Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Biol Chem ; 292(14): 5748-5759, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28213521

RESUMO

Proteins of the Src homology and collagen (Shc) family are typically involved in signal transduction events involving Ras/MAPK and PI3K/Akt pathways. In the nervous system, they function proximal to the neurotrophic factors that regulate cell survival, differentiation, and neuron-specific characteristics. The least characterized homolog, ShcD, is robustly expressed in the developing and mature nervous system, but its contributions to neural cell circuitry are largely uncharted. We now report that ShcD binds to active Ret, TrkA, and TrkB neurotrophic factor receptors predominantly via its phosphotyrosine-binding (PTB) domain. However, in contrast to the conventional Shc adaptors, ShcD suppresses distal phosphorylation of the Erk MAPK. Accordingly, genetic knock-out of mouse ShcD enhances Erk phosphorylation in the brain. In cultured cells, this capacity is tightly aligned to phosphorylation of ShcD CH1 region tyrosine motifs, which serve as docking platforms for signal transducers, such as Grb2. Erk suppression is relieved through independent mutagenesis of the PTB domain and the CH1 tyrosine residues, and successive substitution of these tyrosines breaks the interaction between ShcD and Grb2, thereby promoting TrkB-Grb2 association. Erk phosphorylation can also be restored in the presence of wild type ShcD through Grb2 overexpression. Conversely, mutation of the ShcD SH2 domain results in enhanced repression of Erk. Although the SH2 domain is a less common binding interface in Shc proteins, we demonstrate that it associates with the Ptpn11 (Shp2) phosphatase, which in turn regulates ShcD tyrosine phosphorylation. We therefore propose a model whereby ShcD competes with neurotrophic receptors for Grb2 binding and opposes activation of the MAPK cascade.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Receptor trkA/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Motivos de Aminoácidos , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/genética , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Fosforilação/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-ret/genética , Receptor trkA/genética , Receptor trkB , Proteínas Adaptadoras da Sinalização Shc/genética
2.
NMR Biomed ; 28(5): 566-75, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25808190

RESUMO

Increased lactate production through glycolysis in aerobic conditions is a hallmark of cancer. Some anticancer drugs have been designed to exploit elevated glycolysis in cancer cells. For example, lonidamine (LND) inhibits lactate transport, leading to intracellular acidification in cancer cells. Chemical exchange saturation transfer (CEST) is a novel MRI contrast mechanism that is dependent on intracellular pH. Amine and amide concentration-independent detection (AACID) and apparent amide proton transfer (APT*) represent two recently developed CEST contrast parameters that are sensitive to pH. The goal of this study was to compare the sensitivity of AACID and APT* for the detection of tumor-selective acidification after LND injection. Using a 9.4-T MRI scanner, CEST data were acquired in mice approximately 14 days after the implantation of 10(5) U87 human glioblastoma multiforme (GBM) cells in the brain, before and after the administration of LND (dose, 50 or 100 mg/kg). Significant dose-dependent LND-induced changes in the measured CEST parameters were detected in brain regions spatially correlated with implanted tumors. Importantly, no changes were observed in T1- and T2-weighted images acquired before and after LND treatment. The AACID and APT* contrast measured before and after LND injection exhibited similar pH sensitivity. Interestingly, LND-induced contrast maps showed increased heterogeneity compared with pre-injection CEST maps. These results demonstrate that CEST contrast changes after the administration of LND could help to localize brain cancer and monitor tumor response to chemotherapy within 1 h of treatment. The LND CEST experiment uses an anticancer drug to induce a metabolic change detectable by endogenous MRI contrast, and therefore represents a unique cancer detection paradigm which differs from other current molecular imaging techniques that require the injection of an imaging contrast agent or tracer.


Assuntos
Neoplasias Encefálicas/química , Neoplasias Encefálicas/diagnóstico , Indazóis/uso terapêutico , Imageamento por Ressonância Magnética/métodos , Imagem Molecular/métodos , Animais , Antineoplásicos/uso terapêutico , Meios de Contraste , Humanos , Concentração de Íons de Hidrogênio , Interpretação de Imagem Assistida por Computador/métodos , Camundongos , Camundongos Nus , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Células Tumorais Cultivadas
3.
J Biol Chem ; 288(33): 23807-13, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23749991

RESUMO

TrkA is a cell surface transmembrane receptor tyrosine kinase for nerve growth factor (NGF). TrkA has an NPXY motif and kinase regulatory loop similar to insulin receptor (INSR) suggesting that NGF→TrkA signaling might overlap with insulin→INSR signaling. During insulin or NGF stimulation TrkA, insulin receptor substrate-1 (IRS-1), INSR (and presumably other proteins) forms a complex in PC12 cells. In PC12 cells, tyrosine phosphorylation of INSR and IRS-1 is dependent upon the functional TrkA kinase domain. Moreover, expression of TrkA kinase-inactive mutant blocked the activation of Akt and Erk5 in response to insulin or NGF. Based on these data, we propose that TrkA participates in insulin signaling pathway in PC12 cells.


Assuntos
Insulina/metabolismo , Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Transdução de Sinais , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Ativação Enzimática/efeitos dos fármacos , Glucose/metabolismo , Humanos , Insulina/farmacologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Fator de Crescimento Neural/farmacologia , Células PC12 , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptor de Insulina/química , Receptor de Insulina/metabolismo , Receptor trkA/química , Transdução de Sinais/efeitos dos fármacos
4.
J Neurochem ; 121(6): 861-80, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22404429

RESUMO

Vesicular transport in neurons plays a vital role in neuronal function and survival. Nesca is a novel protein that we previously identified and herein describe its pattern of expression, subcellular localization and protein-protein interactions both in vitro and in vivo. Specifically, a large proportion of Nesca is in tight association with both actin and microtubule cytoskeletal proteins. Nesca binds to F-actin, microtubules, ßIII and acetylated α-tubulin, but not neurofilaments or the actin-binding protein drebrin, in in vitro-binding assays. Nesca co-immunoprecipitates with kinesin heavy chain (KIF5B) and kinesin light-chain motors as well as with the synaptic membrane precursor protein, syntaxin-1, and is a constituent of the post-synaptic density. Moreover, in vitro-binding assays indicate that Nesca directly binds KIF5B, kinesin light-chain and syntaxin-1. In contrast, Nesca does not co-immunoprecipitate with the kinesin motors KIF1B, KIF3A nor does it bind syntaxin-4 or the synaptosome-associated protein 25 kDa (SNAP-25) in vitro. Nesca expression in neurons is highly punctuate, co-stains with syntaxin-1, and is found in fractions containing markers of early endosomes and Golgi suggesting that it is involved in vesicular transport. Collectively, these data suggest that Nesca functions as an adapter involved in neuronal vesicular transport including vesicles containing soluble N-ethylmaleimide sensitive factor attachment protein receptors that are essential to exocytosis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cinesinas/metabolismo , Neurônios/metabolismo , Sintaxina 1/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Western Blotting , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Hipocampo/metabolismo , Humanos , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Neurogênese/fisiologia , Densidade Pós-Sináptica/metabolismo , Transporte Proteico/fisiologia , Membranas Sinápticas/metabolismo , Transfecção
5.
J Neurochem ; 112(4): 882-99, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19943845

RESUMO

Ectopic expression of the TrkA receptor tyrosine kinase in tumors of the nervous system can mediate nerve growth factor (NGF)-dependent cell death by apoptosis and /or autophagy. Herein, we demonstrate that TrkA can also induce cell death in medulloblastoma Daoy cells by a caspase-independent mechanism that involves the hyperstimulation of macropinocytosis. Specifically, NGF-stimulates the uptake of AlexaFluor546-dextran into lysosome-associated membrane protein-1 positive vacuoles which fuse with microtubule associated protein light chain 3 (LC3) positive autophagosomes, to form large intracellular vacuoles (> 1 mum), which then fuse with lysotracker positive lysosomes. While LC3 cleavage and the appearance of LC3 positive vacuoles suggest the induction of autophagy, siRNA reduced expression of four proteins essential to autophagy (beclin-1, Atg5, LC3 and Atg9) neither blocks NGF-induced vacuole formation nor cell death. TrkA activated cell death does not require p38, JNK or Erk1/2 kinases but does require activation of class III PI-3 kinase and is blocked by the casein kinase 1 (CK1) inhibitor, D4476. This inhibitor does not interfere with TrkA activation but does block NGF-dependent AlexaFluor546-dextran uptake and CK1 dependent phosphorylation of beta-catenin. Collectively, these data demonstrate that TrkA stimulates cell death by a novel mechanism involving CK1-dependent hyperstimulation of macropinocytosis.


Assuntos
Autofagia/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Pinocitose/efeitos dos fármacos , Pinocitose/fisiologia , Receptor trkA/metabolismo , Análise de Variância , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 5 Relacionada à Autofagia , Proteína Beclina-1 , Linhagem Celular Tumoral , Citocromos c/metabolismo , Inibidores Enzimáticos/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação/métodos , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana Lisossomal/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/ultraestrutura , Meduloblastoma/patologia , Meduloblastoma/ultraestrutura , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Eletrônica de Transmissão/métodos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção/métodos
6.
J Neurochem ; 112(4): 924-39, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19943849

RESUMO

Fibroblast growth factor (FGF) mediated signaling is essential to many aspects of neural development. Activated FGF receptors signal primarily through the FGF receptor substrate (Frs) adapters, which include Frs2/Frs2alpha and Frs3/Frs2beta. While some studies suggest that Frs3 can compensate for the loss of Frs2 in transfected cells, the lack of an effective Frs3 specific antibody has prevented efforts to determine the role(s) of the endogenous protein. To this end, we have generated a Frs3 specific antibody and have characterized the pattern of Frs3 expression in the developing nervous system, its subcellular localization as well as its biochemical properties. We demonstrate that Frs3 is expressed at low levels in the ventricular zone of developing cortex, between E12 and E15, and it co-localizes with nestin and acetylated alpha-tubulin in radial processes in the ventricular/subventricular zones as well as with betaIII tubulin in differentiated cortical neurons. Subcellular fractionation studies demonstrate that endogenous Frs3 is both soluble and plasma membrane associated while Frs3 expressed in 293T cells associates exclusively with lipid rafts. Lastly, we demonstrate that neuronal Frs3 binds microtubules comparable to the microtubule-associated protein, MAP2, while Frs2 does not. Collectively, these data suggest that neuronal Frs3 functions as a novel microtubule binding protein and they provide the first biochemical evidence that neuronal Frs3 is functionally distinct from Frs2/Frs2alpha.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , AMP Cíclico/farmacologia , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo , Humanos , Técnicas In Vitro , Antígeno Ki-67/metabolismo , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Microdomínios da Membrana/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Neurônios/efeitos dos fármacos , Ligação Proteica , Proteínas com Domínio T/metabolismo , Tubulina (Proteína)/metabolismo
7.
J Cell Biol ; 164(6): 851-62, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15024033

RESUMO

We provide the first characterization of a novel signaling adapter, Nesca, in neurotrophic signal transduction. Nesca contains a RUN domain, a WW domain, a leucine zipper, a carboxyl-terminal SH3 domain, and several proline-rich regions. Nesca is highly expressed in the brain, is serine phosphorylated, and mobilizes from the cytoplasm to the nuclear membrane in response to neurotrophin, but not epidermal growth factor, stimulation in a MEK-dependent process. Overexpression studies in PC12 cells indicate that Nesca facilitates neurotrophin-dependent neurite outgrowth at nonsaturating doses of nerve growth factor (NGF). Similarly, short interfering RNA studies significantly reduce NGF-dependent neuritogenesis in PC12 cells. Mutational analyses demonstrate that the RUN domain is an important structural determinant for the nuclear translocation of Nesca and that the nuclear redistribution of Nesca is essential to its neurite outgrowth-promoting properties. Collectively, these works provide the first functional characterization of Nesca in the context of neurotrophin signaling and suggest that Nesca serves a novel, nuclear-dependent role in neurotrophin-dependent neurite outgrowth.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas de Transporte/metabolismo , Fatores de Crescimento Neural/metabolismo , Neuritos/metabolismo , Membrana Nuclear/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Proteínas de Transporte/genética , Ativação Enzimática , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator de Crescimento Neural/metabolismo , Sinais de Localização Nuclear , Células PC12 , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Distribuição Tecidual
8.
J Mol Neurosci ; 67(1): 97-110, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30547417

RESUMO

Brain-derived neurotrophic factor (BDNF) facilitates multiple aspects of neuronal differentiation and cellular physiology by activating the high-affinity receptor tyrosine kinase, TrkB. While it is known that both BDNF and TrkB modulate cellular processes involved in learning and memory, exactly how TrkB cross-talks and modulates signaling downstream of excitatory ionotropic receptors, such as the NMDA receptor (NMDAR), are not well understood. A model that we have investigated involves the signaling molecule RasGrf1, a guanine nucleotide exchange factor for both Ras and Rac. We previously identified RasGrf1 as a novel Trk binding partner that facilitates neurite outgrowth in response to both nerve growth factor (NGF) (Robinson et al. in J Biol Chem 280:225-235, 2005) and BDNF (Talebian et al. in J Mol Neurosci 49:38-51, 2013); however, RasGrf1 can also bind the NR2B subunit of the NMDAR (Krapivinsky et al. in Neuron 40:775-784, 2003) and stimulate long-term depression (LTD) (Li et al. in J Neurosci 26:1721-1729, 2006). We have addressed a model that TrkB facilitates learning and memory via two processes. First, TrkB uncouples RasGrf1 from NR2B and facilitates a decrease in NMDA signaling associated with LTD (p38-MAPK). Second, the recruitment of RasGrf1 to TrkB enhances neurite outgrowth and pERK activation and signaling associated with learning and memory. We demonstrate that NMDA recruits RasGrf1 to NR2B; however, co-stimulation with BDNF uncouples this association and recruits RasGrf1 to TrkB. In addition, activation of TrkB stimulates the tyrosine phosphorylation of RasGrf1 which increases neurite outgrowth (Talebian et al. in J Mol Neurosci 49:38-51, 2013), and the tyrosine phosphorylation of NR2B (Tyr1472) (Nakazawa et al. in J Biol Chem 276:693-699, 2001) which facilitates NMDAR cell surface retention (Zhang et al. in J Neurosci 28:415-24, 2008). Collectively, these data demonstrate that TrkB alters NMDA signaling by a dual mechanism that uncouples LTD and, in turn, stimulates neuronal growth and the signaling pathways associated with learning and memory.


Assuntos
Encéfalo/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Encéfalo/fisiologia , Células HEK293 , Humanos , Depressão Sináptica de Longo Prazo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Ligação Proteica , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , ras-GRF1/metabolismo
9.
Biochim Biophys Acta ; 1763(4): 366-80, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16697063

RESUMO

We have investigated the signaling properties of the fibroblast growth factor (FGF) receptor substrate 3 (FRS3), also known as SNT-2 or FRS2beta, in neurotrophin-dependent differentiation in comparison with the related adapter FRS2 (SNT1 or FRS2alpha). We demonstrate that FRS3 binds all neurotrophin Trk receptor tyrosine kinases and becomes tyrosine phosphorylated in response to NGF, BDNF, NT-3 and FGF stimulation in transfected cells and/or primary cortical neurons. Second, the signaling molecules Grb2 and Shp2 bind FRS3 at consensus sites that are highly conserved among FRS family members and that Shp2, in turn, becomes tyrosine phosphorylated. While FRS3 over-expression in PC12 cells neither increases NGF-induced neuritogenesis nor activation of Map kinase/AKT, comparable to previous reports on FRS2, over-expression of a chimeric adapter containing the PH/PTB domains of the insulin receptor substrate (IRS) 2, in place of the PTB domain of FRS3 (IRS2-FRS3) supports insulin-dependent Map kinase activation and neurite outgrowth in PC12 cells. Collectively, these data demonstrate that FRS3 supports ligand-induced Map kinase activation and that the chimeric IRS2-FRS3 adapter is stimulating sufficient levels of activated MapK to support neurite outgrowth in PC12 cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores de Crescimento de Fibroblastos/fisiologia , Fatores de Crescimento Neural/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Células PC12 , Ratos , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Receptor trkC/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Especificidade por Substrato/fisiologia
10.
Mol Cell Biol ; 36(20): 2596-611, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27503856

RESUMO

Macropinocytosis is a normal cellular process by which cells internalize extracellular fluids and nutrients from their environment and is one strategy that Ras-transformed pancreatic cancer cells use to increase uptake of amino acids to meet the needs of rapid growth. Paradoxically, in non-Ras transformed medulloblastoma brain tumors, we have shown that expression and activation of the receptor tyrosine kinase TrkA overactivates macropinocytosis, resulting in the catastrophic disintegration of the cell membrane and in tumor cell death. The molecular basis of this uncontrolled form of macropinocytosis has not been previously understood. Here, we demonstrate that the overactivation of macropinocytosis is caused by the simultaneous activation of two TrkA-mediated pathways: (i) inhibition of RhoB via phosphorylation at Ser(185) by casein kinase 1, which relieves actin stress fibers, and (ii) FRS2-scaffolded Src and H-Ras activation of RhoA, which stimulate actin reorganization and the formation of lamellipodia. Since catastrophic macropinocytosis results in brain tumor cell death, improved understanding of the mechanisms involved will facilitate future efforts to reprogram tumors, even those resistant to apoptosis, to die.


Assuntos
Caseína Quinase I/metabolismo , Neoplasias Cerebelares/metabolismo , Meduloblastoma/metabolismo , Pinocitose , Receptor trkA/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo , Actinas/metabolismo , Morte Celular , Linhagem Celular Tumoral , Humanos , Fosforilação , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Serina/metabolismo , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo
11.
Oncogene ; 22(54): 8774-85, 2003 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-14647472

RESUMO

TrkA is the receptor tyrosine kinase (RTK) for nerve growth factor (NGF) and stimulates NGF-dependent cell survival and differentiation in primary neurons and also differentiation of neuroblastomas and apoptosis of medulloblastomas. We have previously shown that aspartic acid and glutamic acid substitution (AspGlu and GluAsp) of the activation loop tyrosines in TrkA (Tyr(683) and Tyr(684)) supports NGF-independent neuritogenesis and cell survival in PC12 cell-derived nnr5 cells. In this study, the AspGlu and GluAsp mutant Trks have been analysed for their ability to support NGF-independent and NGF-dependent neuritogenesis, proliferation and cell signalling in the human neuroblastoma cell line, SY5Y. We find that the AspGlu and GluAsp mutant Trks support NGF-dependent, but not NGF-independent, autophosphorylation, neuritogenic responses and/or inhibit cell cycle progression. The NGF-dependent neuritogenic responses are lower for the mutant Trks (approximately 30-60% for AspGlu and 50-60% for GluAsp), relative to wild-type TrkA. While both the AspGlu and GluAsp mutant Trks support NGF-dependent transient phosphorylation of Shc, PLCgamma-1, AKT, FRS2, SH2B as well as prolonged MAP kinase activation, the GluAsp mutant induces stronger NGF-dependent tyrosine phosphorylation of FRS2 and SH2B, as well as a stronger reduction in bromodeoxyuridine (BrdU) incorporation. Collectively, these data suggest that neither absolute levels of receptor autophosphorylation, high levels of TrkA expression nor the activation of a specific signalling pathway is dominant and absolutely essential for neuritogenesis and cell cycle arrest of SY5Y cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/química , Proteínas de Membrana/química , Fator de Crescimento Neural/fisiologia , Proteínas Serina-Treonina Quinases , Receptor trkA , Tirosina/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Substituição de Aminoácidos , Animais , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Ciclo Celular , Diferenciação Celular , Linhagem Celular Tumoral , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neuritos/fisiologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Fosfolipase C gama , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Coelhos , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Fosfolipases Tipo C/metabolismo
12.
J Mol Neurosci ; 55(3): 663-77, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25159185

RESUMO

The neurotrophins are a family of closely related growth factors that regulate proliferation and differentiation in the developing and mature nervous systems. Neurotrophins stimulate a family of receptor tyrosine kinases (Trk receptors) and utilize an intracellular docking protein termed fibroblast growth factor (FGF) receptor substrate 2 (FRS2) as a major downstream adapter to activate Ras, phosphatidylinositide 3-kinase (PI3K), and mitogen-activated protein kinase (MAPK) signaling cascades. The goals of this study were twofold: first, to investigate the complexity of neurotrophin-induced FRS2 interactions in primary cortical neurons and to determine which pathway(s) are important in regulating neuronal growth and, second, to determine whether the related signaling adapter, FRS3, stimulates neuron growth comparable to FRS2. We find that neurotrophin treatment of primary cortical neurons stimulates the tyrosine phosphorylation of FRS2 and the subsequent recruitment of Shp2, Grb2, and Gab2. With FRS2 mutants deficient in Grb2 or Shp2 binding, we demonstrate that FRS2 binds Gab1 and Gab2 through Grb2, providing an alternative route to activate PI3 kinase and Shp2. Using recombinant adenoviruses expressing FRS2, we demonstrate that FRS2 overexpression promotes neurite outgrowth and branching in cortical neurons relative to controls. In contrast, overexpression of FRS3 does not stimulate neuronal growth. Moreover, we find that while loss of Shp2, but not Grb2, reduces brain-derived neurotrophic factor (BDNF)-induced MAPK activation, the loss of either pathway impairs neuronal growth. Collectively, these experiments demonstrate that FRS2 functions as an adapter of a multiprotein complex that is activated by the Trk receptors and that the activation of both Grb2- and Shp2-dependent pathways facilitates cortical neuronal growth.


Assuntos
Córtex Cerebral/metabolismo , Proteína Adaptadora GRB2/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Animais , Células COS , Células Cultivadas , Córtex Cerebral/citologia , Chlorocebus aethiops , Proteína Adaptadora GRB2/genética , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/genética , Camundongos , Neurônios/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética
13.
Mol Brain ; 8: 41, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26170135

RESUMO

Alzheimer's disease (AD) is characterized by the deposition of Beta-Amyloid (Aß) peptides in the brain. Aß peptides are generated by cleavage of the Amyloid Precursor Protein (APP) by the ß - and γ - secretase enzymes. Although this process is tightly linked to the internalization of cell surface APP, the compartments responsible are not well defined. We have found that APP can be rapidly internalized from the cell surface to lysosomes, bypassing early and late endosomes. Here we show by confocal microscopy and electron microscopy that this pathway is mediated by macropinocytosis. APP internalization is enhanced by antibody binding/crosslinking of APP suggesting that APP may function as a receptor. Furthermore, a dominant negative mutant of Arf6 blocks direct transport of APP to lysosomes, but does not affect classical endocytosis to endosomes. Arf6 expression increases through the hippocampus with the development of Alzheimer's disease, being expressed mostly in the CA1 and CA2 regions in normal individuals but spreading through the CA3 and CA4 regions in individuals with pathologically diagnosed AD. Disruption of lysosomal transport of APP reduces both Aß40 and Aß42 production by more than 30 %. Our findings suggest that the lysosome is an important site for Aß production and that altering APP trafficking represents a viable strategy to reduce Aß production.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Peptídeos beta-Amiloides/biossíntese , Lisossomos/metabolismo , Pinocitose , Fator 1 de Ribosilação do ADP/metabolismo , Fator 6 de Ribosilação do ADP , Actinas/metabolismo , Oxirredutases do Álcool/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Compartimento Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Sobrevivência Celular , Reagentes de Ligações Cruzadas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dextranos/metabolismo , Endossomos/metabolismo , Endossomos/ultraestrutura , Técnicas de Silenciamento de Genes , Hipocampo/metabolismo , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Lisossomos/ultraestrutura , Camundongos , Proteínas Mutantes/metabolismo , Transporte Proteico , RNA Interferente Pequeno/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
14.
Endocrinology ; 144(3): 922-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12586769

RESUMO

The thyroid TRK oncogenes are generated by chromosomal rearrangements juxtaposing the neurotrophic tyrosine receptor kinase type 1 (NTRK1) tyrosine kinase domain to foreign activating sequences. TRK oncoproteins display a constitutive tyrosine kinase activity resulting in the capability to transform NIH3T3 cells. The TRK oncoproteins' signal transduction has been in part elucidated, and it involves several signal transducers activated by the NGF-stimulated NTRK1 receptor. In this paper, we investigate the role of FRS2 and FRS3, two related adapter proteins activated by fibroblast growth factor and NTRK1 receptors, in the signaling of the thyroid TRK-T1 and TRK-T3 oncogenes. By a combination of in vitro and in vivo assays, we demonstrate that both fibroblast growth factor receptor substrate (FRS)2 and FRS3 are recruited and activated by TRK-T1 and TRK-T3. Interaction studies using different TRK-T3 mutants indicate that FRS3 is recruited by the same tyrosine residue interacting with Shc and FRS2. Expression studies show different expression patterns of the FRS adapters in normal and tumor thyroid samples: FRS3 is expressed in both normal and thyroid tumor samples, whereas FRS2 is not expressed in normal thyroid but is differentially expressed in some tumors. Altogether, our data indicate that the FRS2 and FRS3 adapters may have a role in thyroid carcinogenesis triggered by TRK oncogenes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/metabolismo , Lipoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Receptor trkA/fisiologia , Glândula Tireoide/química , Células 3T3 , Animais , Western Blotting , Proteínas de Transporte/genética , Fatores de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Técnicas de Imunoadsorção , Lipoproteínas/genética , Proteínas de Membrana/genética , Camundongos , Mutagênese Sítio-Dirigida , Fosfoproteínas/genética , RNA Mensageiro/análise , Receptor trkA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Neoplasias da Glândula Tireoide/genética , Transfecção
15.
J Neurotrauma ; 19(12): 1531-41, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12542855

RESUMO

Increased intraspinal nerve growth factor (NGF) after spinal cord injury (SCI) is detrimental to the autonomic nervous system. Autonomic dysreflexia is a debilitating condition characterized by episodic hypertension, intense headache, and sweating. Experimentally, it is associated with aberrant primary afferent sprouting in the dorsal horn that is nerve growth factor (NGF)-dependent. Therapeutic strategies that neutralize NGF may ameliorate initial apoptotic cellular responses to the injury and aberrant afferent plasticity that occurs weeks after the injury. Subsequently, the development of autonomic disorders may be suppressed. We constructed a protein including the extracellular portion of trkA fused to the Fc portion of human IgG and expressed it using a baculovirus system. Binding of our trkA-IgG fusion protein was specific for NGF with a K(d) = 4.26 x 10(-11) M and blocked NGF-dependent neuritogenesis in PC-12 cells. We hypothesized that binding of NGF in the injured cord by our trkA-IgG fusion protein would diminish autonomic dysreflexia. Severe, high thoracic SCI was induced with clip compression and the rats were treated with intrathecal infusions (4 microg/day) of trkA-IgG or control IgG. At 14 days post-SCI, the magnitude of autonomic dysreflexia was assessed. Colon distension increased mean arterial pressure (MAP) in control rats by 46 +/- 2 from 96 +/- 5 mmHg. In contrast, MAP of rats treated with trkA-IgG increased by only 30 +/- 2 mmHg. Likewise, the MAP response to cutaneous stimulation was also reduced in rats treated with trkA-IgG (20 +/- 1 vs. 29 +/- 2). In contrast, trkA-IgG treatment had no effect on heart rate responses during colon distension or cutaneous stimulation. These results indicate that treatment with trkA-IgG to block NGF suppresses the development of autonomic dysreflexia after a clinically relevant spinal cord injury.


Assuntos
Disreflexia Autonômica/prevenção & controle , Proteínas de Transporte/uso terapêutico , Proteínas de Membrana/uso terapêutico , Fator de Crescimento Neural/antagonistas & inibidores , Receptor trkA , Compressão da Medula Espinal/fisiopatologia , Animais , Disreflexia Autonômica/metabolismo , Disreflexia Autonômica/fisiopatologia , Baculoviridae/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Proteínas de Transporte/farmacologia , Células Cultivadas , Colo/inervação , Colo/fisiologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Frequência Cardíaca/efeitos dos fármacos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/farmacologia , Imunoglobulina G/uso terapêutico , Proteínas de Membrana/farmacologia , Fator de Crescimento Neural/imunologia , Células PC12/efeitos dos fármacos , Estimulação Física , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Compressão da Medula Espinal/complicações
16.
J Cereb Blood Flow Metab ; 34(4): 690-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24496171

RESUMO

Tissue pH is an indicator of altered cellular metabolism in diseases including stroke and cancer. Ischemic tissue often becomes acidic due to increased anaerobic respiration leading to irreversible cellular damage. Chemical exchange saturation transfer (CEST) effects can be used to generate pH-weighted magnetic resonance imaging (MRI) contrast, which has been used to delineate the ischemic penumbra after ischemic stroke. In the current study, a novel MRI ratiometric technique is presented to measure absolute pH using the ratio of CEST-mediated contrast from amine and amide protons: amine/amide concentration-independent detection (AACID). Effects of CEST were observed at 2.75 parts per million (p.p.m.) for amine protons and at 3.50 p.p.m. for amide protons downfield (i.e., higher frequency) from bulk water. Using numerical simulations and in vitro MRI experiments, we showed that pH measured using AACID was independent of tissue relaxation time constants, macromolecular magnetization transfer effects, protein concentration, and temperature within the physiologic range. After in vivo pH calibration using phosphorus ((31)P) magnetic resonance spectroscopy ((31)P-MRS), local acidosis is detected in mouse brain after focal permanent middle cerebral artery occlusion. In summary, our results suggest that AACID represents a noninvasive method to directly measure the spatial distribution of absolute pH in vivo using CEST MRI.


Assuntos
Acidose Láctica , Amidas/análise , Aminas/análise , Isquemia Encefálica/metabolismo , Concentração de Íons de Hidrogênio , Imageamento por Ressonância Magnética/métodos , Acidose Láctica/diagnóstico , Acidose Láctica/metabolismo , Animais , Biomarcadores/análise , Calibragem , Simulação por Computador , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Químicos
17.
J Mol Neurosci ; 49(1): 38-51, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22744634

RESUMO

We previously demonstrated that the guanine nucleotide exchange factor, RasGrf1, binds nerve growth factor (NGF)-activated TrkA and facilitates neurotrophin-induced neurite outgrowth in PC12 cells. RasGrf1 can activate both Ras and Rac, via intrinsic Cdc25 and DH domains, respectively, suggesting that the activation of both could contribute to this process. Previous studies have assayed constitutive neurite outgrowth following RasGrf1 over-expression in PC12 cells, in either the absence or presence of ectopic H-Ras, and have suggested an essential role for either Ras or Rac depending on the presence of H-Ras over-expression. In contrast, in this study, we have addressed the mechanism of how RasGrf1 facilitates neurite outgrowth in response to the neurotrophins, NGF and BDNF. Using Ras/Rac activation assays and site-directed RasGrf1 mutants, we find that both Ras and Rac are essential to neurotrophin-induced neurite outgrowth. Moreover, we find that H-Ras over-expression rescues the loss of neurite outgrowth observed with a Rac minus mutant and that RasGrf1 differentially stimulates NGF-dependent activation of Rac or Ras, depending on cell type. Collectively, these studies clarify the mechanism of how RasGrf1 expression facilitates neurotrophin-induced neurite outgrowth. Moreover, they suggest that H-Ras over-expression should be used with caution to measure phenotypic responses.


Assuntos
Neuritos/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , ras-GRF1/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células HEK293 , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Mutação , Fator de Crescimento Neural/metabolismo , Neuritos/metabolismo , Células PC12 , Proteínas Proto-Oncogênicas p21(ras)/genética , Ratos , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Regulação para Cima , ras-GRF1/genética
19.
Glycobiology ; 17(7): 725-34, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17389653

RESUMO

Trypanosome trans-sialidase (TS) is a sialic acid-transferring enzyme and a novel ligand of tyrosine kinase (TrkA) receptors but not of neurotrophin receptor p75NTR. Here, we show that TS targets TrkB receptors on TrkB-expressing pheochromocytoma PC12 cells and colocalizes with TrkB receptor internalization and phosphorylation (pTrkB). Wild-type TS but not the catalytically inactive mutant TSDeltaAsp98-Glu induces pTrkB and mediates cell survival responses against death caused by oxidative stress in TrkA- and TrkB-expressing cells like those seen with nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF). These same effects are not observed in Trk deficient PC12(nnr5) cells, but are re-established in PC12(nnr5) cells stably transfected with TrkA or TrkB, are partially blocked by inhibitors of tyrosine kinase (K-252a), mitogen-activated protein/mitogen-activated kinase (PD98059) and completely blocked by LY294002, an inhibitor of phosphatidylinositol 3-kinase (PI3K). Both TrkA- and TrkB-expressing cells pretreated with TS or their natural ligands are protected against cell death caused by serum/glucose deprivation or from hypoxia-induced neurite retraction. The cell survival effects of NGF and BDNF against oxidative stress are significantly inhibited by the neuraminidase inhibitor, Tamiflu. Together, these observations suggest that trypanosome TS mimics neurotrophic factors in cell survival responses against oxidative stress, hypoxia-induced neurite retraction and serum/glucose deprivation.


Assuntos
Glucose/metabolismo , Glicoproteínas/metabolismo , Neuraminidase/metabolismo , Estresse Oxidativo , Receptor trkB/metabolismo , Soro/metabolismo , Trypanosoma cruzi/metabolismo , Animais , Sobrevivência Celular , Inibidores Enzimáticos/farmacologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Hipóxia , Fator de Crescimento Neural/metabolismo , Oseltamivir/farmacologia , Células PC12 , Ratos
20.
J Biol Chem ; 280(20): 19461-71, 2005 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15753086

RESUMO

The human tumorous imaginal disc 1 (TID1) proteins including TID1(L) and TID1(S), members of the DnaJ domain protein family, are involved in multiple intracellular signaling pathways such as apoptosis induction, cell proliferation, and survival. Here we report that TID1 associates with the Trk receptor tyrosine kinases and regulates nerve growth factor (NGF)-induced neurite outgrowth in PC12-derived nnr5 cells. Binding assays and transfection studies showed that the carboxyl-terminal end of TID1 (residues 224-429) bound to Trk at the activation loop (Tyr(P)(683)-Tyr(684)(P)(684) in rat TrkA) and that TID1 was tyrosine phosphorylated by Trk both in yeast and in transfected cells. Moreover endogenous TID1 was also tyrosine phosphorylated by and co-immunoprecipitated with Trk in neurotrophin-stimulated primary rat hippocampal neurons. Overexpression studies showed that both TID1(L) and TID1(S) significantly facilitated NGF-induced neurite outgrowth in TrkA-expressing nnr5 cells possibly through a mechanism involving increased activation of mitogen-activated protein kinase. Consistently knockdown of endogenous TID1, mediated with specific short hairpin RNA, significantly reduced NGF-induced neurite growth in nnr5-TrkA cells. These data provide the first evidence that TID1 is a novel intracellular adaptor that interacts with the Trk receptor tyrosine kinases in an activity-dependent manner to facilitate Trk-dependent intracellular signaling.


Assuntos
Proteínas de Choque Térmico/metabolismo , Receptor trkA/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , DNA/genética , Proteínas de Choque Térmico HSP40 , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Humanos , Cinética , Neuritos/metabolismo , Neuritos/ultraestrutura , Células PC12 , Fosforilação , Interferência de RNA , Ratos , Receptor trkA/química , Receptor trkA/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Tirosina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA