Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Psychiatry ; 26(2): 508-523, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-30610203

RESUMO

Bipolar disorder (BD) is a common mood disorder characterized by recurrent episodes of mania and depression. Both genetic and environmental factors have been implicated in BD etiology, but the biological underpinnings remain elusive. Recently, genome-wide association studies (GWAS) of neuropsychiatric disorders have identified a risk locus for BD containing the SYNE1 gene, a large gene encoding multiple proteins. The BD association signal spans, almost exclusively, the part of SYNE1 encoding CPG2, a brain-specific protein localized to excitatory postsynaptic sites, where it regulates glutamate receptor internalization. Here we show that CPG2 protein levels are significantly decreased in postmortem brain tissue from BD patients, as compared to control subjects, as well as schizophrenia and depression patients. We identify genetic variants within the postmortem brains that map to the CPG2 promoter region, and show that they negatively affect gene expression. We also identify missense single nucleotide polymorphisms (SNPs) in CPG2 coding regions that affect CPG2 expression, localization, and synaptic function. Our findings link genetic variation in the CPG2 region of SYNE1 with a mechanism for glutamatergic synapse dysfunction that could underlie susceptibility to BD in some individuals. Few GWAS hits in human genetics for neuropsychiatric disorders to date have afforded such mechanistic clues. Further, the potential for genetic distinction of susceptibility to BD from other neuropsychiatric disorders with overlapping clinical traits holds promise for improved diagnostics and treatment of this devastating illness.


Assuntos
Transtorno Bipolar , Proteínas do Citoesqueleto/genética , Proteínas do Tecido Nervoso/genética , Esquizofrenia , Transtorno Bipolar/genética , Encéfalo/metabolismo , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Polimorfismo de Nucleotídeo Único
2.
Mol Cell Neurosci ; 71: 46-55, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26704904

RESUMO

Bipolar disorder (BD) is a prevalent and severe mood disorder characterized by recurrent episodes of mania and depression. Both genetic and environmental factors have been implicated in BD etiology, but the biological underpinnings remain elusive. Recent genome-wide association studies (GWAS) for identifying genes conferring risk for schizophrenia, BD, and major depression, identified an association between single-nucleotide polymorphisms (SNPs) in the SYNE1 gene and increased risk of BD. SYNE1 has also been identified as a risk locus for multiple other neurological or neuromuscular genetic disorders. The BD associated SNPs map within the gene region homologous to part of rat Syne1 encompassing the brain specific transcripts encoding CPG2, a postsynaptic neuronal protein localized to excitatory synapses and an important regulator of glutamate receptor internalization. Here, we use RNA-seq, ChIP-seq and RACE to map the human SYNE1 transcriptome, focusing on the CPG2 locus. We validate several CPG2 transcripts, including ones not previously annotated in public databases, and identify and clone a full-length CPG2 cDNA expressed in human neocortex, hippocampus and striatum. Using lenti-viral gene knock down/replacement and surface receptor internalization assays, we demonstrate that human CPG2 protein localizes to dendritic spines in rat hippocampal neurons and is functionally equivalent to rat CPG2 in regulating glutamate receptor internalization. This study provides a valuable gene-mapping framework for relating multiple genetic disease loci in SYNE1 with their transcripts, and for evaluating the effects of missense SNPs identified by patient genome sequencing on neuronal function.


Assuntos
Genoma Humano , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Mapeamento Cromossômico/métodos , Proteínas do Citoesqueleto , Espinhas Dendríticas/metabolismo , Endocitose , Células HEK293 , Humanos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Polimorfismo de Nucleotídeo Único , Ratos , Receptores de Glutamato/metabolismo , Sinapses/metabolismo , Transcriptoma
3.
Proc Natl Acad Sci U S A ; 110(35): 14426-31, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23940334

RESUMO

NMDA receptor activation promotes endocytosis of AMPA receptors, which is an important mechanism underlying long-term synaptic depression. The pH-sensitive GFP variant pHluorin fused to the N terminus of GluA2 (pH-GluA2) has been used to assay NMDA-mediated AMPA receptor endocytosis and recycling. Here, we demonstrate that in somatic and dendritic regions of hippocampal neurons a large fraction of the fluorescent signal originates from intracellular pH-GluA2, and that the decline in fluorescence in response to NMDA and AMPA primarily describes an intracellular acidification, which quenches the pHluorin signal from intracellular receptor pools. Neurons expressing an endoplasmic reticulum-retained mutant of GluA2 (pH-GluA2 ΔC49) displayed a larger response to NMDA than neurons expressing wild-type pH-GluA2. A similar NMDA-elicited decline in pHluorin signal was observed by expressing cytosolic pHluorin alone without fusion to GluA2 (cyto-pHluorin). Intracellular acidification in response to NMDA was further confirmed by using the ratiometric pH indicator carboxy-SNARF-1. The NMDA-induced decline was followed by rapid recovery of the fluorescent signal from both cyto-pHluorin and pH-GluA2. The recovery was sodium-dependent and sensitive to Na(+)/H(+)-exchanger (NHE) inhibitors. Moreover, recovery was more rapid after shRNA-mediated knockdown of the GluA2 binding PDZ domain-containing protein interacting with C kinase 1 (PICK1). Interestingly, the accelerating effect of PICK1 knockdown on the fluorescence recovery was eliminated in the presence of the NHE1 inhibitor zoniporide. Our results indicate that the pH-GluA2 recycling assay is an unreliable assay for studying AMPA receptor trafficking and also suggest a role for PICK1 in regulating intracellular pH via modulation of NHE activity.


Assuntos
Acidose/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Células Cultivadas , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Trombina/metabolismo
4.
J Biol Chem ; 289(36): 25327-40, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25023278

RESUMO

PDZ domain proteins control multiple cellular functions by governing assembly of protein complexes. It remains unknown why individual PDZ domains can bind the extreme C terminus of very diverse binding partners and maintain selectivity. By employing NMR spectroscopy, together with molecular modeling, mutational analysis, and fluorescent polarization binding experiments, we identify here three structural mechanisms explaining why the PDZ domain of PICK1 selectively binds >30 receptors, transporters, and kinases. Class II ligands, including the dopamine transporter, adopt a canonical binding mode with promiscuity obtained via differential packing in the binding groove. Class I ligands, such as protein kinase Cα, depend on residues upstream from the canonical binding sequence that are likely to interact with flexible loop residues of the PDZ domain. Finally, we obtain evidence that the unconventional ligand ASIC1a has a dual binding mode involving a canonical insertion and a noncanonical internal insertion with the two C-terminal residues forming interactions outside the groove. Together with an evolutionary analysis, the data show how unconventional binding modes might evolve for a protein recognition domain to expand the repertoire of functionally important interactions.


Assuntos
Proteínas de Transporte/química , Simulação de Acoplamento Molecular/métodos , Proteínas Nucleares/química , Domínios PDZ , Sequência de Aminoácidos , Sítios de Ligação/genética , Ligação Competitiva , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Polarização de Fluorescência , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Proteína Quinase C-alfa/química , Proteína Quinase C-alfa/genética , Proteína Quinase C-alfa/metabolismo
5.
J Biol Chem ; 288(38): 27534-27544, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-23884410

RESUMO

The dopamine transporter (DAT) is responsible for sequestration of extracellular dopamine (DA). The psychostimulant amphetamine (AMPH) is a DAT substrate, which is actively transported into the nerve terminal, eliciting vesicular depletion and reversal of DA transport via DAT. Here, we investigate the role of the DAT C terminus in AMPH-evoked DA efflux using cell-permeant dominant-negative peptides. A peptide, which corresponded to the last 24 C-terminal residues of DAT (TAT-C24 DAT) and thereby contained the Ca(2+)-calmodulin-dependent protein kinase IIα (CaMKIIα) binding domain and the PSD-95/Discs-large/ZO-1 (PDZ)-binding sequence of DAT, was made membrane-permeable by fusing it to the cell membrane transduction domain of the HIV-1 Tat protein (TAT-C24WT). The ability of TAT-C24WT but not a scrambled peptide (TAT-C24Scr) to block the CaMKIIα-DAT interaction was supported by co-immunoprecipitation experiments in heterologous cells. In heterologous cells, we also found that TAT-C24WT, but not TAT-C24Scr, decreased AMPH-evoked 1-methyl-4-phenylpyridinium efflux. Moreover, chronoamperometric recordings in striatum revealed diminished AMPH-evoked DA efflux in mice preinjected with TAT-C24WT. Both in heterologous cells and in striatum, the peptide did not further inhibit efflux upon KN-93-mediated inhibition of CaMKIIα activity, consistent with a dominant-negative action preventing binding of CaMKIIα to the DAT C terminus. This was further supported by the ability of a peptide with perturbed PDZ-binding sequence, but preserved CaMKIIα binding (TAT-C24AAA), to diminish AMPH-evoked DA efflux in vivo to the same extent as TAT-C24WT. Finally, AMPH-induced locomotor hyperactivity was attenuated following systemic administration of TAT-C24WT but not TAT-C24Scr. Summarized, our findings substantiate that DAT C-terminal protein-protein interactions are critical for AMPH-evoked DA efflux and suggest that it may be possible to target protein-protein interactions to modulate transporter function and interfere with psychostimulant effects.


Assuntos
Anfetamina/farmacologia , Peptídeos Penetradores de Células/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Corpo Estriado/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/farmacologia , Dopamina/metabolismo , Anfetamina/efeitos adversos , Animais , Benzilaminas/farmacologia , Peptídeos Penetradores de Células/metabolismo , Estimulantes do Sistema Nervoso Central/efeitos adversos , Proteínas da Membrana Plasmática de Transporte de Dopamina/farmacocinética , Humanos , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Domínios PDZ , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Sulfonamidas/farmacologia
6.
Proc Natl Acad Sci U S A ; 107(1): 413-8, 2010 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-20018661

RESUMO

Proteins containing PSD-95/Discs-large/ZO-1 homology (PDZ) domains play key roles in the assembly and regulation of cellular signaling pathways and represent putative targets for new pharmacotherapeutics. Here we describe the first small-molecule inhibitor (FSC231) of the PDZ domain in protein interacting with C kinase 1 (PICK1) identified by a screening of approximately 44,000 compounds in a fluorescent polarization assay. The inhibitor bound the PICK1 PDZ domain with an affinity similar to that observed for endogenous peptide ligands (K(i) approximately 10.1 microM). Mutational analysis, together with computational docking of the compound in simulations starting from the PDZ domain structure, identified the binding mode of FSC231. The specificity of FSC231 for the PICK1 PDZ domain was supported by the lack of binding to PDZ domains of postsynaptic density protein 95 (PSD-95) and glutamate receptor interacting protein 1 (GRIP1). Pretreatment of cultured hippocampal neurons with FSC231 inhibited coimmunopreciptation of the AMPA receptor GluR2 subunit with PICK1. In agreement with inhibiting the role of PICK1 in GluR2 trafficking, FSC231 accelerated recycling of pHluorin-tagged GluR2 in hippocampal neurons after internalization in response to NMDA receptor activation. FSC231 blocked the expression of both long-term depression and long-term potentiation in hippocampal CA1 neurons from acute slices, consistent with inhibition of the bidirectional function of PICK1 in synaptic plasticity. Given the proposed role of the PICK1/AMPA receptor interaction in neuropathic pain, excitotoxicity, and cocaine addiction, FSC231 might serve as a lead in the future development of new therapeutics against these conditions.


Assuntos
Carbamatos/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Cinamatos/metabolismo , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Depressão Sináptica de Longo Prazo/fisiologia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Domínios PDZ , Animais , Sítios de Ligação , Células COS , Carbamatos/química , Proteínas de Transporte/química , Proteínas de Transporte/genética , Chlorocebus aethiops , Cinamatos/química , Proteínas do Citoesqueleto , Hipocampo/citologia , Humanos , Modelos Moleculares , Estrutura Molecular , Plasticidade Neuronal/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Proteínas Nucleares/química , Proteínas Nucleares/genética , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Estrutura Terciária de Proteína , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
7.
J Neurochem ; 121(6): 915-23, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22469063

RESUMO

Apart from its hematopoietic activity, erythropoietin (EPO) is also known as a tissue-protective cytokine. In the brain, EPO and its receptor are up-regulated in response to insult and exert pro-survival effects. EPO binds to its receptor (EPOR) via high- and low-affinity binding sites (Sites 1 and 2, respectively), inducing conformational changes in the receptor, followed by the activation of downstream signaling cascades. Based on the crystal structure of the EPO:EPOR(2) complex, we designed a peptide, termed Epobis, whose sequence encompassed amino acids from binding Site 1. The present study shows that the Epobis peptide specifically binds to EPOR and induces neurite outgrowth from primary neurons in an EPOR-expression dependent manner. Furthermore, Epobis promoted the survival of hippocampal and cerebellar neuronal cultures after kainate treatment and KCl deprivation, respectively. Thus, we identified a new functional agonist of EPOR with the potential to promote neuroregeneration and neuroprotection.


Assuntos
Neuritos/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Peptídeos/farmacologia , Receptores da Eritropoetina/agonistas , Receptores da Eritropoetina/metabolismo , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Eritropoetina/química , Eritropoetina/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Modelos Moleculares , Fármacos Neuroprotetores/metabolismo , Peptídeos/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Ressonância de Plasmônio de Superfície , Transfecção
9.
Brain ; 133(Pt 8): 2281-94, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20435631

RESUMO

Erythropoietin, a member of the type 1 cytokine superfamily, controls proliferation and differentiation of erythroid progenitor cells through binding to and dimerization of the erythropoietin receptor. Both erythropoietin and its receptor are also expressed in the central nervous system, where they are involved in tissue protection. However, the use of erythropoietin as a neuroprotective agent may be hampered by its erythropoietic activity. Therefore, developing non-haematopoietic erythropoietin mimetics is important. Based on the crystal structure of the complex of erythropoietin and its receptor, we designed a peptide, termed Epotris, corresponding to the C α-helix region (amino-acid residues 92-111) of human erythropoietin. The peptide specifically bound to the erythropoietin receptor and promoted neurite outgrowth and survival of primary neurons with the same efficiency as erythropoietin, but with 10(3)-fold lower potency. Knockdown of the erythropoietin receptor or interference with its downstream signalling inhibited the Epotris-induced neuritogenic and pro-survival effect. Similarly to erythropoietin, Epotris penetrated the blood-brain barrier. Moreover, treatment with the peptide attenuated seizures, decreased mortality and reduced neurodegeneration in an in vivo model of kainic acid-induced neurotoxicity. In contrast to erythropoietin, Epotris did not stimulate erythropoiesis upon chronic administration. Thus, Epotris is a novel neuroprotective non-haematopoietic erythropoietin mimetic that may offer new opportunities for the treatment of neurological disorders.


Assuntos
Eritropoetina/farmacologia , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptores da Eritropoetina/agonistas , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Células Cultivadas , Eritropoese/efeitos dos fármacos , Eritropoese/fisiologia , Eritropoetina/química , Eritropoetina/metabolismo , Eritropoetina/farmacocinética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/mortalidade , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacocinética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacocinética , Ratos , Ratos Wistar , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Convulsões/tratamento farmacológico , Convulsões/mortalidade
10.
EMBO Mol Med ; 12(6): e11248, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32352640

RESUMO

Maladaptive plasticity involving increased expression of AMPA-type glutamate receptors is involved in several pathologies, including neuropathic pain, but direct inhibition of AMPARs is associated with side effects. As an alternative, we developed a cell-permeable, high-affinity (~2 nM) peptide inhibitor, Tat-P4 -(C5)2 , of the PDZ domain protein PICK1 to interfere with increased AMPAR expression. The affinity is obtained partly from the Tat peptide and partly from the bivalency of the PDZ motif, engaging PDZ domains from two separate PICK1 dimers to form a tetrameric complex. Bivalent Tat-P4 -(C5)2 disrupts PICK1 interaction with membrane proteins on supported cell membrane sheets and reduce the interaction of AMPARs with PICK1 and AMPA-receptor surface expression in vivo. Moreover, Tat-P4 -(C5)2 administration reduces spinal cord transmission and alleviates mechanical hyperalgesia in the spared nerve injury model of neuropathic pain. Taken together, our data reveal Tat-P4 -(C5)2 as a novel promising lead for neuropathic pain treatment and expand the therapeutic potential of bivalent inhibitors to non-tandem protein-protein interaction domains.


Assuntos
Neuralgia , Domínios PDZ , Proteínas de Transporte/metabolismo , Humanos , Neuralgia/tratamento farmacológico , Proteínas Nucleares/metabolismo , Receptores de AMPA/metabolismo
11.
ACS Chem Biol ; 8(9): 2033-41, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-23838571

RESUMO

The natural product argiotoxin-636 (ArgTX-636) found in the venom of the Argiope lobata spider is a potent open-channel blocker of ionotropic glutamate (iGlu) receptors, and recently, two analogues, ArgTX-75 and ArgTX-48, were identified with increased potency and selectivity for iGlu receptor subtypes. Here, we have exploited these analogues as templates in the development of fluorescent iGlu receptor ligands to be employed as unique tools for dynamic studies. Eighteen fluorescent analogues were designed and synthesized, and subsequently pharmacologically evaluated at three iGlu receptor subtypes, which resulted in the discovery of highly potent fluorescent iGlu receptor antagonists with IC50 values as low as 11 nM. The most promising ligands were further characterized showing retention of their mechanism of action, as open-channel blockers of iGlu receptors, as well as preservation of the photophysical properties of the incorporated fluorophores. Finally, we demonstrate the applicability of the developed probes for imaging of iGlu receptors in hippocampal neurons.


Assuntos
Corantes Fluorescentes/química , Hipocampo/citologia , Ácidos Indolacéticos/química , Neurônios/citologia , Poliaminas/química , Receptores Ionotrópicos de Glutamato/análise , Animais , Corantes Fluorescentes/metabolismo , Corantes Fluorescentes/farmacologia , Ácidos Indolacéticos/metabolismo , Ácidos Indolacéticos/farmacologia , Ligantes , Modelos Moleculares , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Poliaminas/metabolismo , Poliaminas/farmacologia , Ratos , Receptores Ionotrópicos de Glutamato/antagonistas & inibidores , Receptores Ionotrópicos de Glutamato/metabolismo , Coloração e Rotulagem
12.
Eur J Cell Biol ; 90(12): 990-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22000729

RESUMO

Ciliary neurotrophic factor (CNTF) induces neuronal differentiation and promotes the survival of various neuronal cell types by binding to a receptor complex formed by CNTF receptor α (CNTFRα), gp130, and the leukemia inhibitory factor (LIF) receptor (LIFR). The CD loop-D helix region of CNTF has been suggested to be important for the cytokine interaction with LIFR. We designed a peptide, termed cintrofin, that encompasses this region. Surface plasmon resonance analysis demonstrated that cintrofin bound to LIFR and gp130, but not to CNTFRα, with apparent KD values of 35 nM and 1.1 nM, respectively. Cintrofin promoted the survival of cerebellar granule neurons (CGNs), in which cell death was induced either by potassium withdrawal or H2O2 treatment. Cintrofin induced neurite outgrowth from CGNs, and this effect was inhibited by specific antibodies against both gp130 and LIFR, indicating that these receptors are involved in the effects of cintrofin. The C-terminal part of the peptide, corresponding to the D helix region of CNTF, was shown to be essential for the neuritogenic action of the peptide. CNTF and LIF induced neurite outgrowth in CGNs plated on laminin-coated slides. On uncoated slides, CNTF and LIF had no neuritogenic effect but were able to inhibit cintrofin-induced neuronal differentiation, indicating that cintrofin and cytokines compete for the same receptors. In addition, cintrofin induced the phosphorylation of STAT3, Akt, and ERK, indicating that it exerts cell signaling properties similar to those induced by CNTF and may be a valuable survival agent with possible therapeutic potential.


Assuntos
Fator Neurotrófico Ciliar/farmacologia , Fator Inibidor de Leucemia/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Citocinas/metabolismo , Sequência de Aminoácidos , Antígenos CD/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fator Neurotrófico Ciliar/metabolismo , Humanos , Interleucina-6/metabolismo , Microscopia Confocal , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA