Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
J Neurochem ; 160(2): 234-255, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34816431

RESUMO

The nervous system monitors the environment to maintain homeostasis, which can be affected by stressful conditions. Using mammalian models of chronic stress, we previously observed altered brain levels of GPM6A, a protein involved in neuronal morphology. However, GPM6A's role in systemic stress responses remains unresolved. The nematode Caenorhabditis elegans expresses a GPM6A ortholog, the neuronal membrane glycoprotein 1 (NMGP-1). Because of the shared features between nematode and mammalian nervous systems and the vast genetic tools available in C. elegans, we used the worm to elucidate the role of GPM6A in the stress response. We first identified nmgp-1 expression in different amphid and phasmid neurons. To understand the nmgp-1 role, we characterized the behavior of nmgp-1(RNAi) animals and two nmgp-1 mutant alleles. Compared to control animals, mutant and RNAi-treated worms exhibited increased recovery time from the stress-resistant dauer stage, altered SDS chemosensation and reduced egg-laying rate resulting in egg retention (bag-of-worms phenotype). Silencing of nmgp-1 expression induced morphological abnormalities in the ASJ sensory neurons, partly responsible for dauer exit. These results indicate that nmgp-1 is required for neuronal morphology and for behaviors associated with chemosensation. Finally, we propose nmgp-1 mutants as a tool to screen drugs for human nervous system pathologies.


Assuntos
Adaptação Fisiológica/fisiologia , Comportamento Animal/fisiologia , Caenorhabditis elegans/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Feminino
2.
Mol Cell Proteomics ; 14(7): 1871-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25922409

RESUMO

Complete characterization of antibody specificities associated to natural infections is expected to provide a rich source of serologic biomarkers with potential applications in molecular diagnosis, follow-up of chemotherapeutic treatments, and prioritization of targets for vaccine development. Here, we developed a highly-multiplexed platform based on next-generation high-density peptide microarrays to map these specificities in Chagas Disease, an exemplar of a human infectious disease caused by the protozoan Trypanosoma cruzi. We designed a high-density peptide microarray containing more than 175,000 overlapping 15 mer peptides derived from T. cruzi proteins. Peptides were synthesized in situ on microarray slides, spanning the complete length of 457 parasite proteins with fully overlapped 15 mers (1 residue shift). Screening of these slides with antibodies purified from infected patients and healthy donors demonstrated both a high technical reproducibility as well as epitope mapping consistency when compared with earlier low-throughput technologies. Using a conservative signal threshold to classify positive (reactive) peptides we identified 2,031 disease-specific peptides and 97 novel parasite antigens, effectively doubling the number of known antigens and providing a 10-fold increase in the number of fine mapped antigenic determinants for this disease. Finally, further analysis of the chip data showed that optimizing the amount of sequence overlap of displayed peptides can increase the protein space covered in a single chip by at least ∼ threefold without sacrificing sensitivity. In conclusion, we show the power of high-density peptide chips for the discovery of pathogen-specific linear B-cell epitopes from clinical samples, thus setting the stage for high-throughput biomarker discovery screenings and proteome-wide studies of immune responses against pathogens.


Assuntos
Doença de Chagas/imunologia , Mapeamento de Epitopos/métodos , Epitopos de Linfócito B/imunologia , Ensaios de Triagem em Larga Escala/métodos , Peptídeos/metabolismo , Análise Serial de Proteínas/métodos , Proteômica/métodos , Antígenos de Protozoários/imunologia , Linfócitos B/imunologia , Bases de Dados de Proteínas , Ensaio de Imunoadsorção Enzimática , Humanos , Reprodutibilidade dos Testes
3.
Mol Cell Neurosci ; 77: 95-104, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27793698

RESUMO

Neuronal glycoprotein M6a belongs to the tetraspan proteolipid protein (PLP) family. Mutations in GPM6A gene have been related to mental disorders like schizophrenia, bipolar disorders and claustrophobia. M6a is expressed mainly in neuronal cells of the central nervous system and it has been extensively related to neuronal plasticity. M6a induces neuritogenesis and axon/filopodium outgrowth; however its mechanism of action is still unresolved. We recently reported that the integrity of the transmembrane domains (TMDs) 2 and 4 are critical for M6a filopodia induction. There is also experimental data suggesting that M6a might be involved in synaptogenesis. In this regard, we have previously determined that M6a is involved in filopodia motility, a process that is described in the first step of the filopodial model for synaptogenesis. In this work we analyzed the possible involvement of M6a in synaptogenesis and spinogenesis, and evaluated the effect of two non-synonymous SNPs present in the coding region of TMD2-GPM6A in these processes. The results showed that endogenous M6a colocalized with both, pre-synaptic (synaptophysin) and post-synaptic (NMDA-R1), markers along of neuronal soma and dendrites. M6a-overexpressing neurons displayed an increased number of synaptophysin and NMDA-R1 puncta and, also, an increased number of colocalization puncta between both markers. Conversely, the number of synaptic puncta markers in neurons expressing nsSNP variants was similar to those of control neurons. Overexpression of M6a is accompanied by an increase in spine density, particularly in mature spines, as compared with neurons expressing mGFP or GPM6A nsSNP variants. Taken together, these results suggest that M6a contributes positively to spine and, likely, synapse formation.


Assuntos
Espinhas Dendríticas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Sinapses/metabolismo , Animais , Feminino , Hipocampo/citologia , Hipocampo/embriologia , Hipocampo/metabolismo , Glicoproteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Sinaptofisina/metabolismo
4.
J Neurochem ; 137(1): 46-61, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26809475

RESUMO

Stress-responsive neuronal membrane glycoprotein M6a (Gpm6a) functions in neurite extension, filopodium and spine formation and synaptogenesis. The mechanisms of Gpm6a action in these processes are incompletely understood. Previously, we identified the actin regulator coronin-1a (Coro1a) as a putative Gpm6a interacting partner. Here, we used co-immunoprecipitation assays with the anti-Coro1a antibody to show that Coro1a associates with Gpm6a in rat hippocampal neurons. By immunofluorescence microscopy, we demonstrated that in hippocampal neurons Coro1a localizes in F-actin-enriched regions and some of Coro1a spots co-localize with Gpm6a labeling. Notably, the over-expression of a dominant-negative form of Coro1a as well as its down-regulation by siRNA interfered with Gpm6a-induced filopodium formation. Coro1a is known to regulate the plasma membrane translocation and activation of small GTPase Rac1. We show that Coro1a co-immunoprecipitates with Rac1 together with Gpm6a. Pharmacological inhibition of Rac1 resulted in a significant decrease in filopodium formation by Gpm6a. The same was observed upon the co-expression of Gpm6a with the inactive GDP-bound form of Rac1. In this case, the elevated membrane recruitment of GDP-bound Rac1 was detected as well. Moreover, the kinase activity of the p21-activated kinase 1 (Pak1), a main downstream effector of Rac1 that acts downstream of Coro1a, was required for Gpm6a-induced filopodium formation. Taken together, our results provide evidence that a signaling pathway including Coro1a, Rac1, and Pak1 facilitates Gpm6a-induced filopodium formation. Formation of filopodia by membrane glycoprotein M6a (Gpm6a) requires actin regulator coronin-1a (Coro1a), known to regulate plasma membrane localization and activation of Rac1 and its downstream effector Pak1. Coro1a associates with Gpm6a. Blockage of Coro1a, Rac1, or Pak1 interferes with Gpm6a-induced filopodium formation. Moreover, Gpm6a facilitates Rac1 membrane recruitment. Altogether, a mechanistic insight into the process of Gpm6a-induced neuronal filopodium formation is provided.


Assuntos
Glicoproteínas de Membrana/fisiologia , Proteínas dos Microfilamentos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/ultraestrutura , Pseudópodes/fisiologia , Quinases Ativadas por p21/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Actinas/análise , Animais , Células Cultivadas , Regulação para Baixo , Genes Reporter , Hipocampo/citologia , Proteínas dos Microfilamentos/genética , Proteínas do Tecido Nervoso/antagonistas & inibidores , Biogênese de Organelas , Cultura Primária de Células , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores
5.
J Neurochem ; 134(3): 499-512, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25940868

RESUMO

Membrane glycoprotein M6a, which belongs to the tetraspan proteolipid protein family, promotes structural plasticity in neurons and cell lines by unknown mechanisms. This glycoprotein is encoded by Gpm6a, a stress-regulated gene. The hippocampus of animals chronically stressed by either psychosocial or physical stressors shows decreased M6a expression. Stressed Gpm6a-null mice develop a claustrophobia-like phenotype. In humans, de novo duplication of GPM6A results in learning/behavioral abnormalities, and two single-nucleotide polymorphisms (SNPs) in the non-coding region are linked to mood disorders. Here, we studied M6a dimerization in neuronal membranes and its functional relevance. We showed that the self-interaction of M6a transmembrane domains (TMDs) might be driving M6a dimerization, which is required to induce filopodia formation. Glycine mutants located in TMD2 and TMD4 of M6a affected its dimerization, thus preventing M6a-induced filopodia formation in neurons. In silico analysis of three non-synonymous SNPs located in the coding region of TMDs suggested that these mutations induce protein instability. Indeed, these SNPs prevented M6a from being functional in neurons, owing to decreased stability, dimerization or improper folding. Interestingly, SNP3 (W141R), which caused endoplasmic reticulum retention, is equivalent to that mutated in PLP1, W161L, which causes demyelinating Pelizaeus-Merzbacher disease. In this work we analyzed the functional contribution of transmembrane domains (TMDs) of the neuronal membrane glycoprotein M6a. We determined that certain glycines present in TMD2 and TMD4 are critical for filopodia induction in neurons. In addition, three nsSNPs located in the coding region of TMD2 and TMD3 of GPM6A impair M6a function by affecting its stability, folding and dimer formation.


Assuntos
Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/ultraestrutura , Sequência de Aminoácidos , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Immunoblotting , Imuno-Histoquímica , Glicoproteínas de Membrana/química , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Polimorfismo de Nucleotídeo Único , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley
6.
J Neurosci Res ; 93(2): 215-29, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25242528

RESUMO

Neuronal glycoprotein M6a is involved in neuronal plasticity, promoting neurite and filopodia outgrowth and, likely, synaptogenesis. Polymorphisms in the human M6a gene GPM6A have recently been associated with mental illnesses such as schizophrenia, bipolar disorders, and claustrophobia. Nevertheless, the molecular bases underlying these observations remain unknown. We have previously documented that, to induce filopodia formation, M6a depends on the association of membrane lipid microdomains and the activation of Src and mitogen-activated protein kinase kinases. Here, in silico analysis of the phosphorylation of tyrosine 251 (Y251) at the C-terminus of M6a showed that it could be a target of Src kinases. We examined whether phosphorylation of M6a at Y251 affects neurite and filopodia outgrowth and the targets involved in its signal propagation. This work provides evidence that the Src kinase family and the phosphatidylinositide 3-kinase (PI3K), but not Ras, participate in M6a signal cascade leading to neurite/filopodia outgrowth in hippocampal neurons and murine neuroblastoma N2a cells. Phosphorylation of M6a at Y251 is essential only for neurite outgrowth by the PI3K/AKT-mediated pathway and, moreover, rescues the inhibition caused by selective Src inhibitor and external M6a monoclonal antibody treatment. Thus, we suggest that phosphorylation of M6a at Y251 is critical for a specific stage of neuronal development and triggers redundant signaling pathways leading to neurite extension.


Assuntos
Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Tirosina/metabolismo , Animais , Linhagem Celular Transformada , Cromonas/farmacologia , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Técnicas In Vitro , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Camundongos , Morfolinas/farmacologia , Mutação/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Pseudópodes/fisiologia , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
7.
J Biol Chem ; 287(23): 19058-69, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22500021

RESUMO

Trypanosoma cruzi, the agent of Chagas disease, does not seem to control gene expression through regulation of transcription initiation and makes use of post-transcriptional mechanisms. We report here a 43-nt U-rich RNA element located in the 3'-untranslated region (3'-UTR) of a large number of T. cruzi mRNAs that is important for mRNA abundance in the intracellular amastigote stage of the parasite. Whole genome scan analysis, differential display RT-PCR, Northern blot, and RT-PCR analyses were used to determine the transcript levels of more than 900 U-rich-containing mRNAs of large gene families as well as single and low copy number genes. Our results indicate that the 43-nt U-rich mRNA element is preferentially present in amastigotes. The cis-element of a protein kinase 3'-UTR but not its mutated version promoted the expression of the green fluorescent protein reporter gene in amastigotes. The regulatory cis-element, but not its mutated version, was also shown to interact with the trypanosome-specific RNA-binding protein (RBP) TcUBP1 but not with other related RBPs. Co-immunoprecipitation experiments of TcUBP1-containing ribonucleoprotein complexes formed in vivo validated the interaction with representative endogenous RNAs having the element. These results suggest that this 43-nt U-rich element together with other yet unidentified sequences might be involved in the modulation of abundance and/or translation of subsets of transcripts in the amastigote stage.


Assuntos
Regiões 3' não Traduzidas/fisiologia , Genoma de Protozoário/fisiologia , RNA de Protozoário/metabolismo , Proteínas de Ligação a RNA/metabolismo , Trypanosoma cruzi/fisiologia , RNA de Protozoário/genética , Proteínas de Ligação a RNA/genética
8.
J Biol Chem ; 286(51): 43959-43971, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22039054

RESUMO

The protist parasite Trypanosoma cruzi has evolved the ability to transit between completely different hosts and to replicate in adverse environments. In particular, the epimastigote form, the replicative stage inside the vector, is subjected to nutritional and osmotic stresses during its development. In this work, we describe the biochemical and global gene expression changes of epimastigotes under hyperosmotic conditions. Hyperosmotic stress resulted in cell shrinking within a few minutes. Depending on the medium osmolarity, this was followed by lack of volume recovery for at least 2 h or by slow recovery. Experiments with inhibitors, or with cells in which an aquaporin gene (TcAQP1) was knocked down or overexpressed, revealed its importance for the cellular response to hyperosmotic stress. Furthermore, the adaptation to this new environment was shown to involve the regulation of the polyphosphate polymerization state as well as changes in amino acid catabolism to generate compatible osmolytes. A genome-wide transcriptional analysis of stressed parasites revealed down-regulation of genes belonging to diverse functional categories and up-regulation of genes encoding trans-sialidase-like and ribosomal proteins. Several of these changes were confirmed by Northern blot analyses. Sequence analysis of the 3'UTRs of up- and down-regulated genes allowed the identification of conserved structural RNA motifs enriched in each group, suggesting that specific ribonucleoprotein complexes could be of great importance in the adaptation of this parasite to different environments through regulation of transcript abundance.


Assuntos
Aminoácidos/química , Aquaporinas/química , Regulação da Expressão Gênica , Polifosfatos/química , Trypanosoma cruzi/metabolismo , Animais , Membrana Celular/metabolismo , Regulação para Baixo , Expressão Gênica , Microscopia Eletrônica/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Osmose , RNA Mensageiro/metabolismo , Fatores de Tempo , Regulação para Cima
9.
Glycobiology ; 22(10): 1363-73, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22653661

RESUMO

The trans-sialidase of Trypanosoma cruzi (TcTS) catalyzes the transfer of sialic acid from host glycoconjugates to terminal ß-galactopyranosides in the mucins of the parasite. During infection, the enzyme is actively shed by the parasite to the bloodstream inducing hematological alterations. Lactitol prevents cell apoptosis caused by the TcTS, although it is rapidly eliminated from the circulatory system. Linear polyethyleneglycol (PEG) conjugates of lactose analogs were prepared but their clearance from blood was still quite fast. With the aim of improving their circulating half-lives in vivo, we now synthesized covalent conjugates of eight-arm PEG. The star-shape of these conjugates allows an increase in the molecular weight together with the loading of the active sugar. Two approaches were used for PEGylation of disaccharide derivatives containing ß-D-Galp as the non-reducing unit. (1) Amide formation between benzyl ß-D-galactopyranosyl-(1→6)-2-amino-2-deoxy-α-D-glucopyranoside and a succinimide-activated PEG. (2) Conjugation of lactobionolactone with amino end-functionalized PEG. Two 8-arm PEG derivatives (20 and 40 kDa) were used for each sugar. Substitution of all arms was proved by (1)H nuclear magnetic resonance (NMR) spectroscopy. The bioavailability of the conjugates in mice plasma was considerably improved with respect to the 5 kDa linear PEG conjugates retaining their inhibitory properties.


Assuntos
Inibidores Enzimáticos/farmacologia , Glicoproteínas/antagonistas & inibidores , Lactose/farmacologia , Neuraminidase/antagonistas & inibidores , Polietilenoglicóis/química , Trypanosoma cruzi/enzimologia , Disponibilidade Biológica , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Glicoproteínas/metabolismo , Lactose/análogos & derivados , Lactose/química , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Neuraminidase/metabolismo , Relação Estrutura-Atividade
10.
BMC Neurosci ; 13: 78, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22762289

RESUMO

BACKGROUND: Members of the proteolipid protein family, including the four-transmembrane glycoprotein M6a, are involved in neuronal plasticity in mammals. Results from our group previously demonstrated that M6, the only proteolipid protein expressed in Drosophila, localizes to the cell membrane in follicle cells. M6 loss triggers female sterility, which suggests a role for M6 in follicular cell remodeling. These results were the basis of the present study, which focused on the function and requirements of M6 in the fly nervous system. RESULTS: The present study identified two novel, tissue-regulated M6 isoforms with variable N- and C- termini, and showed that M6 is the functional fly ortholog of Gpm6a. In the adult brain, the protein was localized to several neuropils, such as the optic lobe, the central complex, and the mushroom bodies. Interestingly, although reduced M6 levels triggered a mild rough-eye phenotype, hypomorphic M6 mutants exhibited a defective response to light. CONCLUSIONS: Based on its ability to induce filopodium formation we propose that M6 is key in cell remodeling processes underlying visual system function. These results bring further insight into the role of M6/M6a in biological processes involving neuronal plasticity and behavior in flies and mammals.


Assuntos
Comportamento Animal/fisiologia , Olho/metabolismo , Regulação da Expressão Gênica/fisiologia , Glicoproteínas de Membrana/fisiologia , Vias Visuais/metabolismo , Processamento Alternativo/genética , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Clonagem Molecular , Sequência Conservada/genética , Drosophila , Proteínas de Drosophila/genética , Olho/ultraestrutura , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Expectativa de Vida , Glicoproteínas de Membrana/genética , Microscopia Eletrônica de Varredura , Atividade Motora/genética , Mutação/genética , Neuroblastoma/patologia , Neurópilo/metabolismo , Neurópilo/ultraestrutura , Lobo Óptico de Animais não Mamíferos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Pseudópodes/metabolismo , RNA Mensageiro/metabolismo , Transfecção , Vias Visuais/ultraestrutura
11.
J Neurochem ; 119(3): 521-31, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21426347

RESUMO

A neuronal integral membrane glycoprotein M6a has been suggested to be involved in a number of biological processes, including neuronal remodeling and differentiation, trafficking of mu-opioid receptors, and Ca(2+) transportation. Moreover, pathological situations such as chronic stress in animals and depression in humans have been associated with alterations in M6a sequence and expression. The mechanism of action of M6a is essentially unknown. In this work, we analyze the relevance of M6a distribution in plasma membrane, namely its lipid microdomain targeting, for its biological function in filopodia formation. We demonstrate that M6a is localized in membrane microdomains compatible with lipid rafts in cultured rat hippocampal neurons. Removal of cholesterol from neuronal membranes with methyl-ß-cyclodextrin decreases M6a-induced filopodia formation, an effect that is reversed by the addition of cholesterol. Inhibition of Src kinases and MAPK prevents filopodia formation in M6a-over-expressing neurons. Src-deficient SYF cells over-expressing M6a fail to promote filopodia formation. Taken together, our findings reveal that the association of M6a with lipid rafts is important for its role in filopodia formation and Src and MAPK kinases participate in M6a signal propagation.


Assuntos
Colesterol/metabolismo , Glicoproteínas de Membrana/fisiologia , Microdomínios da Membrana/química , Microdomínios da Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Pseudópodes/química , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/química , Neurônios/metabolismo , Ligação Proteica/fisiologia , Pseudópodes/metabolismo , Ratos , Ratos Sprague-Dawley
12.
J Biol Chem ; 284(50): 35015-28, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19801539

RESUMO

RNA-binding proteins (RBPs) and RNA metabolism are considered to be important for modulating gene expression in trypanosomes, because these protozoan parasites mainly rely on post-transcriptional mechanisms to regulate protein levels. Previously, we have identified TcUBP1, a single RNA recognition motif (RRM)-type RBP from Trypanosoma cruzi. TcUBP1 is a cytoplasmic protein with roles in stabilization/degradation of mRNAs and in the protection of transcripts through their recruitment into cytoplasmic granules. We now show that TcUBP1, and the closely related protein TcUBP2, can be found in small amounts in the nucleus under normal conditions, and are able to accumulate in the nucleus under arsenite stress. The kinetics of nuclear accumulation, and export to the cytoplasm, are consistent with the shuttling of TcUBP1 between the nucleus and the cytoplasm. The sequence required for TcUBP1 nuclear accumulation was narrowed to the RRM, and point mutations affecting RNA binding abolished nuclear import. This RRM was also shown to be efficiently exported from the nucleus in unstressed parasites, a property that relied on the binding to RNA. TcUBP1 nuclear accumulation was dependent on active transcription, and colocalized with transcripts in the nucleus, suggesting nuclear binding of the mRNA. We propose that TcUBP1 could be linking the mRNA metabolism at both sides of the nuclear pore complex, using the RRM as a nuclear localization signal, and being exported as a cargo on mRNA.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Proteínas de Ligação a RNA/metabolismo , Trypanosoma cruzi/metabolismo , Sequência de Aminoácidos , Animais , Arsenitos/metabolismo , Sequência de Bases , Núcleo Celular/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Sinais de Localização Nuclear , Poro Nuclear/metabolismo , Estresse Oxidativo , Conformação Proteica , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transcrição Gênica , Trypanosoma cruzi/citologia
13.
J Biol Chem ; 284(46): 32075-88, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19737934

RESUMO

Gpm6a was identified as a stress-responsive gene in the hippocampal formation. This gene is down-regulated in the hippocampus of both socially and physically stressed animals, and this effect can be reversed by antidepressant treatment. Previously we showed that the stress-regulated protein M6a is a key modulator for neurite outgrowth and filopodium/spine formation. In the present work, mutational analysis was used to characterize the action of M6a at the molecular level. We show that four cysteines 162, 174, 192, and 202 within EC2 are functionally crucial sites. The presence of cysteines 162 and 202 is essential for the efficient cell surface expression of the M6a protein. In contrast, cysteines 174 and 192, which form a disulfide bridge as shown by biochemical analysis, are not required for the efficient surface expression of M6a. Their mutation to alanine does not interfere with the localization of M6a to filopodial protrusions in primary hippocampal neurons. The neurons expressing C174A and/or C192A mutants display decreased filopodia number. In non-permeabilized cells, these mutant proteins are not recognized by a function-blocking monoclonal antibody directed to M6a. Moreover, neurons in contact with axons expressing C174A/C192A mutant display significantly lower density of presynaptic clusters over their dendrites. Taken together, this study demonstrates that cysteines in the EC2 domain are critical for the role of M6a in filopodium outgrowth and synaptogenesis.


Assuntos
Membrana Celular/metabolismo , Cisteína/genética , Hipocampo/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Estresse Fisiológico , Animais , Western Blotting , Células COS , Células Cultivadas , Chlorocebus aethiops , Cisteína/metabolismo , Hipocampo/citologia , Técnicas Imunoenzimáticas , Glicoproteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Ratos , Ratos Wistar/embriologia
14.
J Exp Med ; 195(4): 401-13, 2002 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-11854354

RESUMO

Chagas' disease is a major health and economic problem caused by the protozoan Trypanosoma cruzi. Multiple independently evolving clones define a complex parasite population that can be arranged into two broad genetic lineages termed T. cruzi I and II. These lineages have different evolutionary origin and display distinct ecological and biological traits. Here we describe a novel molecule termed TSSA for trypomastigote small surface antigen that provides the first immunological marker allowing discrimination between lineages. TSSA is a surface, glycosylphosphatidyl inositol (GPI)-anchored mucin-like protein, highly antigenic during the infection. TSSA sequences from different parasite isolates reveal a population dimorphism that perfectly matches with the two T. cruzi lineages. Interestingly, this dimorphism is restricted to the central region of the molecule, which comprises the immunodominant B cell epitopes. This sequence variability has a major impact on TSSA antigenicity, leading to no immunological cross-reactivity between both isoforms for antibodies present either in immunization or infection sera. Furthermore, the absolute seroprevalence for TSSA in confirmed Chagasic patients is restricted to T. cruzi II isoform, strongly suggesting that human infections are due to this particular subgroup. Even though association of T. cruzi II with Chagas' disease has been proposed based on molecular markers, this is the first immunological evidence supporting this hypothesis. The implications of these results for the future research on Chagas' disease could be envisaged.


Assuntos
Antígenos de Protozoários/imunologia , Antígenos de Superfície/imunologia , Doença de Chagas/imunologia , Doença de Chagas/parasitologia , Trypanosoma cruzi/classificação , Trypanosoma cruzi/imunologia , Glicoproteínas Variantes de Superfície de Trypanosoma , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Antígenos de Superfície/química , Antígenos de Superfície/genética , Argentina , Sequência de Bases , Western Blotting , Brasil , Doença de Chagas/epidemiologia , Chile , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Regulação da Expressão Gênica , Genes de Protozoários/genética , Humanos , Soros Imunes/imunologia , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coelhos , Ratos , Homologia de Sequência de Aminoácidos , Trypanosoma cruzi/química , Trypanosoma cruzi/genética
15.
Eur J Neurosci ; 31(2): 195-202, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20074218

RESUMO

M6a is a neuronal membrane glycoprotein whose expression diminishes during chronic stress. M6a overexpression in rat primary hippocampal neurons induces the formation of filopodial protrusions that could be spine precursors. As the filopodium and spine motility has been associated with synaptogenesis, we analysed the motility of M6a-induced protrusions by time-lapse imaging. Our data demonstrate that the motile protrusions formed by the neurons overexpressing M6a were more abundant and moved faster than those formed in control cells. When different putative M6a phosphorylation sites were mutated, the neurons transfected with a mutant lacking intracellular phosphorylation sites bore filopodia, but these protrusions did not move as fast as those formed by cells overexpressing wild-type M6a. This suggests a role for M6a phosphorylation state in filopodium motility. Furthermore, we show that M6a-induced protrusions could be stabilized upon contact with presynaptic region. The motility of filopodia contacting or not neurites overexpressing synaptophysin was analysed. We show that the protrusions that apparently contacted synaptophysin-labeled cells exhibited less motility. The behavior of filopodia from M6a-overexpressing cells and control cells was alike. Thus, M6a-induced protrusions may be spine precursors that move to reach presynaptic membrane. We suggest that M6a is a key molecule for spine formation during development.


Assuntos
Movimento Celular/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios , Pseudópodes , Sinapses/fisiologia , Animais , Hipocampo/citologia , Glicoproteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/fisiologia , Pseudópodes/metabolismo , Pseudópodes/ultraestrutura , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sinapses/ultraestrutura
16.
J Neurosci Res ; 88(6): 1298-308, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19937804

RESUMO

Chronic stress causes morphological alterations in the hippocampus of rodents and tree shrews, including atrophy of CA3 dendrites and loss of synapses. The molecular mechanisms underlying these structural changes remain largely unknown. We have previously identified M6a as a stress responsive gene and shown that M6a is involved in filopodium/spine outgrowth and, likely, synapse formation. M6a belongs to the proteolipid protein (PLP) family, all of their members having four transmembrane domains that allow their localization at the plasma membrane. In the present work, we analyzed other members of this family, the closely related M6b as well as PLP and its splice variant DM20. We found that chronic restraint stress in mice reduces M6b and DM20, but not PLP, mRNA levels in the hippocampus. In addition, M6b and DM20, but again not PLP, induce filopodium formation in primary cultures of hippocampal neurons. Several M6b protein isoforms were studied, all of them having similar effects except for the one lacking the transmembrane domains. Our results reveal a conserved cellular function and a stress-mediated regulation among members of the proteolipid protein family, suggesting an involvement of proteolipid proteins in the stress response.


Assuntos
Hipocampo/metabolismo , Neurônios/metabolismo , Proteolipídeos/metabolismo , Estresse Psicológico/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Doença Crônica , Modelos Animais de Doenças , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Proteolipídica de Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Restrição Física
17.
Glycoconj J ; 27(5): 549-59, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20645127

RESUMO

Trypanosoma cruzi, the agent of Chagas disease, expresses a unique enzyme, the trans-sialidase (TcTS) involved in the transfer of sialic acid from host glycoconjugates to mucins of the parasite. The enzyme is shed to the medium and may affect the immune system of the host. We have previously described that lactose derivatives effectively inhibited the transfer of sialic acid to N-acetyllactosamine. Lactitol also prevented the apoptosis caused by the TcTS, although it is rapidly eliminated from the circulatory system. In this paper we report covalent conjugation of polyethylene glycol (PEG) with lactose, lactobionolactone and benzyl beta-D-galactopyranosyl-(1-->6)-2-amino-2-deoxy-alpha-D-glucopyranoside (1) with the hope to improve the bioavailability, though retaining their inhibitory properties. Different conjugation methods have been used and the behavior of the PEGylated products in the TcTS reaction was studied.


Assuntos
Dissacarídeos/química , Glicoproteínas/metabolismo , Neuraminidase/metabolismo , Polietilenoglicóis/química , Polietilenoglicóis/síntese química , Polietilenoglicóis/farmacologia , Tripanossomicidas/síntese química , Trypanosoma cruzi/enzimologia , Configuração de Carboidratos , Sequência de Carboidratos , Portadores de Fármacos , Glicoproteínas/antagonistas & inibidores , Lactose/análogos & derivados , Lactose/síntese química , Lactose/química , Lactose/metabolismo , Lactose/farmacologia , Dados de Sequência Molecular , Mucinas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/antagonistas & inibidores , Tripanossomicidas/química , Tripanossomicidas/farmacologia
18.
RNA Biol ; 7(3): 339-44, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20458169

RESUMO

In eukaryotic cells, a regulated import and export of factors is required to fulfill the requirements of precise gene expression. Post-transcriptional regulation of gene expression has proven to provide ubiquitous control, as well as a quick response to environmental changes when required. RNA-binding proteins (RBP) are involved in the several steps at which mRNA biogenesis, stability, translation and decay is exerted. The most characterized RBPs contain single or multiple copies of an RNA Recognition Motif (RRM). Here, we concentrate on RRMs mediating protein nuclear import by virtue of its ability to interact with proteins, besides interacting with nucleic acids. The consensus on how RRM-protein interactions take place is non-existent, and so is the involvement of the RRM as a nuclear localization signal (NLS). Within the cases examined, the single RRM from a trypanosome RBP behaves as a structural NLS, alternating nuclear import and RNA-binding.


Assuntos
Núcleo Celular/metabolismo , Domínios e Motivos de Interação entre Proteínas/fisiologia , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação/fisiologia , Humanos , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular
19.
Artigo em Inglês | MEDLINE | ID: mdl-32848694

RESUMO

Nowadays, great efforts are made to gain insight into the molecular mechanisms that underlie structural neuronal plasticity. Moreover, the identification of signaling pathways involved in the development of psychiatric disorders aids the screening of possible therapeutic targets. Genetic variations or alterations in GPM6A expression are linked to neurological disorders such as schizophrenia, depression, and Alzheimer's disease. GPM6A encodes the neuronal surface glycoprotein M6a that promotes filopodia/spine, dendrite, and synapse formation by unknown mechanisms. A substantial body of evidence suggests that the extracellular loops of M6a command its function. However, the proteins that associate with them and that modulate neuronal plasticity have not been determined yet. To address this question, we generated a chimera protein that only contains the extracellular loops of M6a and performed a co-immunoprecipitation with rat hippocampus samples followed by TMT/MS. Here, we report 72 proteins, which are good candidates to interact with M6a's extracellular loops and modify its function. Gene ontology (GO) analysis showed that 63% of the potential M6a's interactor proteins belong to the category "synapse," at both sides of the synaptic cleft, "neuron projections" (51%) and "presynapse" (49%). In this sense, we showed that endogenous M6a interacts with piccolo, synaptic vesicle protein 2B, and synapsin 1 in mature cultured hippocampal neurons. Interestingly, about 28% of the proteins left were related to the "myelin sheath" annotation, suggesting that M6a could interact with proteins at the surface of oligodendrocytes. Indeed, we demonstrated the (cis and trans) interaction between M6a and proteolipid protein (PLP) in neuroblastoma N2a cells. Finally, the 72 proteins were subjected to disease-associated genes and variants screening by DisGeNET. Apart from the diseases that have already been associated with M6a, most of the proteins are also involved in "autistic disorder," "epilepsy," and "seizures" increasing the spectrum of disorders in which M6a could play a role. Data are available via ProteomeXchange with identifier PXD017347.

20.
Nat Rev Microbiol ; 4(3): 229-36, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16489349

RESUMO

The surface of the protozoan parasite Trypanosoma cruzi is covered in mucins, which contribute to parasite protection and to the establishment of a persistent infection. Their importance is highlighted by the fact that the approximately 850 mucin-encoding genes comprise approximately 1% of the parasite genome and approximately 6% of all predicted T. cruzi genes. The coordinate expression of a large repertoire of mucins containing variable regions in the mammal-dwelling stages of the T. cruzi life cycle suggests a possible strategy to thwart the host immune response. Here, we discuss the expression profiling of T. cruzi mucins, the mechanisms leading to the acquisition of mucin diversity and the possible consequences of a mosaic surface coat in the interplay between parasite and host.


Assuntos
Trypanosoma cruzi/química , Animais , Doença de Chagas/parasitologia , Evolução Molecular , Genes de Protozoários/genética , Interações Hospedeiro-Parasita , Humanos , Mucinas/química , Mucinas/genética , Mucinas/fisiologia , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/fisiologia , Trypanosoma cruzi/patogenicidade , Trypanosoma cruzi/fisiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA