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










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38187761

RESUMO

Primary cilia are conserved sensory hubs essential for signaling transduction and embryonic development. Ciliary dysfunction causes a variety of developmental syndromes with neurological features and cognitive impairment, whose basis mostly remains unknown. Despite connections to neural function, the primary cilium remains an overlooked organelle in the brain. Most neurons have a primary cilium; however, it is still unclear how this organelle modulates brain architecture and function, given the lack of any systemic dissection of neuronal ciliary signaling. Here, we present the first in vivo glance at the molecular composition of cilia in the mouse brain. We have adapted in vivo BioID (iBioID), targeting the biotin ligase BioID2 to primary cilia in neurons. We identified tissue-specific signaling networks enriched in neuronal cilia, including Eph/Ephrin and GABA receptor signaling pathways. Our iBioID ciliary network presents a wealth of neural ciliary hits that provides new insights into neurological disorders. Our findings are a promising first step in defining the fundamentals of ciliary signaling and their roles in shaping neural circuits and behavior. This work can be extended to pathological conditions of the brain, aiming to identify the molecular pathways disrupted in the brain cilium. Hence, finding novel therapeutic strategies will help uncover and leverage the therapeutic potential of the neuronal cilium.

2.
Life Sci Alliance ; 5(8)2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35470240

RESUMO

Microtubule (MT) dynamics are modulated through the coordinated action of various MT-associated proteins (MAPs). However, the regulatory mechanisms underlying MT dynamics remain unclear. We show that the MAP7 family protein Map7D2 stabilizes MTs to control cell motility and neurite outgrowth. Map7D2 directly bound to MTs through its N-terminal half and stabilized MTs in vitro. Map7D2 localized prominently to the centrosome and partially on MTs in mouse N1-E115 neuronal cells, which expresses two of the four MAP7 family members, Map7D2 and Map7D1. Map7D2 loss decreased the resistance to the MT-destabilizing agent nocodazole without affecting acetylated/detyrosinated stable MTs, suggesting that Map7D2 stabilizes MTs via direct binding. In addition, Map7D2 loss increased the rate of random cell migration and neurite outgrowth, presumably by disturbing the balance between MT stabilization and destabilization. Map7D1 exhibited similar subcellular localization and gene knockdown phenotypes to Map7D2. However, in contrast to Map7D2, Map7D1 was required for the maintenance of acetylated stable MTs. Taken together, our data suggest that Map7D2 and Map7D1 facilitate MT stabilization through distinct mechanisms in cell motility and neurite outgrowth.


Assuntos
Microtúbulos , Neurônios , Animais , Movimento Celular/genética , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo , Nocodazol/metabolismo , Nocodazol/farmacologia
3.
Nature ; 588(7837): 296-302, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33177716

RESUMO

Perisynaptic astrocytic processes are an integral part of central nervous system synapses1,2; however, the molecular mechanisms that govern astrocyte-synapse adhesions and how astrocyte contacts control synapse formation and function are largely unknown. Here we use an in vivo chemico-genetic approach that applies a cell-surface fragment complementation strategy, Split-TurboID, and identify a proteome that is enriched at astrocyte-neuron junctions in vivo, which includes neuronal cell adhesion molecule (NRCAM). We find that NRCAM is expressed in cortical astrocytes, localizes to perisynaptic contacts and is required to restrict neuropil infiltration by astrocytic processes. Furthermore, we show that astrocytic NRCAM interacts transcellularly with neuronal NRCAM coupled to gephyrin at inhibitory postsynapses. Depletion of astrocytic NRCAM reduces numbers of inhibitory synapses without altering glutamatergic synaptic density. Moreover, loss of astrocytic NRCAM markedly decreases inhibitory synaptic function, with minor effects on excitation. Thus, our results present a proteomic framework for how astrocytes interface with neurons and reveal how astrocytes control GABAergic synapse formation and function.


Assuntos
Astrócitos/química , Astrócitos/metabolismo , Neurônios/metabolismo , Proteoma/metabolismo , Proteômica , Sinapses/química , Sinapses/metabolismo , Animais , Astrócitos/citologia , Moléculas de Adesão Celular Neuronais/metabolismo , Forma Celular , Feminino , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Teste de Complementação Genética , Células HEK293 , Humanos , Masculino , Camundongos , Inibição Neural , Neurônios/citologia , Ácido gama-Aminobutírico/metabolismo
4.
Elife ; 82019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31829939

RESUMO

Human mutations in the dystroglycan complex (DGC) result in not only muscular dystrophy but also cognitive impairments. However, the molecular architecture critical for the synaptic organization of the DGC in neurons remains elusive. Here, we report Inhibitory Synaptic protein 1 (InSyn1) is a critical component of the DGC whose loss alters the composition of the GABAergic synapses, excitatory/inhibitory balance in vitro and in vivo, and cognitive behavior. Association of InSyn1 with DGC subunits is required for InSyn1 synaptic localization. InSyn1 null neurons also show a significant reduction in DGC and GABA receptor distribution as well as abnormal neuronal network activity. Moreover, InSyn1 null mice exhibit elevated neuronal firing patterns in the hippocampus and deficits in fear conditioning memory. Our results support the dysregulation of the DGC at inhibitory synapses and altered neuronal network activity and specific cognitive tasks via loss of a novel component, InSyn1.


Assuntos
Distroglicanas/metabolismo , Neurônios GABAérgicos/metabolismo , Hipocampo/fisiologia , Memória , Sinapses/metabolismo , Sinapsinas/metabolismo , Animais , Células Cultivadas , Cognição , Humanos , Camundongos Endogâmicos C57BL
5.
Neuron ; 103(4): 583-597.e8, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31272828

RESUMO

Analysis of endogenous protein localization, function, and dynamics is fundamental to the study of all cells, including the diversity of cell types in the brain. However, current approaches are often low throughput and resource intensive. Here, we describe a CRISPR-Cas9-based homology-independent universal genome engineering (HiUGE) method for endogenous protein manipulation that is straightforward, scalable, and highly flexible in terms of genomic target and application. HiUGE employs adeno-associated virus (AAV) vectors of autonomous insertional sequences (payloads) encoding diverse functional modifications that can integrate into virtually any genomic target loci specified by easily assembled gene-specific guide-RNA (GS-gRNA) vectors. We demonstrate that universal HiUGE donors enable rapid alterations of proteins in vitro or in vivo for protein labeling and dynamic visualization, neural-circuit-specific protein modification, subcellular rerouting and sequestration, and truncation-based structure-function analysis. Thus, the "plug-and-play" nature of HiUGE enables high-throughput and modular analysis of mechanisms driving protein functions in cellular neurobiology.


Assuntos
Técnicas de Introdução de Genes/métodos , Genômica/métodos , Engenharia de Proteínas/métodos , Processamento de Proteína Pós-Traducional , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Sistemas CRISPR-Cas , Células Cultivadas , Dependovirus/genética , Edição de Genes/métodos , Vetores Genéticos/genética , Humanos , Imunoquímica/métodos , Inteínas , Camundongos , Mutagênese Insercional , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteômica , RNA Guia de Cinetoplastídeos/genética , Proteínas Recombinantes de Fusão/genética , Homologia de Sequência do Ácido Nucleico
6.
Methods Mol Biol ; 2008: 107-119, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31124092

RESUMO

Two anatomically and functionally distinct types of synapses are present in the central nervous system, excitatory synapses, and inhibitory synapses. Purification and analysis of the protein complex at the excitatory postsynapses have led to fundamental insights into neurobiology. In contrast, the biochemical purification and analysis of the inhibitory postsynaptic density have been largely intractable. The recently developed method called BioID employs the biotin ligase mutant, BirA*, fused to a bait protein to label and capture proximal proteins. We adapted the BioID approach to enable in vivo BioID, or iBioID of inhibitory synaptic complexes in the mouse brain. This protocol describes the iBioID method to allow synaptic bait proteins to target synaptic complexes, label, and purify biotinylated proteins from the mouse brain. This technique can be easily adapted to target other substructures in vivo that have been difficult to purify and analyze in the past.


Assuntos
Química Encefálica , Encéfalo/metabolismo , Proteínas do Tecido Nervoso , Sinapses , Animais , Biotinilação , Carbono-Nitrogênio Ligases/química , Proteínas de Escherichia coli/química , Camundongos , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/metabolismo , Proteínas Repressoras/química , Sinapses/química , Sinapses/metabolismo
7.
Nat Commun ; 10(1): 386, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30674877

RESUMO

Excitatory synapse formation during development involves the complex orchestration of both structural and functional alterations at the postsynapse. However, the molecular mechanisms that underlie excitatory synaptogenesis are only partially resolved, in part because the internal machinery of developing synapses is largely unknown. To address this, we apply a chemicogenetic approach, in vivo biotin identification (iBioID), to discover aspects of the proteome of nascent synapses. This approach uncovered sixty proteins, including a previously uncharacterized protein, CARMIL3, which interacts in vivo with the synaptic cytoskeletal regulator proteins SrGAP3 (or WRP) and actin capping protein. Using new CRISPR-based approaches, we validate that endogenous CARMIL3 is localized to developing synapses where it facilitates the recruitment of capping protein and is required for spine structural maturation and AMPAR recruitment associated with synapse unsilencing. Together these proteomic and functional studies reveal a previously unknown mechanism important for excitatory synapse development in the developing perinatal brain.


Assuntos
Citoesqueleto/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteoma/metabolismo , Proteômica , Sinapses/metabolismo , Proteínas de Capeamento de Actina/genética , Proteínas de Capeamento de Actina/metabolismo , Animais , Biotina , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Proteínas do Citoesqueleto/metabolismo , Espinhas Dendríticas/metabolismo , Proteínas Ativadoras de GTPase , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Microtúbulos/metabolismo , Neurogênese/genética , Neurogênese/fisiologia , Neurônios/metabolismo , Proteoma/genética , Sinapses/genética
8.
EMBO Rep ; 19(7)2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29880710

RESUMO

The Wnt signaling pathway can be grouped into two classes, the ß-catenin-dependent and ß-catenin-independent pathways. Wnt5a signaling through a ß-catenin-independent pathway promotes microtubule (MT) remodeling during cell-substrate adhesion, cell migration, and planar cell polarity formation. Although Wnt5a signaling and MT remodeling are known to form an interdependent regulatory loop, the underlying mechanism remains unknown. Here we show that in HeLa cells, the paralogous MT-associated proteins Map7 and Map7D1 (Map7/7D1) form an interdependent regulatory loop with Disheveled, the critical signal transducer in Wnt signaling. Map7/7D1 bind to Disheveled, direct its cortical localization, and facilitate the cortical targeting of MT plus-ends in response to Wnt5a signaling. Wnt5a signaling also promotes Map7/7D1 movement toward MT plus-ends, and depletion of the Kinesin-1 member Kif5b abolishes the Map7/7D1 dynamics and Disheveled localization. Furthermore, Disheveled stabilizes Map7/7D1. Intriguingly, Map7/7D1 and its Drosophila ortholog, Ensconsin show planar-polarized distribution in both mouse and fly epithelia, and Ensconsin influences proper localization of Drosophila Disheveled in pupal wing cells. These results suggest that the role of Map7/7D1/Ensconsin in Disheveled localization is evolutionarily conserved.


Assuntos
Evolução Molecular , Proteínas Associadas aos Microtúbulos/genética , Proteína Wnt-5a/genética , Animais , Movimento Celular/genética , Polaridade Celular/genética , Proteínas Desgrenhadas/genética , Drosophila/genética , Células HeLa , Humanos , Cinesinas/genética , Camundongos , Ligação Proteica , Via de Sinalização Wnt/genética , beta Catenina/genética
9.
Science ; 353(6304): 1123-9, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27609886

RESUMO

Inhibitory synapses dampen neuronal activity through postsynaptic hyperpolarization. The composition of the inhibitory postsynapse and the mechanistic basis of its regulation, however, remain poorly understood. We used an in vivo chemico-genetic proximity-labeling approach to discover inhibitory postsynaptic proteins. Quantitative mass spectrometry not only recapitulated known inhibitory postsynaptic proteins but also revealed a large network of new proteins, many of which are either implicated in neurodevelopmental disorders or are of unknown function. Clustered regularly interspaced short palindromic repeats (CRISPR) depletion of one of these previously uncharacterized proteins, InSyn1, led to decreased postsynaptic inhibitory sites, reduced the frequency of miniature inhibitory currents, and increased excitability in the hippocampus. Our findings uncover a rich and functionally diverse assemblage of previously unknown proteins that regulate postsynaptic inhibition and might contribute to developmental brain disorders.


Assuntos
Encefalopatias/metabolismo , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Inibição Neural , Densidade Pós-Sináptica/metabolismo , Proteoma/metabolismo , Animais , Encefalopatias/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Espectrometria de Massas , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteínas do Tecido Nervoso/genética
10.
Nat Neurosci ; 18(6): 883-91, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25938885

RESUMO

Psychiatric and neurodevelopmental disorders may arise from anomalies in long-range neuronal connectivity downstream of pathologies in dendritic spines. However, the mechanisms that may link spine pathology to circuit abnormalities relevant to atypical behavior remain unknown. Using a mouse model to conditionally disrupt a critical regulator of the dendritic spine cytoskeleton, the actin-related protein 2/3 complex (Arp2/3), we report here a molecular mechanism that unexpectedly reveals the inter-relationship of progressive spine pruning, elevated frontal cortical excitation of pyramidal neurons and striatal hyperdopaminergia in a cortical-to-midbrain circuit abnormality. The main symptomatic manifestations of this circuit abnormality are psychomotor agitation and stereotypical behaviors, which are relieved by antipsychotics. Moreover, this antipsychotic-responsive locomotion can be mimicked in wild-type mice by optogenetic activation of this circuit. Collectively these results reveal molecular and neural-circuit mechanisms, illustrating how diverse pathologies may converge to drive behaviors relevant to psychiatric disorders.


Assuntos
Antipsicóticos/farmacologia , Espinhas Dendríticas/fisiologia , Dopamina/metabolismo , Locomoção/efeitos dos fármacos , Neostriado/metabolismo , Rede Nervosa/metabolismo , Proteína 3 Relacionada a Actina/genética , Proteína 3 Relacionada a Actina/metabolismo , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/genética , Angiopoietinas/metabolismo , Animais , Feminino , Masculino , Mesencéfalo/efeitos dos fármacos , Camundongos , Camundongos Knockout , Mutação , Neostriado/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Optogenética , Técnicas de Patch-Clamp , Córtex Pré-Frontal/efeitos dos fármacos , Agitação Psicomotora/psicologia , Células Piramidais/efeitos dos fármacos , Comportamento Estereotipado/efeitos dos fármacos
11.
Elife ; 32014 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-25517933

RESUMO

During cortical synaptic development, thalamic axons must establish synaptic connections despite the presence of the more abundant intracortical projections. How thalamocortical synapses are formed and maintained in this competitive environment is unknown. Here, we show that astrocyte-secreted protein hevin is required for normal thalamocortical synaptic connectivity in the mouse cortex. Absence of hevin results in a profound, long-lasting reduction in thalamocortical synapses accompanied by a transient increase in intracortical excitatory connections. Three-dimensional reconstructions of cortical neurons from serial section electron microscopy (ssEM) revealed that, during early postnatal development, dendritic spines often receive multiple excitatory inputs. Immuno-EM and confocal analyses revealed that majority of the spines with multiple excitatory contacts (SMECs) receive simultaneous thalamic and cortical inputs. Proportion of SMECs diminishes as the brain develops, but SMECs remain abundant in Hevin-null mice. These findings reveal that, through secretion of hevin, astrocytes control an important developmental synaptic refinement process at dendritic spines.


Assuntos
Astrócitos/citologia , Espinhas Dendríticas/fisiologia , Animais , Astrócitos/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/ultraestrutura , Proteínas da Matriz Extracelular/metabolismo , Camundongos , Microscopia Confocal , Microscopia Imunoeletrônica , Sinapses/metabolismo
12.
Proc Natl Acad Sci U S A ; 109(43): E2929-38, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23027962

RESUMO

Spatial regulation of tyrosine phosphorylation is important for many aspects of cell biology. However, phosphotyrosine accounts for less than 1% of all phosphorylated substrates, and it is typically a very transient event in vivo. These factors complicate the identification of key tyrosine kinase substrates, especially in the context of their extraordinary spatial organization. Here, we describe an approach to identify tyrosine kinase substrates based on their subcellular distribution from within cells. This method uses an unnatural amino acid-modified Src homology 2 (SH2) domain that is expressed within cells and can covalently trap phosphotyrosine proteins on exposure to light. This SH2 domain-based photoprobe was targeted to cellular structures, such as the actin cytoskeleton, mitochondria, and cellular membranes, to capture tyrosine kinase substrates unique to each cellular region. We demonstrate that RhoA, one of the proteins associated with actin, can be phosphorylated on two tyrosine residues within the switch regions, suggesting that phosphorylation of these residues might modulate RhoA signaling to the actin cytoskeleton. We conclude that expression of SH2 domains within cellular compartments that are capable of covalent phototrapping can reveal the spatial organization of tyrosine kinase substrates that are likely to be important for the regulation of subcellular structures.


Assuntos
Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Frações Subcelulares/metabolismo , Domínios de Homologia de src , Compartimento Celular , Células HEK293 , Humanos , Espectrometria de Massas , Fosforilação
13.
PLoS One ; 7(5): e37035, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606326

RESUMO

Many protein interaction domains bind short peptides based on canonical sequence consensus motifs. Here we report the development of a peptide array-based proteomics tool to identify proteins directly interacting with ligand peptides from cell lysates. Array-formatted bait peptides containing an amino acid-derived cross-linker are photo-induced to crosslink with interacting proteins from lysates of interest. Indirect associations are removed by high stringency washes under denaturing conditions. Covalently trapped proteins are subsequently identified by LC-MS/MS and screened by cluster analysis and domain scanning. We apply this methodology to peptides with different proline-containing consensus sequences and show successful identifications from brain lysates of known and novel proteins containing polyproline motif-binding domains such as EH, EVH1, SH3, WW domains. These results suggest the capacity of arrayed peptide ligands to capture and subsequently identify proteins by mass spectrometry is relatively broad and robust. Additionally, the approach is rapid and applicable to cell or tissue fractions from any source, making the approach a flexible tool for initial protein-protein interaction discovery.


Assuntos
Domínios e Motivos de Interação entre Proteínas , Proteômica/métodos , Sequência de Aminoácidos , Animais , Química Encefálica , Reagentes de Ligações Cruzadas , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Biblioteca de Peptídeos , Espectrometria de Massas em Tandem , Família de Proteínas da Síndrome de Wiskott-Aldrich/química , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo
14.
Sci Signal ; 4(201): rs13, 2011 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-22126966

RESUMO

Rho family GAPs [guanosine triphosphatase (GTPase) activating proteins] negatively regulate Rho family GTPase activity and therefore modulate signaling events that control cytoskeletal dynamics. The spatial distribution of these GAPs and their specificity toward individual GTPases are controlled by their interactions with various proteins within signaling complexes. These interactions are likely mediated through the Src homology 3 (SH3) domain, which is abundant in the Rho family GAP proteome and exhibits a micromolar binding affinity, enabling the Rho family GAPs to participate in transient interactions with multiple binding partners. To capture these elusive GAP signaling complexes in situ, we developed a domain-based proteomics approach, starting with in vivo phototrapping of SH3 domain-binding proteins and the mass spectrometry identification of associated proteins for nine representative Rho family GAPs. After the selection of candidate binding proteins by cluster analysis, we performed peptide array-based high-throughput in vitro binding assays to confirm the direct interactions and map the SH3 domain-binding sequences. We thereby identified 54 SH3-mediated binding interactions (including 51 previously unidentified ones) for nine Rho family GAPs. We constructed Rho family GAP interactomes that provided insight into the functions of these GAPs. We further characterized one of the predicted functions for the Rac-specific GAP WRP and identified a role for WRP in mediating clustering of the postsynaptic scaffolding protein gephyrin and the GABA(A) (γ-aminobutyric acid type A) receptor at inhibitory synapses.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Modelos Biológicos , Receptores de GABA-A/metabolismo , Transdução de Sinais/fisiologia , Domínios de Homologia de src/fisiologia , Proteínas Ativadoras de GTPase/genética , Células HEK293 , Humanos , Proteômica/métodos , Receptores de GABA-A/genética
15.
Genes Cells ; 16(8): 868-78, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21762413

RESUMO

We have previously shown that SGIP1α is an endocytic protein specifically expressed in neural tissues. SGIP1α has a lipid-binding domain called the MP domain, which shows no significant homology to any other domains. In this study, we characterized FCHO2, a protein with a high level of homology to SGIP1α. FCHO2 lacks the MP domain but has another lipid-binding domain, the EFC/F-BAR domain. FCHO2 was ubiquitously expressed. The FCHO2 EFC domain bound to phosphatidylserine and phosphoinositides and deformed the plasma membrane and liposomes into narrow tubes. FCHO2 localized to clathrin-coated pits at the plasma membrane and bound to Eps15, an important adaptor protein in clathrin-mediated endocytosis. FCHO2 knockdown reduced transferrin endocytosis. These results suggest that FCHO2 regulates clathrin-mediated endocytosis through its interactions with membranes and Eps15. These properties of FCHO2 are similar to those of SGIP1α. FCHO2 is likely to be a ubiquitous homologue of SGIP1α. We furthermore found that FCHO2 was subjected to monoubiquitination, and gel filtration analysis showed that FCHO2 formed an oligomer. These new properties might also contribute to the role of FCHO2 in clathrin-mediated endocytosis.


Assuntos
Proteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Células COS , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Chlorocebus aethiops , Clatrina/genética , Clatrina/metabolismo , Endocitose/fisiologia , Proteínas de Ligação a Ácido Graxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Espaço Intracelular/metabolismo , Metabolismo dos Lipídeos/fisiologia , Camundongos , Complexos Multiproteicos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica/fisiologia , Multimerização Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas/genética , Ratos , Fator de Transcrição AP-2/metabolismo , Transferrina/metabolismo , Ubiquitinação/fisiologia
16.
Genes Cells ; 13(4): 295-312, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18363962

RESUMO

Microtubules (MTs) play crucial roles in a variety of cell functions, such as mitosis, vesicle transport and cell motility. MTs also compose specialized structures, such as centrosomes, spindles and cilia. However, molecular mechanisms of these MT-based functions and structures are not fully understood. Here, we analyzed MT co-sedimented proteins from rat brain by tandem mass spectrometry (MS) upon ion exchange column chromatography. We identified a total of 391 proteins. These proteins were grouped into 12 categories: 57 MT cytoskeletal proteins, including MT-associated proteins (MAPs) and motor proteins; 66 other cytoskeletal proteins; 4 centrosomal proteins; 10 chaperons; 5 Golgi proteins; 7 mitochondrial proteins; 62 nucleic acid-binding proteins; 14 nuclear proteins; 13 ribosomal proteins; 28 vesicle transport proteins; 83 proteins with diverse function and/or localization; and 42 uncharacterized proteins. Of these uncharacterized proteins, six proteins were expressed in cultured cells, resulting in the identification of three novel components of centrosomes and cilia. Our present method is not specific for MAPs, but is useful for identifying low abundant novel MAPs and components of MT-based structures. Our analysis provides an extensive list of potential candidates for future study of the molecular mechanisms of MT-based functions and structures.


Assuntos
Química Encefálica , Proteínas dos Microtúbulos/análise , Proteínas do Tecido Nervoso/análise , Animais , Sequência de Bases , Linhagem Celular , Centrossomo/química , Cílios/química , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/isolamento & purificação , Primers do DNA/genética , DNA Complementar/genética , Cães , Humanos , Proteínas dos Microtúbulos/classificação , Proteínas dos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/análise , Proteínas Associadas aos Microtúbulos/classificação , Proteínas Associadas aos Microtúbulos/genética , Proteínas Mitocondriais/análise , Proteínas Mitocondriais/isolamento & purificação , Chaperonas Moleculares/análise , Chaperonas Moleculares/isolamento & purificação , Proteínas Motores Moleculares/análise , Proteínas Motores Moleculares/isolamento & purificação , Proteínas do Tecido Nervoso/classificação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/análise , Proteínas Nucleares/isolamento & purificação , Ratos , Proteínas Recombinantes/genética , Proteínas Ribossômicas/análise , Proteínas Ribossômicas/isolamento & purificação , Espectrometria de Massas em Tandem , Transfecção
17.
Biochem Biophys Res Commun ; 366(4): 958-62, 2008 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-18086554

RESUMO

Centrosomes function as the major microtubule (MT)-organizing center. They are composed of a pair of centrioles which are surrounded by the pericentriolar material. Here, we describe the molecular characterization of a novel protein, named centlein (centrosomal protein). Centlein was a protein of 721 amino acids with a calculated molecular weight of 82,717 and possessed coiled-coil domains. Western blot analysis indicated that centlein was ubiquitously expressed. Endogenous centlein as well as enhanced green fluorescent protein-fused centlein was localized at centrosomes in interphase and mitosis. When centrosomes were isolated from cells treated with nocodazole, an MT-disrupting agent, centlein and the centrosomal protein, gamma-tubulin, were enriched in the same fractions. These data indicate that centlein is a widespread centrosomal protein and that its association with centrosomes is independent of MTs. Centlein appeared to be enriched in the mother centriole in G1 phase, suggesting possible involvement of centlein in mother-centriole-related functions, such as duplication of centrioles and generation of primary cilia.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Animais , Células CHO , Células COS , Chlorocebus aethiops , Clonagem Molecular , Cricetinae , Cricetulus , Células HeLa , Humanos , Camundongos , Mitose , Peso Molecular , Células NIH 3T3 , Transporte Proteico , Ratos
18.
J Biol Chem ; 282(36): 26481-9, 2007 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-17626015

RESUMO

SGIP1 has been shown to be an endophilin-interacting protein that regulates energy balance, but its function is not fully understood. Here, we identified its splicing variant of SGIP1 and named it SGIP1alpha. SGIP1alpha bound to phosphatidylserine and phosphoinositides and deformed the plasma membrane and liposomes into narrow tubules, suggesting the involvement in vesicle formation during endocytosis. SGIP1alpha furthermore bound to Eps15, an important adaptor protein of clathrin-mediated endocytic machinery. SGIP1alpha was colocalized with Eps15 and the AP-2 complex. Upon epidermal growth factor (EGF) stimulation, SGIP1alpha was colocalized with EGF at the plasma membrane, indicating the localization of SGIP1alpha at clathrin-coated pits/vesicles. SGIP1alpha overexpression reduced transferrin and EGF endocytosis. SGIP1alpha knockdown reduced transferrin endocytosis but not EGF endocytosis; this difference may be due to the presence of redundant pathways in EGF endocytosis. These results suggest that SGIP1alpha plays an essential role in clathrin-mediated endocytosis by interacting with phospholipids and Eps15.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Vesículas Revestidas por Clatrina/metabolismo , Metabolismo Energético/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfolipídeos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Processamento Alternativo/fisiologia , Animais , Sequência de Bases , Células COS , Proteínas de Ligação ao Cálcio/genética , Proteínas de Transporte/genética , Chlorocebus aethiops , Vesículas Revestidas por Clatrina/genética , Endocitose , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana , Dados de Sequência Molecular , Fosfolipídeos/genética , Fosfoproteínas/genética , Ligação Proteica/fisiologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Transferrina/genética , Transferrina/metabolismo
19.
J Neurochem ; 85(5): 1216-27, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12753081

RESUMO

Ca2+/calmodulin-dependent protein kinase I (CaMKI), originally identified as a protein kinase phosphorylating synapsin I, has been shown to constitute a family of closely related isoforms (alpha, beta and gamma). Here, we have isolated and determined the complete primary structures of two alternatively splicing isoforms of CaMKI termed CaMKI gamma 1 and -gamma 2. CaMKI gamma 1 and -gamma 2 contain an identical N-terminal catalytic domain with different C-terminal regions due to the deletion of the 425-bp nucleotide sequence of CaMKI gamma 1 in CaMKI gamma 2. In vitro kinase assay has demonstrated the marked enhancement of the Ca2+/CaM-dependent activity of CaMKI gamma 1 by the preincubation with Ca2+/calmodulin-dependent protein kinase kinase (CaMKK), but no significant activation of CaMKI gamma 2. Northern blot analysis has demonstrated the predominant expression of CaMKI gamma in the brain. RT-PCR analysis has revealed similar expression patterns between CaMKI gamma 1 and CaMKI gamma 2 in various brain regions. In situ hybridization analysis has demonstrated that CaMKI gamma mRNA is expressed in a distinct pattern from other isoforms of CaMKI with predominant expression in some restricted brain regions such as the olfactory bulb, hippocampal pyramidal cell layer of CA3, central amygdaloid nuclei, ventromedial hypothalamic nucleus and pineal gland. In the primary hippocampal neurons and NG108-15 cells, transfected CaMKI gamma 1 and -gamma 2 are localized primarily in the cytoplasm and neurites but not in the nucleus. These findings suggest that both isoforms of CaMKI gamma may be involved in Ca2+ signal transduction in the cytoplasmic compartment of certain neuronal population.


Assuntos
Processamento Alternativo/genética , Encéfalo/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Proteínas do Tecido Nervoso/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Encéfalo/enzimologia , Células COS , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/biossíntese , Células Cultivadas , Clonagem Molecular , Expressão Gênica , Células Híbridas , Hibridização In Situ , Isoenzimas/biossíntese , Isoenzimas/genética , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/biossíntese , Neurônios/metabolismo , Especificidade de Órgãos , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Transfecção
20.
J Neurochem ; 82(3): 585-93, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12153482

RESUMO

We have focused on activation mechanisms of calcium/calmodulin-dependent protein kinase (CaM) kinase I in the hippocampal neurons and compared them with that of CaM kinase IV. Increased activation of CaM kinase I occurred by stimulation with glutamate and depolarization in cultured rat hippocampal neurons. Similar to CaM kinases II and IV, CaM kinase I was essentially activated by stimulation with the NMDA receptor. Although both CaM kinases I and IV seem to be activated by CaM kinase kinase, the activation of CaM kinase I was persistent during stimulation with glutamate in contrast to a transient activation of CaM kinase IV. In addition, CaM kinase I was activated in a lower concentration of glutamate than that of CaM kinase IV. Depolarization-induced activation of CaM kinase I was also evident in the cultured neurons and was largely blocked by nifedipine. In the experiment with 32P-labeled cells, phosphorylation of CaM kinase I was stimulated by glutamate treatment and depolarization. The glutamate- and depolarization-induced phosphorylation was inhibited by the NMDA receptor antagonist and nifedipine, respectively. These results suggest that, although CaM kinases I and IV are activated by the NMDA receptor and depolarization stimulation, these kinase activities are differently regulated in the hippocampal neurons.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Hipocampo/enzimologia , Neurônios/enzimologia , Animais , Especificidade de Anticorpos , Bloqueadores dos Canais de Cálcio/farmacologia , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina , Células Cultivadas , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Nifedipino/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...