Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Oncogene ; 25(53): 7086-95, 2006 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-16732321

RESUMO

In patients with medullary thyroid carcinoma (MTC) and type 2A multiple endocrine neoplasia (MEN2A), mutations of cysteine residues in the extracellular juxtamembrane region of the RET receptor tyrosine kinase cause the formation of covalent receptor dimers linked by intermolecular disulfide bonds between unpaired cysteines, followed by oncogenic activation of the RET kinase. The close proximity to the plasma membrane of the affected cysteine residues prompted us to investigate the possible role of the transmembrane (TM) domain of RET (RET-TM) in receptor-receptor interactions underlying dimer formation. Strong self-association of the RET-TM was observed in a biological membrane. Mutagenesis studies indicated the involvement of the evolutionary conserved residues Ser-649 and Ser-653 in RET-TM oligomerization. Unexpectedly, RET-TM interactions were also abrogated in the A639G/A641R double mutant, first identified in a sporadic case of MTC. In agreement with this, no transforming activity could be detected in full-length RET carrying the A639G and A641R mutations, which remained fully responsive to glial cell-line-derived neurotrophic factor (GDNF) stimulation. When introduced in the context of C634R - a cysteine replacement that is prevalent in MEN2A cases - the A639G/A641R mutations significantly reduced dimer formation and transforming activity in this otherwise highly oncogenic RET variant. These data suggest that a strong propensity to self-association in the RET-TM underlies - and may be required for - dimer formation and oncogenic activation of juxtamembrane cysteine mutants of RET, and explains the close proximity to the plasma membrane of cysteine residues implicated in MEN2A and MTC syndromes.


Assuntos
Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Neoplasia Endócrina Múltipla Tipo 2a/genética , Neoplasia Endócrina Múltipla Tipo 2a/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Alanina/genética , Alanina/metabolismo , Sequência de Aminoácidos , Animais , Cisteína/genética , Cisteína/metabolismo , Dimerização , Dissulfetos/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster , Expressão Gênica/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Humanos , Dados de Sequência Molecular , Mutação/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-ret/química , Proteínas Proto-Oncogênicas c-ret/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Neoplasias da Glândula Tireoide/genética
2.
Cell Mol Life Sci ; 61(4): 488-96, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14999407

RESUMO

Using a search engine called Motifer, we searched the public database of the human genome for genes matching a consensus pattern of cysteine residues derived from members of the transforming growth factor-beta (TGF-beta) superfamily. We identified two genes (named MDF451 and MDF628) that display sequence similarity to members of the TGF-beta superfamily in the arrangement of six conserved cysteine residues. Phylogenetic analyses revealed that MDF451 and MDF628 constitute a distinct subgroup within the TGF-beta superfamily, distantly related to the GDNF subfamily of ligands. Both genes could be identified in several primate species in addition to human, including chimpanzee, gorilla, guereza, and green and gray monkey, but not in rodents or other non-primate mammals, and appear not to be present in the genomes of mouse, rat or zebrafish. RNAs for MDF451 and MDF628 were expressed at low levels within distinct regions of the human central nervous system, including adult cerebellum, adult spinal cord and fetal brain. Despite expression at the RNA level, both genes presented a transcribed upstream stop codon that would prevent translation of the TGF-beta-like reading frame. The coding potential of alternative reading frames was not immediately apparent. The two genes may represent TGF-beta-like pseudogenes that have recently appeared in evolution in a common ancestor of the primate lineage by duplication from a GDNF/TGF-beta-like ancestral gene.


Assuntos
Evolução Molecular , Família Multigênica , Primatas/genética , Pseudogenes , Fator de Crescimento Transformador beta/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Humanos , Dados de Sequência Molecular , Sistema Nervoso/metabolismo , Filogenia , Primatas/metabolismo
3.
Genes Dev ; 15(15): 2010-22, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11485994

RESUMO

Nodal proteins have crucial roles in mesendoderm formation and left-right patterning during vertebrate development. The molecular mechanisms of signal transduction by Nodal and related ligands, however, are not fully understood. In this paper, we present biochemical and functional evidence that the orphan type I serine/threonine kinase receptor ALK7 acts as a receptor for mouse Nodal and Xenopus Nodal-related 1 (Xnr1). Receptor reconstitution experiments indicate that ALK7 collaborates with ActRIIB to confer responsiveness to Xnr1 and Nodal. Both receptors can independently bind Xnr1. In addition, Cripto, an extracellular protein genetically implicated in Nodal signaling, can independently interact with both Xnr1 and ALK7, and its expression greatly enhances the ability of ALK7 and ActRIIB to respond to Nodal ligands. The Activin receptor ALK4 is also able to mediate Nodal signaling but only in the presence of Cripto, with which it can also interact directly. A constitutively activated form of ALK7 mimics the mesendoderm-inducing activity of Xnr1 in Xenopus embryos, whereas a dominant-negative ALK7 specifically blocks the activities of Nodal and Xnr1 but has little effect on other related ligands. In contrast, a dominant-negative ALK4 blocks all mesoderm-inducing ligands tested, including Nodal, Xnr1, Xnr2, Xnr4, and Activin. In agreement with a role in Nodal signaling, ALK7 mRNA is localized to the ectodermal and organizer regions of Xenopus gastrula embryos and is expressed during early stages of mouse embryonic development. Therefore, our results indicate that both ALK4 and ALK7 can mediate signal transduction by Nodal proteins, although ALK7 appears to be a receptor more specifically dedicated to Nodal signaling.


Assuntos
Embrião não Mamífero/fisiologia , Fator de Crescimento Epidérmico , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio , Glicoproteínas de Membrana , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/metabolismo , Fatores de Transcrição , Fator de Crescimento Transformador beta/fisiologia , Proteínas de Xenopus , Xenopus laevis/embriologia , Receptores de Ativinas , Receptores de Ativinas Tipo I , Sequência de Aminoácidos , Animais , Anormalidades Congênitas/embriologia , Anormalidades Congênitas/genética , Ectoderma/fisiologia , Proteínas Ligadas por GPI , Gástrula/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Ligantes , Proteínas de Membrana , Camundongos , Dados de Sequência Molecular , Morfogênese , Proteínas de Neoplasias/metabolismo , Proteína Nodal , Filogenia , Proteínas Serina-Treonina Quinases/química , RNA Mensageiro/genética , Receptores de Fatores de Crescimento/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Vertebrados/embriologia
4.
J Biol Chem ; 276(38): 35808-17, 2001 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-11445581

RESUMO

Using bioinformatic tools, mutagenesis, and binding studies, we have investigated the structural organization of the extracellular region of the RET receptor tyrosine kinase, a functional receptor for glial cell line-derived neurotrophic factor (GDNF). Multiple sequence alignments of seven vertebrate sequences and one invertebrate RET sequence delineated four distinct N-terminal domains, each of about 110 residues, containing many of the consensus motifs of the cadherin fold. Based on these alignments and the crystal structures of epithelial and neural cadherins, we have generated molecular models of each of the four cadherin-like domains in the extracellular region of human RET. The modeled structures represent realistic models from both energetic and geometrical points of view and are consistent with previous observations gathered from biochemical analyses of the effects of Hirschsprung's disease mutations affecting the folding and stability of the RET molecule, as well as our own site-directed mutagenesis studies of RET cadherin-like domain 1. We have also investigated the role of Ca(2+) in ligand binding by RET and found that Ca(2+) ions are required for RET binding to GDNF but not for GDNF binding to the GFRalpha1 co-receptor. In agreement with these results, RET, but not GFRalpha1, was found to bind Ca(2+) directly. Our results indicate that the overall architecture of the extracellular region of RET is more closely related to cadherins than previously thought. The models of the cadherin-like domains of human RET represent valuable tools with which to guide future site-directed mutagenesis studies aimed at identifying residues involved in ligand binding and receptor activation.


Assuntos
Caderinas/metabolismo , Cálcio/metabolismo , Proteínas de Drosophila , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Caderinas/química , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Filogenia , Conformação Proteica , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ret , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/genética , Homologia de Sequência de Aminoácidos
5.
Mol Cell Neurosci ; 18(1): 56-67, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11461153

RESUMO

Neurotrophin-4 (NT-4) is produced by slow muscle fibers in an activity-dependent manner and promotes growth and remodeling of adult motorneuron innervation. However, both muscle fibers and motor neurons express NT-4 receptors, suggesting bidirectional NT-4 signaling at the neuromuscular junction. Mice lacking NT-4 displayed enlarged and fragmented neuromuscular junctions with disassembled postsynaptic acetylcholine receptor (AChR) clusters, reduced AChR binding, and acetylcholinesterase activity. Electromyographic responses, posttetanic potentiation, and action potential amplitude were also significantly reduced in muscle fibers from NT-4 knock-out mice. Slow-twitch soleus muscles from these mice fatigued twice as rapidly as those from wild-type mice during repeated tetanic stimulation. Thus, muscle-derived NT-4 is required for maintenance of postsynaptic AChR regions, normal muscular electrophysiological responses, and resistance to muscle fatigue. This neurotrophin may therefore be a key component of an activity-dependent feedback mechanism regulating maintenance of neuromuscular connections and muscular performance.


Assuntos
Fadiga Muscular/fisiologia , Músculo Esquelético/fisiologia , Fatores de Crescimento Neural/genética , Junção Neuromuscular/fisiologia , Acetilcolinesterase/metabolismo , Fatores Etários , Animais , Eletromiografia , Camundongos , Camundongos Knockout , Neurônios Motores/fisiologia , Contração Muscular/fisiologia , Fibras Musculares de Contração Lenta/enzimologia , Músculo Esquelético/citologia , Músculo Esquelético/inervação , Receptores Colinérgicos/metabolismo
6.
Neuron ; 29(1): 171-84, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11182089

RESUMO

Although both c-Ret and GFRalpha1 are required for responsiveness to GDNF, GFRalpha1 is widely expressed in the absence of c-Ret, suggesting alternative roles for "ectopic" sites of GFRalpha1 expression. We show that GFRalpha1 is released by neuronal cells, Schwann cells, and injured sciatic nerve. c-Ret stimulation in trans by soluble or immobilized GFRalpha1 potentiates downstream signaling, neurite outgrowth, and neuronal survival, and elicits dramatic localized expansions of axons and growth cones. Soluble GFRalpha1 mediates robust recruitment of c-Ret to lipid rafts via a novel mechanism requiring the c-Ret tyrosine kinase. Activated c-Ret associates with different adaptor proteins inside and outside lipid rafts. These results provide an explanation for the tissue distribution of GFRalpha1, supporting the physiological importance of c-Ret activation in trans as a novel mechanism to potentiate and diversify the biological responses to GDNF.


Assuntos
Proteínas de Drosophila , Microdomínios da Membrana/metabolismo , Fatores de Crescimento Neural , Neurônios/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Animais , Axônios/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/metabolismo , Meios de Cultivo Condicionados/farmacologia , Inibidores Enzimáticos/farmacologia , Matriz Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Glicosilfosfatidilinositóis/metabolismo , Cones de Crescimento/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Compressão Nervosa , Proteínas do Tecido Nervoso/farmacologia , Neurônios/citologia , Estrutura Terciária de Proteína/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Proto-Oncogênicas c-ret , Ratos , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/farmacologia , Células de Schwann/citologia , Células de Schwann/metabolismo , Nervo Isquiático/citologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/metabolismo
7.
J Biol Chem ; 276(2): 1450-8, 2001 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-11018032

RESUMO

The glial cell line-derived neurotrophic factor (GDNF) family comprise a subclass of cystine-knot superfamily ligands that interact with a multisubunit receptor complex formed by the c-Ret tyrosine kinase and a cystine-rich glycosyl phosphatidylinositol-anchored binding subunit called GDNF family receptor alpha (GFRalpha). All four GDNF family ligands utilize c-Ret as a common signaling receptor, whereas specificity is conferred by differential binding to four distinct GFRalpha homologues. To understand how the different GFRalphas discriminate ligands, we have constructed a large set of chimeric and truncated receptors and analyzed their ligand binding and signaling capabilities. The major determinant of ligand binding was found in the most conserved region of the molecule, a central domain predicted to contain four conserved alpha helices and two beta strands. Distinct hydrophobic and positively charged residues in this central region were required for binding of GFRalpha1 to GDNF. Interaction of GFRalpha1 and GFRalpha2 with GDNF and neurturin required distinct subsegments within this central domain, which allowed the construction of chimeric receptors that responded equally well to both ligands. C-terminal segments adjacent to the central domain are necessary and have modulatory function in ligand binding. In contrast, the N-terminal domain was dispensable without compromising ligand binding specificity. Ligand-independent interaction with c-Ret also resides in the central domain of GFRalpha1, albeit within a distinct and smaller region than that required for ligand binding. Our results indicate that the central region of this class of receptors constitutes a novel binding domain for cystine-knot superfamily ligands.


Assuntos
Proteínas de Drosophila , Fatores de Crescimento Neural , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Linhagem Celular , Galinhas , Chlorocebus aethiops , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Ligantes , Camundongos , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ret , Ratos , Receptores Proteína Tirosina Quinases/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Transfecção
8.
J Biol Chem ; 276(7): 5140-6, 2001 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-11084022

RESUMO

The signaling capabilities and biological functions of activin receptor-like kinase 7 (ALK7), a type I receptor serine/threonine kinase predominantly expressed in the nervous system, are unknown. We have constructed a cell line derived from the rat pheochromocytoma PC12 in which expression of a constitutively active mutant of ALK7 (T194D) is under the control of a tetracycline-inducible promoter. For comparison, another cell line was engineered with tetracycline-regulated expression of a constitutively active variant of the transforming growth factor-beta type I receptor ALK5. Expression of activated ALK7 in PC12 cells resulted in activation of Smad2 and Smad3, but not Smad1, as well as the mitogen-activated protein kinases extracellular signal-regulated kinase and c-Jun N-terminal kinase. Reporter assays demonstrated that ALK7 activation stimulates transcription from the Smad-binding element of the Jun-B gene, the plasminogen activator inhibitor-1 gene, and AP-1 elements. In addition, ALK7 activation induced expression of endogenous gene products, including Smad7, c-fos mRNA, and plasminogen activator inhibitor-1. Thymidine incorporation assays revealed an anti-proliferative effect of ALK7 activation in PC12 cells, which correlated with increased transcription from the promoters of cycline-dependent kinase inhibitors p15(INK4B) and p21. Unexpectedly, ALK7 signaling produced a remarkable change in cell morphology characterized by cell flattening and elaboration of blunt, short cell processes. Interestingly, no such changes were observed upon induction of activated ALK5. The alterations in cell morphology upon ALK7 activation were more pronounced in cultures grown in full serum, were accompanied by rearrangements of actin filaments, and were maintained for several days after withdrawal of treatment. PC12 cultures that had been "primed" in this way showed an accelerated and augmented differentiation response to nerve growth factor. These results indicate that ALK7 may participate in the control of proliferation of neuronal precursors and morphological differentiation of postmitotic neurons.


Assuntos
Receptores de Ativinas Tipo I , Neurônios/citologia , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais , Actinas/metabolismo , Receptores de Ativinas , Animais , Diferenciação Celular , Divisão Celular , DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Doxiciclina/farmacologia , Sistema de Sinalização das MAP Quinases , Mutação , Neurônios/metabolismo , Células PC12 , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/biossíntese , Ratos , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad2 , Proteína Smad3 , Transativadores/metabolismo , Ativação Transcricional , Transfecção , Fator de Crescimento Transformador beta/genética
9.
Development ; 127(21): 4531-40, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11023857

RESUMO

Brain derived neurotrophic factor, BDNF, is a neurotrophin best characterized for its survival and differentiative effects on neurons expressing the trk B receptor tyrosine kinase. Although many of these neurons are lost in the BDNF(-)(/)(- )mouse, the early postnatal lethality of these animals suggests a wider function for this growth factor. Here, we demonstrate that deficient expression of BDNF impairs the survival of endothelial cells in intramyocardial arteries and capillaries in the early postnatal period, although the embryonic vasculature can remodel into arteries, capillaries and veins. BDNF deficiency results in a reduction in endothelial cell-cell contacts and in endothelial cell apoptosis, leading to intraventricular wall hemorrhage, depressed cardiac contractility and early postnatal death. Vascular hemorrhage is restricted to cardiac vessels, reflecting the localized expression of BDNF and trk B by capillaries and arterioles in this vascular bed. Conversely, ectopic BDNF overexpression in midgestational mouse hearts results in an increase in capillary density. Moreover, BDNF activation of endogenous trk B receptors supports the survival of cardiac microvascular endothelial cells cultured from neonatal mice. These results establish an essential role for BDNF in maintaining vessel stability in the heart through direct angiogenic actions on endothelial cells.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Vasos Coronários/fisiologia , Endotélio Vascular/citologia , Cardiopatias Congênitas/genética , Animais , Animais Recém-Nascidos , Apoptose , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/genética , Capilares/crescimento & desenvolvimento , Capilares/fisiologia , Comunicação Celular , Sobrevivência Celular , Circulação Coronária , Vasos Coronários/crescimento & desenvolvimento , Cruzamentos Genéticos , Endotélio Vascular/fisiologia , Coração/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor trkB/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
J Biol Chem ; 275(50): 39159-66, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-10995764

RESUMO

Proximal signaling events and protein-protein interactions initiated after activation of the c-Ret receptor tyrosine kinase by its ligand, glial cell line-derived neurotrophic factor (GDNF), were investigated in cells carrying native and mutated forms of this receptor. Mutation of Tyr-1062 (Y1062F) in the cytoplasmic tail of c-Ret abolished receptor binding and phosphorylation of the adaptor Shc and eliminated activation of Ras by GDNF. Phosphorylation of Erk kinases was also greatly attenuated but not eliminated by this mutation. This residual wave of Erk phosphorylation was independent of the kinase activity of c-Ret. Mutation of Tyr-1096 (Y1096F), a binding site for the adaptor Grb2, had no effect on Erk activation by GDNF. Activation of phosphatidylinositol-3 kinase (PI3K) and its downstream effector Akt was also reduced in the Y1062F mutant but not completely abolished unless Tyr-1096 was also mutated. Ligand stimulation of neuronal cells induced the assembly of a large protein complex containing c-Ret, Grb2, and tyrosine-phosphorylated forms of Shc, p85(PI3K), the adaptor Gab2, and the protein-tyrosine phosphatase SHP-2. In agreement with Ras-independent activation of PI3K by GDNF in neuronal cells, survival of sympathetic neurons induced by GDNF was dependent on PI3K but was not affected by microinjection of blocking anti-Ras antibodies, which did compromise neuronal survival by nerve growth factor, suggesting that Ras is not required for GDNF-induced survival of sympathetic neurons. These results indicate that upon ligand stimulation, at least two distinct protein complexes assemble on phosphorylated Tyr-1062 of c-Ret via Shc, one leading to activation of the Ras/Erk pathway through recruitment of Grb2/Sos and another to the PI3K/Akt pathway through recruitment of Grb2/Gab2 followed by p85(PI3K) and SHP-2. This latter complex can also assemble directly onto phosphorylated Tyr-1096, offering an alternative route to PI3K activation by GDNF.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Proteínas de Drosophila , Fatores de Crescimento Neural , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Proteínas ras/metabolismo , Células 3T3 , Animais , Sítios de Ligação , Western Blotting , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , Citoplasma/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fibroblastos/metabolismo , Proteína Adaptadora GRB2 , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Glutationa Transferase/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Morfolinas/farmacologia , Mutagênese Sítio-Dirigida , Mutação , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Fosforilação , Testes de Precipitina , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-ret , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Adaptadoras da Sinalização Shc , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Fatores de Tempo , Tirosina/química
11.
J Biol Chem ; 275(50): 39427-34, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-10958791

RESUMO

Four members of the glial cell line-derived neurotrophic factor family have been identified (GDNF, neurturin, persephin, and enovin/artemin). They bind to a specific membrane-anchored GDNF family receptor as follows: GFRalpha-1 for GDNF, GFRalpha-2 for neurturin, GFRalpha-3 for enovin/artemin, and (chicken) GFRalpha-4 for persephin. Subsequent signaling occurs through activation of a common transmembrane tyrosine kinase, cRET. GFRalpha-4, the coreceptor for persephin, was previously identified in chicken only. We describe the cloning and characterization of a mammalian persephin receptor GFRalpha-4. The novel GFRalpha receptor is substantially different in sequence from all known GFRalphas, including chicken GFRalpha-4, and lacks the first cysteine-rich domain present in all previously characterized GFRalphas. At least two different GFRalpha-4 splice variants exist in rat tissues, differing at their respective COOH termini. GFRalpha-4 mRNA is expressed at low levels in different brain areas in the adult as well as in some peripheral tissues including testis and heart. Recombinant rat GFRalpha-4 variants were expressed in mammalian cells and shown to be at least partially secreted from the cells. Persephin binds specifically and with high affinity (K(D) = 6 nm) to the rat GFRalpha-4 receptor, but no cRET activation could be demonstrated. Although the newly characterized mammalian GFRalpha-4 receptor is structurally divergent from previously characterized GFRalpha family members, we suggest that it is a mammalian orthologue of the chicken persephin receptor. This discovery will allow a more detailed investigation of the biological targets of persephin action and its potential involvement in diseases of the nervous system.


Assuntos
Proteínas Aviárias , Proteínas de Drosophila , Glicoproteínas de Membrana/metabolismo , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Fator de Crescimento Neural , Processamento Alternativo , Sequência de Aminoácidos , Animais , Northern Blotting , Western Blotting , Encéfalo/metabolismo , Células CHO , Galinhas , Mapeamento Cromossômico , Clonagem Molecular , Cricetinae , Cisteína/química , DNA Complementar/metabolismo , Embrião de Mamíferos/metabolismo , Embrião não Mamífero , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Hibridização In Situ , Cinética , Glicoproteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ret , RNA Mensageiro/metabolismo , Ratos , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Superfície Celular/química , Proteínas Recombinantes/metabolismo , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Distribuição Tecidual , Transfecção
13.
EMBO J ; 18(21): 5901-10, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10545102

RESUMO

Ligand-induced receptor oligomerization is a widely accepted mechanism for activation of cell-surface receptors. We investigated ligand-receptor interactions in the glial cell-line derived neurotrophic factor (GDNF) receptor complex, formed by the c-Ret receptor tyrosine kinase and the glycosylphosphatidylinositol (GPI)-anchored subunit GDNF family receptor alpha-1 (GFRalpha1). As only GFRalpha1 can bind GDNF directly, receptor complex formation is thought to be initiated by GDNF binding to this receptor. Here we identify an interface in GDNF formed by exposed acidic and hydrophobic residues that is critical for binding to GFRalpha1. Unexpectedly, several GDNF mutants deficient in GFRalpha1 binding retained the ability to bind and activate c-Ret at normal levels. Although impaired in binding GFRalpha1 efficiently, these mutants still required GFRalpha1 for c-Ret activation. These findings support a role for c-Ret in ligand binding and indicate that GDNF does not initiate receptor complex formation, but rather interacts with a pre-assembled GFRalpha1- c-Ret complex.


Assuntos
Proteínas de Drosophila , Fatores de Crescimento Neural , Proteínas do Tecido Nervoso/química , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Reagentes de Ligações Cruzadas , Dimerização , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Camundongos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Fosforilação , Fosfotirosina/análise , Ligação Proteica/genética , Conformação Proteica , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas c-ret , Relação Estrutura-Atividade
14.
J Biol Chem ; 274(30): 20885-94, 1999 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-10409632

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) has been shown to signal through a multicomponent receptor complex consisting of the Ret receptor tyrosine kinase and a member of the GFRalpha family of glycosylphosphatidylinositol-anchored receptors. In the current model of GDNF signaling, Ret delivers the intracellular signal but cannot bind ligand on its own, while GFRalphas bind ligand but are thought not to signal in the absence of Ret. We have compared signaling pathways activated by GDNF in two neuronal cell lines expressing different complements of GDNF receptors. In a motorneuron-derived cell line expressing Ret and GFRalphas, GDNF stimulated sustained activation of the Ras/ERK and phosphatidylinositol 3-kinase/Akt pathways, cAMP response element-binding protein phosphorylation, and increased c-fos expression. Unexpectedly, GDNF also promoted biochemical and biological responses in a line of conditionally immortalized neuronal precursors that express high levels of GFRalphas but not Ret. GDNF treatment did not activate the Ras/ERK pathway in these cells, but stimulated a GFRalpha1-associated Src-like kinase activity in detergent-insoluble membrane compartments, rapid phosphorylation of cAMP response element-binding protein, up-regulation of c-fos mRNA, and cell survival. Together, these results offer new insights into the dynamics of GDNF signaling in neuronal cells, and indicate the existence of novel signaling mechanisms directly or indirectly mediated by GFRalpha receptors acting in a cell-autonomous manner independently of Ret.


Assuntos
Proteínas de Drosophila , Proteínas do Tecido Nervoso/farmacologia , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/fisiologia , Proteínas Proto-Oncogênicas c-ret , Ratos
15.
Mol Reprod Dev ; 53(3): 274-81, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10369388

RESUMO

Previous studies reported the presence of choline acetyltransferase (ChAT) mRNA and protein in the mammalian testis. We have now found that none of the ChAT mRNAs produced in the testis is capable of encoding a full-length ChAT protein. Two ChAT cDNAs were isolated from an adult rat testis cDNA library encoding N-terminally truncated ChAT proteins of 450 and 414 amino acids (aa), respectively, the former containing a novel N-terminal extension of 69 residues. Rapid Amplification of cDNA Ends (RACE) analysis revealed a complex pattern of 5' untranslated mRNA termini generated from the ChAT gene locus in the testis, all representing truncated versions of the ChAT enzyme. Two of these proteins were produced in transfected fibroblasts and found to lack ChAT activity. Neither did they show binding to the ChAT substrates, acetyl CoA and choline, in a competition assay. These results indicate that mammalian testis lacks a bona fide ChAT enzyme but expresses truncated ChAT proteins with a possible unique function to the testis.


Assuntos
Colina O-Acetiltransferase/genética , Testículo/enzimologia , Regiões 5' não Traduzidas , Animais , Sequência de Bases , Células COS , Colina O-Acetiltransferase/biossíntese , Colina O-Acetiltransferase/metabolismo , Clonagem Molecular , DNA Complementar , Masculino , Dados de Sequência Molecular , Neurônios/enzimologia , Biossíntese de Proteínas , RNA Mensageiro , Ratos , Ratos Sprague-Dawley , Ribonucleases
17.
J Neurosci ; 19(9): 3507-18, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10212310

RESUMO

Brain-derived neurotrophic factor (BDNF) is a survival factor for different classes of neurons, including gustatory neurons. We have studied innervation and development of the gustatory system in transgenic mice overexpressing BDNF under the control of regulatory sequences from the nestin gene, an intermediate filament gene expressed in precursor cells of the developing nervous system and muscle. In transgenic mice, the number and size of gustatory papillae were decreased, circumvallate papillae had a deranged morphology, and there was also a severe loss of lingual taste buds. Paradoxically, similar deficits have been found in BDNF knock-out mice, which lack gustatory neurons. However, the number of neurons in gustatory ganglia was increased in BDNF-overproducing mice. Although gustatory fibers reached the tongue in normal numbers, the amount and density of nerve fibers in gustatory papillae were reduced in transgenic mice compared with wild-type littermates. Gustatory fibers appeared stalled at the base of the tongue, a site of ectopic BDNF expression, where they formed abnormal branches and sprouts. Interestingly, palatal taste buds, which are innervated by gustatory neurons whose afferents do not traverse sites of ectopic BDNF expression, appeared unaffected. We suggest that lingual gustatory deficits in BDNF overexpressing mice are a consequence of the failure of their BDNF-dependent afferents to reach their targets because of the effects of ectopically expressed BDNF on fiber growth. Our findings suggest that mammalian taste buds and gustatory papillae require proper BDNF-dependent gustatory innervation for development and that the correct spatial expression of BDNF in the tongue epithelium is crucial for appropriate target invasion and innervation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Proteínas de Filamentos Intermediários/fisiologia , Proteínas do Tecido Nervoso , Papilas Gustativas/fisiologia , Língua/fisiologia , Animais , Encéfalo/fisiologia , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/genética , Desenvolvimento Embrionário e Fetal , Regulação da Expressão Gênica , Gânglio Geniculado/fisiologia , Proteínas de Filamentos Intermediários/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Fibras Nervosas/fisiologia , Nestina , Neurônios/fisiologia , Especificidade de Órgãos , Valores de Referência , Sequências Reguladoras de Ácido Nucleico , Papilas Gustativas/anormalidades , Papilas Gustativas/patologia , Língua/embriologia , Língua/inervação , Língua/ultraestrutura
18.
Trends Neurosci ; 21(10): 438-44, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9786342

RESUMO

Neurotrophic factors control the survival, differentiation and maintenance of neurons in the peripheral and central nervous systems. Their discovery and characterization have been instrumental to our understanding of a wide range of phenomena in the development, plasticity and repair of the nervous system. Their potential importance in the development of therapeutic agents against neurodegenerative disorders and nerve injury has led to a flurry of activity towards understanding their structure, function and signaling mechanisms. This knowledge has increased dramatically in recent years, in particular due to the elucidation of three-dimensional structures, the discovery of families of structurally related neurotrophic factors and the characterization of receptors and downstream signaling components. Common themes are emerging from these recent studies that allow us to make new insights and predictions as to the function and possible clinical utility of these molecules.


Assuntos
Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/fisiologia , Animais , Sítios de Ligação , Transporte Biológico/fisiologia , Humanos , Estrutura Molecular , Fatores de Crescimento Neural/uso terapêutico , Tecido Nervoso/fisiologia , Doenças Neurodegenerativas/tratamento farmacológico , Relação Estrutura-Atividade , Transmissão Sináptica/fisiologia
19.
Neuron ; 21(2): 305-15, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9728912

RESUMO

Cajal-Retzius (CR) cells of the cerebral cortex express receptors for the neurotrophin brain-derived neurotrophic factor (BDNF) and downregulate expression of the extracellular matrix protein Reelin during early postnatal development, coincident with the onset of cortical BDNF expression. During this period, mice lacking BDNF have elevated levels of Reelin in CR cells. Acute BDNF stimulation of cortical neuron cultures and overexpression of BDNF in the developing brain of transgenic mice prior to the onset of endogenous production causes a profound, dose-dependent reduction of Reelin expression in CR cells. In addition, overexpression of BDNF produces gaps and heterotopias in the marginal zone and disorganization and aggregation of cortical CR cells and induces several other malformations, including aberrant cortical lamination, similar to the phenotype of reeler mutant mice, which lack Reelin. These results demonstrate a role for BDNF on cortical CR cells and identify Reelin as a direct effector of this neurotrophin during brain development.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Córtex Cerebral/metabolismo , Proteínas da Matriz Extracelular/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Neurônios/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Regulação para Baixo , Proteínas de Filamentos Intermediários/metabolismo , Camundongos , Camundongos Transgênicos , Nestina , Ratos , Proteína Reelina
20.
Proc Natl Acad Sci U S A ; 95(9): 5269-74, 1998 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-9560265

RESUMO

Peripheral nerve injury markedly regulates expression of neurotrophins and their receptors in the lesioned nerve. However, the role of endogenously produced neurotrophins in the process of nerve regeneration is unclear. Expression of a multifunctional neurotrophin, pan-neurotrophin-1 (PNT-1), was targeted to the peripheral nerves of transgenic mice by using a gene promoter that is specifically activated after nerve lesion but that is otherwise silent in all other tissues and during development. PNT-1 is a chimeric neurotrophin that combines the active sites of the neurotrophins nerve growth factor, brain-derived neurotrophic factor, and neurotrophin-3 and binds and activates all known neurotrophin receptors. In adult transgenic mice, PNT-1 was highly expressed in transected but not in intact sciatic nerve. Morphometric analyses at the electron microscopy level showed increased and accelerated recovery of axon diameter of myelinated fibers in crushed peripheral nerves of transgenic mice compared with wild type. Examination of nerve bundles in target tissues indicated accelerated reinnervation of foot pad dermis and flexor plantaris muscle in transgenic mice. Moreover, transected sensory and motor axons of transgenic mice showed faster and increased return of neurophysiological responses, suggesting an accelerated rate of axonal elongation. Importantly, transgenic mice also showed a markedly ameliorated loss of skeletal muscle weight, indicating functional regeneration of motor axons. Together, these data provide evidence, at both the anatomical and functional levels, that neurotrophins endogenously produced by the lesioned nerve are capable of significantly accelerating the regeneration of both sensory and motor axons after peripheral nerve damage. In addition, our results indicate that exogenous PNT-1 administration may be an effective therapeutic treatment of peripheral nerve injuries.


Assuntos
Neurônios Motores/citologia , Fatores de Crescimento Neural/administração & dosagem , Regeneração Nervosa , Neurônios Aferentes/citologia , Animais , Axônios/fisiologia , Camundongos , Camundongos Transgênicos , Músculo Esquelético/anatomia & histologia , Músculos/inervação , Compressão Nervosa , Fatores de Crescimento Neural/genética , Tamanho do Órgão , Proteínas Recombinantes de Fusão , Nervo Isquiático , Pele/inervação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA