpha inhibits Kv channels more strongly than PTPepsilon; this correlates with constitutive association of PTPalpha with Kv2.1, driven by membranal localization of PTPalpha. PTPalpha, but not PTPepsilon, activates Src in sciatic nerve extracts, suggesting Src deregulation is not responsible exclusively for the observed phenotypes and highlighting an unexpected difference between both PTPs. Developmentally, sciatic nerve myelination is reduced transiently in mice lacking either PTP and more so in mice lacking both PTPs, suggesting both PTPs support myelination but are not fully redundant. We conclude that PTPepsilon and PTPalpha differ significantly in their regulation of Kv channels and Src in the system examined and that similarity between PTPs does not necessarily result in full functional redundancy in vivo.


Asunto(s)
Regulación de la Expresión Génica , Canales de Potasio con Entrada de Voltaje/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Células de Schwann/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Ratones , Ratones Noqueados , Vaina de Mielina/metabolismo , Fosforilación , Canales de Potasio con Entrada de Voltaje/fisiología , Proteínas Tirosina Fosfatasas/genética , Nervio Ciático/metabolismo , Canales de Potasio Shab/metabolismo , Familia-src Quinasas/metabolismo
8.
Psychopharmacology (Berl) ; 234(4): 575-587, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28025742

RESUMEN

RATIONALE: The receptor protein tyrosine phosphatase PTPRG has been genetically associated with psychiatric disorders and is a ligand for members of the contactin family, which are themselves linked to autism spectrum disorders. OBJECTIVE: Based on our finding of a phosphatase-null de novo mutation in PTPRG associated with a case of sporadic schizophrenia, we used PTPRG knockout (KO) mice to model the effect of a loss-of-function mutation. We compared the results with loss-of-function in its close paralogue PTPRZ, previously associated with schizophrenia. We tested PTPRG -/- , PTPRZ -/- , and wild-type male mice for effects on social behavior, forced swim test, and anxiety, as well as on regional brain neurochemistry. RESULTS: The most notable behavioral consequences of PTPRG gene inactivation were reduced immobilization in the forced swim test, suggestive of some negative symptoms of schizophrenia. By contrast, PTPRZ -/- mice demonstrated marked social alteration with increased aggressivity, reminiscent of some positive symptoms of schizophrenia. Both knockouts showed elevated dopamine levels in prefrontal cortex, hippocampus, and most particularly amygdala, but not striatum, accompanied by reduced dopamine beta hydroxylase activity only in amygdala. In addition, PTPRG KO elicited a distinct increase in hippocampal serotonin level not observed in PTPRZ KO. CONCLUSION: PTPRG and PTPRZ gene loss therefore induces distinct patterns of behavioral change and region-specific alterations in neurotransmitters, highlighting their usefulness as models for neuropsychiatric disorder mechanisms and making these receptors attractive targets for therapy.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Dopamina/metabolismo , Hipocampo/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Serotonina/metabolismo , Conducta Social , Animales , Cuerpo Estriado/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Esquizofrenia/metabolismo , Natación
9.
Arthritis Rheumatol ; 68(2): 359-69, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26414708

RESUMEN

OBJECTIVE: During rheumatoid arthritis (RA), fibroblast-like synoviocytes (FLS) critically promote disease pathogenesis by aggressively invading the extracellular matrix of the joint. The focal adhesion kinase (FAK) signaling pathway is emerging as a contributor to the anomalous behavior of RA FLS. The receptor protein tyrosine phosphatase α (RPTPα), which is encoded by the PTPRA gene, is a key promoter of FAK signaling. The aim of this study was to investigate whether RPTPα mediates FLS aggressiveness and RA pathogenesis. METHODS: Through RPTPα knockdown, we assessed FLS gene expression by quantitative polymerase chain reaction analysis and enzyme-linked immunosorbent assay, invasion and migration by Transwell assays, survival by annexin V and propidium iodide staining, adhesion and spreading by immunofluorescence microscopy, and activation of signaling pathways by Western blotting of FLS lysates. Arthritis development was examined in RPTPα-knockout (KO) mice using the K/BxN serum-transfer model. The contribution of radiosensitive and radioresistant cells to disease was evaluated by reciprocal bone marrow transplantation. RESULTS: RPTPα was enriched in the RA synovial lining. RPTPα knockdown impaired RA FLS survival, spreading, migration, invasiveness, and responsiveness to platelet-derived growth factor, tumor necrosis factor, and interleukin-1 stimulation. These phenotypes correlated with increased phosphorylation of Src on inhibitory Y(527) and decreased phosphorylation of FAK on stimulatory Y(397) . Treatment of RA FLS with an inhibitor of FAK phenocopied the knockdown of RPTPα. RPTPα-KO mice were protected from arthritis development, which was due to radioresistant cells. CONCLUSION: By regulating the phosphorylation of Src and FAK, RPTPα mediates proinflammatory and proinvasive signaling in RA FLS, correlating with the promotion of disease in an FLS-dependent model of RA.


Asunto(s)
Artritis Experimental/genética , Artritis Reumatoide/genética , Fibroblastos/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/genética , Familia-src Quinasas/metabolismo , Animales , Articulación del Tobillo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Western Blotting , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/efectos de los fármacos , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Interleucina-1/farmacología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Fosforilación/genética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Reacción en Cadena de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Membrana Sinovial/citología , Factor de Necrosis Tumoral alfa/farmacología , Familia-src Quinasas/efectos de los fármacos
10.
Mol Biol Cell ; 26(7): 1249-62, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25631816

RESUMEN

Cell-cell adhesion couples the contractile cortices of epithelial cells together, generating tension to support a range of morphogenetic processes. E-cadherin adhesion plays an active role in generating junctional tension by promoting actin assembly and cortical signaling pathways that regulate myosin II. Multiple myosin II paralogues accumulate at mammalian epithelial cell-cell junctions. Earlier, we found that myosin IIA responds to Rho-ROCK signaling to support junctional tension in MCF-7 cells. Although myosin IIB is also found at the zonula adherens (ZA) in these cells, its role in junctional contractility and its mode of regulation are less well understood. We now demonstrate that myosin IIB contributes to tension at the epithelial ZA. Further, we identify a receptor type-protein tyrosine phosphatase alpha-Src family kinase-Rap1 pathway as responsible for recruiting myosin IIB to the ZA and supporting contractile tension. Overall these findings reinforce the concept that orthogonal E-cadherin-based signaling pathways recruit distinct myosin II paralogues to generate the contractile apparatus at apical epithelial junctions.


Asunto(s)
Uniones Adherentes/fisiología , Cadherinas/metabolismo , Células Epiteliales/fisiología , Miosina Tipo IIB no Muscular/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Familia-src Quinasas/metabolismo , Uniones Adherentes/metabolismo , Células Epiteliales/metabolismo , Femenino , Humanos , Células MCF-7 , Transducción de Señal
11.
PLoS One ; 8(6): e65371, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785422

RESUMEN

Alterations in function of the neurotrophin BDNF are associated with neurodegeneration, cognitive decline, and psychiatric disorders. BDNF promotes axonal outgrowth and branching, regulates dendritic tree morphology and is important for axonal regeneration after injury, responses that largely result from activation of its tyrosine kinase receptor TrkB. Although intracellular neurotrophin (NT) signaling presumably reflects the combined action of kinases and phosphatases, little is known about the contributions of the latter to TrkB regulation. The issue is complicated by the fact that phosphatases belong to multiple independently evolved families, which are rarely studied together. We undertook a loss-of-function RNA-interference-based screen of virtually all known (254) human phosphatases to understand their function in BDNF/TrkB-mediated neurite outgrowth in differentiated SH-SY5Y cells. This approach identified phosphatases from diverse families, which either positively or negatively modulate BDNF-TrkB-mediated neurite outgrowth, and most of which have little or no previously established function related to NT signaling. "Classical" protein tyrosine phosphatases (PTPs) accounted for 13% of the candidate regulatory phosphatases. The top classical PTP identified as a negative regulator of BDNF-TrkB-mediated neurite outgrowth was PTPN12 (also called PTP-PEST). Validation and follow-up studies showed that endogenous PTPN12 antagonizes tyrosine phosphorylation of TrkB itself, and the downstream activation of ERK1/2. We also found PTPN12 to negatively regulate phosphorylation of p130cas and FAK, proteins with previously described functions related to cell motility and growth cone behavior. Our data provide the first comprehensive survey of phosphatase function in NT signaling and neurite outgrowth. They reveal the complexity of phosphatase control, with several evolutionarily unrelated phosphatase families cooperating to affect this biological response, and hence the relevance of considering all phosphatase families when mining for potentially druggable targets.


Asunto(s)
Neuritas/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Receptor trkB/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proteína Sustrato Asociada a CrK/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Quinasa 1 de Adhesión Focal/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Neuritas/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fenotipo , Monoéster Fosfórico Hidrolasas/genética , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Interferencia de ARN , Reproducibilidad de los Resultados , Transducción de Señal , Tretinoina/farmacología
12.
Biol Psychiatry ; 70(7): 626-35, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21831360

RESUMEN

BACKGROUND: Solid evidence links schizophrenia (SZ) susceptibility to neurodevelopmental processes involving tyrosine phosphorylation-mediated signaling. Mouse studies implicate the Ptpra gene, encoding protein tyrosine phosphatase RPTPα, in the control of radial neuronal migration, cortical cytoarchitecture, and oligodendrocyte differentiation. The human gene encoding RPTPα, PTPRA, maps to a chromosomal region (20p13) associated with susceptibility to psychotic illness. METHODS: We characterized neurobehavioral parameters, as well as gene expression in the central nervous system, of mice with a null mutation in the Ptpra gene. We searched for genetic association between polymorphisms in PTPRA and schizophrenia risk (two independent cohorts, 1420 cases and 1377 controls), and we monitored PTPRA expression in prefrontal dorsolateral cortex of SZ patients (35 cases, 2 control groups of 35 cases). RESULTS: We found that Ptpra⁻/⁻ mice reproduce neurobehavioral endophenotypes of human SZ: sensitization to methamphetamine-induced hyperactivity, defective sensorimotor gating, and defective habituation to a startle response. Ptpra loss of function also leads to reduced expression of multiple myelination genes, mimicking the hypomyelination-associated changes in gene expression observed in postmortem patient brains. We further report that a polymorphism at the PTPRA locus is genetically associated with SZ, and that PTPRA mRNA levels are reduced in postmortem dorsolateral prefrontal cortex of subjects with SZ. CONCLUSIONS: The implication of this well-studied signaling protein in SZ risk and endophenotype manifestation provides novel entry points into the etiopathology of this disease.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/fisiología , Esquizofrenia/genética , Esquizofrenia/fisiopatología , Psicología del Esquizofrénico , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Expresión Génica , Estudios de Asociación Genética/métodos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/genética , Polimorfismo Genético , Corteza Prefrontal/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/biosíntesis , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/genética
13.
Antioxid Redox Signal ; 13(2): 109-25, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20055753

RESUMEN

The role of NO in regulating the focal adhesion proteins, Src, FAK, p130 Cas, and PTP-alpha, was investigated. Fibroblasts expressing PTP-alpha (PTP-alpha(WT) cells), fibroblasts "knockout" for PTP-alpha (PTP-alpha(-/-) cells), and "rescued" "knockout" fibroblasts (PTP-alpha A5/3 cells) were stimulated with either S-nitroso-N-acetylpenicillamine (SNAP) or fetal bovine serum (FBS). FBS increased inducible NO synthase in both cell lines. Activation of Src mediated either by SNAP or by FBS occurred independent of dephosphorylation of Tyr527 in PTP-alpha(-/-) cells. Both stimuli promoted dephosphorylation of Tyr527 and activation of Src kinase in PTP-alpha(WT) cells. NO-mediated activation of Src kinase affected the activities of FAK and p130Cas and was dependent on the expression of PTP-alpha. Analogous to tyrosine phosphorylation, SNAP and FBS stimulated differential generation of NO and S-nitrosylation of Src kinase in both cell lines. Incubation with SNAP resulted in higher levels of NO and S-nitrosylation of immunoprecipitated Src in PTP-alpha(-/-) cells (oxidizing redox environment) as compared with the levels of NO and S-nitrosylated Src in PTP-alpha(WT) cells (reducing redox environment). SNAP differentially stimulated cell proliferation of both cell lines is dependent on the intracellular redox environment, Src activity, and PTP-alpha expression. This dependence also is observed with FBS-stimulated cell migration.


Asunto(s)
Proteína Sustrato Asociada a CrK/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Óxido Nítrico/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Familia-src Quinasas/metabolismo , Animales , Bovinos , Movimiento Celular , Proliferación Celular , Células Cultivadas , Proteína Sustrato Asociada a CrK/genética , Fibroblastos/citología , Fibroblastos/fisiología , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Ratones , Ratones Noqueados , Óxido Nítrico/genética , Donantes de Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oxidación-Reducción , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/genética , S-Nitroso-N-Acetilpenicilamina/metabolismo , Familia-src Quinasas/genética
14.
J Biol Chem ; 283(51): 35815-24, 2008 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-18948260

RESUMEN

Src family tyrosine kinases (SFKs) function in multiple signaling pathways, raising the question of how appropriate regulation and substrate choice are achieved. SFK activity is modulated by several protein-tyrosine phosphatases, among which RPTPalpha and SHP2 are the best established. We studied how RPTPalpha affects substrate specificity and regulation of c-Src and Fyn in response to epidermal growth factor and platelet-derived growth factor. We find that RPTPalpha, in a growth factor-specific manner, directs the specificity of these kinases toward a specific subset of SFK substrates, particularly the focal adhesion protein Paxillin and the lipid raft scaffolding protein Cbp/PAG. A significant fraction of RPTPalpha is present in lipid rafts, where its targets Fyn and Cbp/PAG reside, and growth factor-mediated SFK activation within this compartment is strictly dependent on RPTPalpha. Forced concentration of RPTPalpha into lipid rafts is compatible with activation of Fyn. Finally, RPTPalpha-induced phosphorylation of Paxillin and Cbp/PAG induces recruitment of the SFK inhibitory kinase Csk, indicative of negative feedback loops limiting SFK activation by RPTPalpha. Our findings indicate that individual SFK-controlling PTPs play important and specific roles in dictating SFK substrate specificity and regulatory mechanism.


Asunto(s)
Genes src/fisiología , Microdominios de Membrana/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Animales , Línea Celular , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Factor de Crecimiento Epidérmico/farmacología , Péptidos y Proteínas de Señalización Intercelular , Microdominios de Membrana/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Paxillin/genética , Paxillin/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/genética , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/fisiología
15.
J Immunol ; 180(7): 5017-27, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18354227

RESUMEN

Helicobacter pylori VacA induces multiple effects on susceptible cells, including vacuolation, mitochondrial damage, inhibition of cell growth, and enhanced cyclooxygenase-2 expression. To assess the ability of H. pylori to modulate the production of inflammatory mediators, we examined the mechanisms by which VacA enhanced IL-8 production by promonocytic U937 cells, which demonstrated the greatest VacA-induced IL-8 release of the cells tested. Inhibitors of p38 MAPK (SB203580), ERK1/2 (PD98059), IkappaBalpha ((E)-3-(4-methylphenylsulfonyl)-2-propenenitrile), Ca(2+) entry (SKF96365), and intracellular Ca(2+) channels (dantrolene) blocked VacA-induced IL-8 production. Furthermore, an intracellular Ca(2+) chelator (BAPTA-AM), which inhibited VacA-activated p38 MAPK, caused a dose-dependent reduction in VacA-induced IL-8 secretion by U937 cells, implying a role for intracellular Ca(2+) in mediating activation of MAPK and the canonical NF-kappaB pathway. VacA stimulated translocation of NF-kappaBp65 to the nucleus, consistent with enhancement of IL-8 expression by activation of the NF-kappaB pathway. In addition, small interfering RNA of activating transcription factor (ATF)-2 or CREB, which is a p38MAPK substrate and binds to the AP-1 site of the IL-8 promoter, inhibited VacA-induced IL-8 production. VacA activated an IL-8 promoter containing an NF-IL-6 site, but not a mutated AP-1 or NF-kappaB site, suggesting direct involvement of the ATF-2/CREB binding region or NF-kappaB-binding regions in VacA-induced IL-8 promoter activation. Thus, in U937 cells, VacA directly increases IL-8 production by activation of the p38 MAPK via intracellular Ca(2+) release, leading to activation of the transcription factors, ATF-2, CREB, and NF-kappaB.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Proteínas Bacterianas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Helicobacter pylori/metabolismo , Interleucina-8/biosíntesis , FN-kappa B/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Bacterianas/genética , Hidroxianisol Butilado/farmacología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Dantroleno/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Activación Enzimática/efectos de los fármacos , Helicobacter pylori/genética , Humanos , Imidazoles/farmacología , Interleucina-8/genética , Interleucina-8/metabolismo , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Sulfonas/farmacología , Tapsigargina/farmacología , Células U937 , Regulación hacia Arriba
16.
J Gen Virol ; 89(Pt 7): 1777-1788, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18559949

RESUMEN

Prion diseases are caused by conversion of a normally folded, non-pathogenic isoform of the prion protein (PrP(C)) to a misfolded, pathogenic isoform (PrP(Sc)). Prion inoculation experiments in mice expressing homologous PrP(C) molecules on different genetic backgrounds displayed different incubation times, indicating that the conversion reaction may be influenced by other gene products. To identify genes that contribute to prion pathogenesis, we analysed incubation times of prions in mice in which the gene product was inactivated, knocked out or overexpressed. We tested 20 candidate genes, because their products either colocalize with PrP, are associated with Alzheimer's disease, are elevated during prion disease, or function in PrP-mediated signalling, PrP glycosylation, or protein maintenance. Whereas some of the candidates tested may have a role in the normal function of PrP(C), our data show that many genes previously implicated in prion replication have no discernible effect on the pathogenesis of prion disease. While most genes tested did not significantly affect survival times, ablation of the amyloid beta (A4) precursor protein (App) or interleukin-1 receptor, type I (Il1r1), and transgenic overexpression of human superoxide dismutase 1 (SOD1) prolonged incubation times by 13, 16 and 19 %, respectively.


Asunto(s)
Enfermedades por Prión/genética , Priones/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Dosificación de Gen , Silenciador del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Priones/genética , Receptores Tipo I de Interleucina-1/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Análisis de Supervivencia
17.
J Cell Sci ; 120(Pt 21): 3895-904, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17940065

RESUMEN

Receptor-like protein tyrosine phosphatase alpha (RPTPalpha)-knockout mice have severe hippocampal abnormalities similar to knockouts of the Src family kinase Fyn. These enzymes are linked to the matrix-rigidity response in fibroblasts, but their function in neurons is unknown. The matrix-rigidity response of fibroblasts appears to differ from that of neuronal growth cones but it is unknown whether the rigidity detection mechanism or response pathway is altered. Here, we report that RPTPalpha is required for rigidity-dependent reinforcement of fibronectin (FN)-cytoskeleton bonds and the rigidity response in hippocampal neuron growth cones, like in fibroblasts. In control neurons, rigid FN surfaces inhibit neurite extension and neuron differentiation relative to soft surfaces. In RPTPalpha(-/-) neurons, no inhibition of extension and differentiation is found on both rigid and soft surfaces. The RPTPalpha-dependent rigidity response in neurons is FN-specific, and requires clustering of alpha(v)beta(6) integrin at the leading edge of the growth cones. Further, RPTPalpha is necessary for the rigidity-dependent concentration of Fyn and p130Cas phosphorylation at the leading edge of the growth cone, like it is in fibroblasts. Although neurons respond to rigid FN surfaces in the opposite way to fibroblasts, we suggest that the mechanism of detecting FN rigidity is similar and involves rigidity-dependent RPTPalpha recruitment of Fyn.


Asunto(s)
Diferenciación Celular/fisiología , Hipocampo/citología , Neuronas , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Proteína Sustrato Asociada a CrK/genética , Proteína Sustrato Asociada a CrK/metabolismo , Citoesqueleto/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Integrinas/metabolismo , Ratones , Ratones Noqueados , Neuronas/citología , Neuronas/fisiología , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/genética , Resistencia a la Tracción , Vitronectina/metabolismo , Familia-src Quinasas/metabolismo
18.
Infect Immun ; 75(9): 4472-81, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17591797

RESUMEN

Treatment of AZ-521 cells with Helicobacter pylori VacA increased cyclooxygenase 2 (COX-2) mRNA in a time- and dose-dependent manner. A p38 mitogen-activated protein kinase (MAPK) inhibitor, SB203580, blocked elevation of COX-2 mRNA levels, whereas PD98059, which blocks the Erk1/2 cascade, partially suppressed the increase. Consistent with involvement of p38 MAPK, VacA-induced accumulation of COX-2 mRNA was reduced in AZ-521 cells overexpressing a dominant-negative p38 MAPK (DN-p38). Phosphatidylinositol-specific phospholipase C, which inhibits VacA-induced p38 MAPK activation, blocked VacA-induced COX-2 expression. In parallel with COX-2 expression, VacA increased prostaglandin E(2) (PGE(2)) production, which was inhibited by SB203580 and NS-398, a COX-2 inhibitor. VacA-induced PGE(2) production was markedly attenuated in AZ-521 cells stably expressing DN-p38. VacA increased transcription of a COX-2 promoter reporter gene and activated a COX-2 promoter containing mutated NF-kappaB or NF-interleukin-6 sites but not a mutated cis-acting replication element (CRE) site, suggesting direct involvement of the activating transcription factor 2 (ATF-2)/CREB-binding region in VacA-induced COX-2 promoter activation. The reduction of ATF-2 expression in AZ-521 cells transformed with ATF-2-small interfering RNA duplexes resulted in suppression of COX-2 expression. Thus, VacA enhances PGE(2) production by AZ-521 cells through induction of COX-2 expression via the p38 MAPK/ATF-2 cascade, leading to activation of the CRE site in the COX-2 promoter.


Asunto(s)
Factor de Transcripción Activador 2/fisiología , Factores de Transcripción Activadores/fisiología , Proteínas Bacterianas/fisiología , Ciclooxigenasa 2/biosíntesis , Dinoprostona/biosíntesis , Helicobacter pylori/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Línea Celular Tumoral , Ciclooxigenasa 2/genética , Inducción Enzimática/fisiología , Humanos , ARN Mensajero/biosíntesis , ARN Mensajero/metabolismo , Regulación hacia Arriba/genética
19.
Infect Immun ; 74(12): 6571-80, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17030583

RESUMEN

Helicobacter pylori vacuolating cytotoxin, VacA, induces multiple effects on epithelial cells through different cellular events: one involves pore formation, leading to vacuolation, mitochondrial damage, and apoptosis, and the second involves cell signaling, resulting in stimulation of proinflammatory responses and cell detachment. Our recent data demonstrated that VacA uses receptor-like protein tyrosine phosphatase beta (RPTPbeta) as a receptor, of which five residues (QTTQP) at positions 747 to 751 are involved in binding. In AZ-521 cells, which mainly express RPTPbeta, VacA, after binding to RPTPbeta in non-lipid raft microdomains on the cell surface, is localized with RPTPbeta in lipid rafts in a temperature- and VacA concentration-dependent process. Methyl-beta-cyclodextrin (MCD) did not block binding to RPTPbeta but inhibited translocation of VacA with RPTPbeta to lipid rafts and all subsequent events. On the other hand, 5-nitro-2-(3-phenylpropylamino)-benzoic acid (NPPB), which disrupts anion channels, did not inhibit translocation of VacA to lipid rafts or VacA-induced activation of p38 mitogen-activated protein (MAP) kinase, but inhibited VacA internalization followed by vacuolation. Thus, p38 MAP kinase activation did not appear to be required for internalization. In contrast, phosphatidylinositol-specific phospholipase C (PI-PLC) inhibited translocation, as well as p38 MAP kinase/ATF-2 activation, internalization, and VacA-induced vacuolation. Neither NPPB nor PI-PLC affected VacA binding to cells and to its receptor, RPTPbeta. Thus, receptor-dependent translocation of VacA to lipid rafts is critical for signaling pathways leading to p38 MAP kinase/ATF-2 activation and vacuolation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Microdominios de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Vacuolas/metabolismo , Factor de Transcripción Activador 2/agonistas , Factor de Transcripción Activador 2/metabolismo , Proteínas Bacterianas/análisis , Células Cultivadas , Humanos , Microdominios de Membrana/química , Proteínas del Tejido Nervioso/análisis , Nitrobenzoatos/farmacología , Fosfatidilinositol Diacilglicerol-Liasa/farmacología , Fosfoinositido Fosfolipasa C , Transporte de Proteínas/efectos de los fármacos , Proteínas Tirosina Fosfatasas/análisis , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores , Vacuolas/química , beta-Ciclodextrinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
J Biol Chem ; 277(20): 17406-14, 2002 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-11867627

RESUMEN

SRC family kinases have been consistently and recurrently implicated in neurite extension events, yet the mechanism underlying their neuritogenic role has remained elusive. We report that epidermal growth factor (EGF) can be converted from a non-neuritogenic into a neuritogenic factor through moderate activation of endogenous SRC by receptor-protein-tyrosine phosphatase alpha (a physiological SRC activator). We show that such a qualitative change in the response to EGF is not accompanied by changes in the extent or kinetics of ERK induction in response to this factor. Instead, the pathway involved relies on increased tyrosine phosphorylation of, and recruitment of Crk to, the SRC substrate Sin/Efs. The latter is a scaffolding protein structurally similar to the SRC substrate Cas, tyrosine phosphorylation of which is critical for migration in fibroblasts and epithelial cells. Expression of a dominant negative version of Sin interfered with receptor-protein-tyrosine phosphatase alpha/EGF- as well as fibroblast growth factor-induced neurite outgrowth. These observations uncouple neuritogenic signaling in PC12 cells from sustained activation of ERK kinases and for the first time identify an effector of SRC function in neurite extension.


Asunto(s)
Genes src/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuritas/fisiología , Neuronas/fisiología , Proteínas Tirosina Fosfatasas/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Superficie Celular , Animales , Activación Enzimática , Factor de Crecimiento Epidérmico/metabolismo , Técnica del Anticuerpo Fluorescente , Cinética , Células PC12 , Fosforilación , Proteínas Proto-Oncogénicas c-crk , Ratas , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Transducción de Señal/fisiología , Tirosina/metabolismo , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA