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.
Nat Commun ; 11(1): 475, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980622

RESUMO

We reported previously that acid-sensing ion channel 1a (ASIC1a) mediates acidic neuronal necroptosis via recruiting receptor-interacting protein kinase 1 (RIPK1) to its C terminus (CT), independent of its ion-conducting function. Here we show that the N-terminus (NT) of ASIC1a interacts with its CT to form an auto-inhibition that prevents RIPK1 recruitment/activation under resting conditions. The interaction involves glutamate residues at distal NT and is disrupted by acidosis. Expression of mutant ASIC1a bearing truncation or glutamate-to-alanine substitutions at distal NT causes constitutive cell death. The NT-CT interaction is further disrupted by N-ethylmaleimide-sensitive fusion ATPase (NSF), which associates with ASIC1a-NT under acidosis, facilitating RIPK1 interaction with ASIC1a-CT. Importantly, a membrane-penetrating synthetic peptide representing the distal 20 ASIC1a NT residues, NT1-20, reduced neuronal damage in both in vitro model of acidotoxicity and in vivo mouse model of ischemic stroke, demonstrating the therapeutic potential of targeting the auto-inhibition of ASIC1a for neuroprotection against acidotoxicity.


Assuntos
Canais Iônicos Sensíveis a Ácido/química , Canais Iônicos Sensíveis a Ácido/fisiologia , Necroptose/fisiologia , Neurônios/fisiologia , Canais Iônicos Sensíveis a Ácido/genética , Acidose/patologia , Acidose/fisiopatologia , Substituição de Aminoácidos , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Modelos Neurológicos , Proteínas Sensíveis a N-Etilmaleimida/farmacologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Neurônios/citologia , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia , Transdução de Sinais , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
2.
Neuropharmacology ; 112(Pt A): 66-75, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27543417

RESUMO

Long lasting synaptic plasticity involves both functional and morphological changes, but how these processes are molecularly linked to achieve coordinated plasticity remains poorly understood. Cofilin is a common target of multiple signaling pathways at the synapse and is required for both functional and spine plasticity, but how it is regulated is unclear. In this study, we investigate whether the involvement of cofilin in plasticity is developmentally regulated by examining the role of cofilin in hippocampal long-term depression (LTD) in both young (2 weeks) and mature (2 months) mice. We show that both total protein level of cofilin and its activation undergo significant changes as the brain matures, so that although the amount of cofilin decreases significantly in mature mice, its regulation by protein phosphorylation becomes increasingly important. Consistent with these biochemical data, we show that cofilin-mediated actin reorganization is essential for LTD in mature, but not in young mice. In contrast to cofilin, the GluA2 interactions with NSF and PICK1 appear to be required in both young and mature mice, indicating that AMPAR internalization is a common key mechanism for LTD expression regardless of the developmental stages. These results establish the temporal specificity of cofilin in LTD regulation and suggest that cofilin-mediated actin reorganization may serve as a key mechanism underlying developmental regulation of synaptic and spine plasticity. This article is part of the Special Issue entitled 'Ionotropic glutamate receptors'.


Assuntos
Fatores de Despolimerização de Actina/fisiologia , Actinas/fisiologia , Hipocampo/fisiologia , Depressão Sináptica de Longo Prazo , Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Animais , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Proteínas de Ciclo Celular , Potenciais Pós-Sinápticos Excitadores , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia
3.
PLoS One ; 11(6): e0157837, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27355324

RESUMO

BACKGROUND: The Acute Respiratory Distress Syndrome (ARDS), remains a significant source of morbidity and mortality in critically ill patients. Pneumonia and sepsis are leading causes of ARDS, the pathophysiology of which includes increased pulmonary microvascular permeability and hemodynamic instability resulting in organ dysfunction. We hypothesized that N-ethylmaleimide sensitive factor (NSF) regulates exocytosis of inflammatory mediators, such as Angiopoietin-2 (Ang-2), and cytoskeletal stability by modulating myosin light chain (MLC) phosphorylation. Therefore, we challenged pulmonary cells, in vivo and in vitro, with Gram Positive bacterial cell wall components, lipoteichoic acid (LTA), and peptidoglycan (PGN) and examined the effects of NSF inhibition. METHODS: Mice were pre-treated with an inhibitor of NSF, TAT-NSF700 (to prevent Ang-2 release). After 30min, LTA and PGN (or saline alone) were instilled intratracheally. Pulse oximetry was assessed in awake mice prior to, and 6 hour post instillation. Post mortem, tissues were collected for studies of inflammation and Ang-2. In vitro, pulmonary endothelial cells were assessed for their responses to LTA and PGN. RESULTS: Pulmonary challenge induced signs of airspace and systemic inflammation such as changes in neutrophil counts and protein concentration in bronchoalveolar lavage fluid and tissue Ang-2 concentration, and decreased physiological parameters including oxygen saturation and pulse distention. TAT-NSF700 pre-treatment reduced LTA-PGN induced changes in lung tissue Ang-2, oxygen saturation and pulse distention. In vitro, LTA-PGN induced a rapid (<2 min) release of Ang-2, which was significantly attenuated by TAT-NSF700 or anti TLR2 antibody. Furthermore, TAT-NSF700 reduced LTA-PGN-induced MLC phosphorylation at low concentrations of 1-10 nM. CONCLUSIONS: TAT-NSF700 decreased Ang-2 release, improved oxygen saturation and pulse distention following pulmonary challenge by inhibiting MLC phosphorylation, an important component of endothelial cell retraction. The data suggest that inhibition of NSF in pneumonia and sepsis may be beneficial to prevent the pulmonary microvascular and hemodynamic instability associated with ARDS.


Assuntos
Infecções Bacterianas/complicações , Pulmão/microbiologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Síndrome do Desconforto Respiratório/complicações , Angiopoietina-1/metabolismo , Animais , Vasos Sanguíneos/patologia , Linhagem Celular , Parede Celular/efeitos dos fármacos , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Exocitose , Bactérias Gram-Positivas , Humanos , Inflamação , Lipopolissacarídeos/química , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microcirculação , Proteínas Sensíveis a N-Etilmaleimida/antagonistas & inibidores , Oxigênio/química , Peptidoglicano/química , Fosforilação , Pneumonia/metabolismo , Síndrome do Desconforto Respiratório/microbiologia , Sepse/metabolismo , Ácidos Teicoicos/química , Doenças Vasculares/metabolismo , Doenças Vasculares/prevenção & controle
5.
Circ Res ; 115(1): 10-22, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24753547

RESUMO

RATIONALE: Several reports suggest that antisense oligonucleotides against miR-33 might reduce cardiovascular risk in patients by accelerating the reverse cholesterol transport pathway. However, conflicting reports exist about the impact of anti-miR-33 therapy on the levels of very low-density lipoprotein-triglycerides (VLDL-TAG). OBJECTIVE: We test the hypothesis that miR-33 controls hepatic VLDL-TAG secretion. METHODS AND RESULTS: Using therapeutic silencing of miR-33 and adenoviral overexpression of miR-33, we show that miR-33 limits hepatic secretion of VLDL-TAG by targeting N-ethylmaleimide-sensitive factor (NSF), both in vivo and in primary hepatocytes. We identify conserved sequences in the 3'UTR of NSF as miR-33 responsive elements and show that Nsf is specifically recruited to the RNA-induced silencing complex following induction of miR-33. In pulse-chase experiments, either miR-33 overexpression or knock-down of Nsf lead to decreased secretion of apolipoproteins and TAG in primary hepatocytes, compared with control cells. Importantly, Nsf rescues miR-33-dependent reduced secretion. Finally, we show that overexpression of Nsf in vivo increases global hepatic secretion and raises plasma VLDL-TAG. CONCLUSIONS: Together, our data reveal key roles for the miR-33-NSF axis during hepatic secretion and suggest that caution should be taken with anti-miR-33-based therapies because they might raise proatherogenic VLDL-TAG levels.


Assuntos
Lipoproteínas VLDL/metabolismo , MicroRNAs/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Triglicerídeos/metabolismo , Animais , Apolipoproteína B-100 , Apolipoproteínas B/metabolismo , Apolipoproteínas B/fisiologia , Proteínas de Transporte/fisiologia , Hepatócitos/metabolismo , Lipoproteínas VLDL/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de LDL/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 2/fisiologia , Triglicerídeos/sangue
6.
Neural Plast ; 2012: 392695, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22900206

RESUMO

Signal transmission from the human retina to visual cortex and connectivity of visual brain areas are relatively well understood. How specific visual perceptions transform into corresponding long-term memories remains unknown. Here, I will review recent Blood Oxygenation Level-Dependent functional Magnetic Resonance Imaging (BOLD fMRI) in humans together with molecular biology studies (animal models) aiming to understand how the retinal image gets transformed into so-called visual (retinotropic) maps. The broken object paradigm has been chosen in order to illustrate the complexity of multisensory perception of simple objects subject to visual--rather than semantic--type of memory encoding. The author explores how amygdala projections to the visual cortex affect the memory formation and proposes the choice of experimental techniques needed to explain our massive visual memory capacity. Maintenance of the visual long-term memories is suggested to require recycling of GluR2-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) and ß(2)-adrenoreceptors at the postsynaptic membrane, which critically depends on the catalytic activity of the N-ethylmaleimide-sensitive factor (NSF) and protein kinase PKMζ.


Assuntos
Encéfalo/fisiologia , Memória de Longo Prazo/fisiologia , Córtex Visual/fisiologia , Tonsila do Cerebelo/fisiologia , Animais , Encéfalo/enzimologia , Percepção de Forma/fisiologia , Humanos , Imageamento por Ressonância Magnética , Rememoração Mental/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/enzimologia , Neurônios/fisiologia , Proteínas Quinases/genética , Proteínas Quinases/fisiologia , Receptores de AMPA/fisiologia , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Córtex Visual/citologia , Córtex Visual/enzimologia
7.
J Neurogenet ; 26(3-4): 348-59, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22817636

RESUMO

By the middle of the 20th century, development of powerful genetic approaches had ensured that the fruit fly would remain a model organism of choice for genetic and developmental studies. But in the 1970s, a few pioneering groups turned their attention to the prospect of using the fly for neurophysiological experiments. They proposed that in a poikilothermic organism such as Drosophila, temperature-sensitive or "ts" mutations in proteins that controlled nerve function would translate to a "ts" paralytic phenotype. This was by no means an obvious or even a likely assumption. However, following directed screens these groups soon reported dramatic demonstrations of reversible ts paralysis in fly mutants. Resultantly, these "simple" experiments led to the isolation of a number of conditional mutations including shibire, paralytic, and comatose. All have since been cloned and have enabled deep mechanistic insights into synaptic transmission and nerve conduction. comatose (comt) mutations, for example, were found to map to missense changes in dNSF1, a neuron-specific fly homolog of mammalian NSF (N-ethylmaleimide-sensitive fusion factor). Studies on comt were also some of the first to discriminate between nuanced models of NSF function during presynaptic transmitter release that have since been borne out by experiments in multiple preparations. Here, the authors present an overview of NSF function as it is understood today, with an emphasis on contributions from Drosophila beginning with experiments carried out by Obaid Siddiqi in the Benzer laboratory. The authors also outline initial results from a genetic screen for phenotypic modifiers of comt that hold the promise of further elucidating NSF function at the synapse. Over the years, the neuromuscular system of Drosophila has served as a uniquely accessible model to unravel mechanisms underlying synaptic transmission. To this day, ts paralysis remains one of the most emphatic demonstrations of nerve function in an intact organism.


Assuntos
Mutação/genética , Proteínas Sensíveis a N-Etilmaleimida/genética , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Neurônios/metabolismo , Animais , Animais Geneticamente Modificados , Drosophila/genética , Proteínas de Drosophila , Eletrorretinografia , Metanossulfonato de Etila/farmacologia , Exocitose/efeitos dos fármacos , Exocitose/genética , Testes Genéticos , História do Século XX , Mutagênese/efeitos dos fármacos , Mutagênese/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/história , Paralisia/genética , Paralisia/terapia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/genética , Vesículas Sinápticas/metabolismo , Fatores de Tempo
8.
Dev Neurobiol ; 72(11): 1399-414, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22076955

RESUMO

Plasmalemmal repair (sealing) is necessary for survival of damaged eukaryotic cells. Ca(2+) influx through plasmalemmal disruptions activates pathways that initiate sealing, which is commonly assessed by exclusion of extracellular dye. These sealing pathways include PKA, Epac, and cytosolic oxidation. In this article, we investigate whether PKA, Epac, and/or cytosolic oxidation, activate specific proteins required to produce a plasmalemmal seal. We report that toxin cleavage of proteins required for neurotransmitter release (SNAP-25), inhibition of Golgi trafficking (with Brefeldin A: Bref A) or inhibition of N-ethylmaleimide sensitive factor (NSF) all decrease sealing of rat B104 hippocampal cells with transected neuritis in vitro. Epac, but not PKA or cytosolic oxidation, partly overcomes the decrease in sealing produced by cleavage of SNAP-25. PKA and increased cytosolic oxidation, but not Epac, can partly overcome the decrease in sealing due to Bref A. PKA, Epac, and/or cytosolic oxidation cannot overcome NSF inhibition. Substances that affect plasmalemmal sealing of B104 neurites in vitro have similar effects on plasmalemmal sealing in rat sciatic axons ex vivo. From these and other data, we propose a model of plasmalemmal sealing having three redundant, evolutionarily conserved, parallel pathways that all converge on NSF.


Assuntos
Axônios/fisiologia , Membrana Celular/fisiologia , Regeneração Nervosa/fisiologia , Transdução de Sinais/fisiologia , Animais , Axotomia , Cálcio/fisiologia , Linhagem Celular Tumoral , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Citosol/fisiologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Hipocampo/fisiologia , Técnicas In Vitro , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Neuritos/fisiologia , Oxirredução , Ratos , Nervo Isquiático/fisiologia , Proteína 25 Associada a Sinaptossoma/fisiologia
9.
PLoS One ; 6(11): e27146, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22073277

RESUMO

Vesicle fusion contributes to the maintenance of synapses in the nervous system by mediating synaptic transmission, release of neurotrophic factors, and trafficking of membrane receptors. N-ethylmaleimide-sensitive factor (NSF) is indispensible for dissociation of the SNARE-complex following vesicle fusion. Although NSF function has been characterized extensively in vitro, the in vivo role of NSF in vertebrate synaptogenesis is relatively unexplored. Zebrafish possess two nsf genes, nsf and nsfb. Here, we examine the function of either Nsf or Nsfb in the pre- and postsynaptic cells of the zebrafish lateral line organ and demonstrate that Nsf, but not Nsfb, is required for maintenance of afferent synapses in hair cells. In addition to peripheral defects in nsf mutants, neurodegeneration of glutamatergic synapses in the central nervous system also occurs in the absence of Nsf function. Expression of an nsf transgene in a null background indicates that stabilization of synapses requires Nsf function in both hair cells and afferent neurons. To identify potential targets of Nsf-mediated fusion, we examined the expression of genes implicated in stabilizing synapses and found that transcripts for multiple genes including brain-derived neurotrophic factor (bdnf) were significantly reduced in nsf mutants. With regard to trafficking of BDNF, we observed a striking accumulation of BDNF in the neurites of nsf mutant afferent neurons. In addition, injection of recombinant BDNF protein partially rescued the degeneration of afferent synapses in nsf mutants. These results establish a role for Nsf in the maintenance of synaptic contacts between hair cells and afferent neurons, mediated in part via the secretion of trophic signaling factors.


Assuntos
Células Ciliadas Auditivas/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Sinapses/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Apoptose , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Células Ciliadas Auditivas/patologia , Mutação , Proteínas Sensíveis a N-Etilmaleimida/genética , Neurogênese , Sinapses/patologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
10.
J Neurosci ; 30(42): 13955-65, 2010 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20962217

RESUMO

Disturbances in GABA(A) receptor trafficking contribute to several neurological and psychiatric disorders by altering inhibitory neurotransmission. Identifying mechanisms that regulate GABA(A) receptor trafficking could lead to better understanding of disease pathogenesis and treatment. Here, we show that protein kinase Cε (PKCε) regulates the N-ethylmaleimide-sensitive factor (NSF), an ATPase critical for membrane fusion events, and thereby promotes the trafficking of GABA(A) receptors. Activation of PKCε decreased cell surface expression of GABA(A) receptors and attenuated GABA(A) currents. Activated PKCε associated with NSF, phosphorylated NSF at serine 460 and threonine 461, and increased NSF ATPase activity, which was required for GABA(A) receptor downregulation. These findings identify new roles for NSF and PKCε in regulating synaptic inhibition through downregulation of GABA(A) receptors. Reducing NSF activity by inhibiting PKCε could help restore synaptic inhibition in disease states in which it is impaired.


Assuntos
Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Proteína Quinase C-épsilon/fisiologia , Receptores de GABA-A/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Biotinilação , Linhagem Celular , Membrana Celular/metabolismo , Eletrofisiologia , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Técnicas de Patch-Clamp , Fosforilação , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/isolamento & purificação , Receptores de Superfície Celular/metabolismo
11.
Proc Natl Acad Sci U S A ; 106(34): 14658-63, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19706552

RESUMO

Current models of synaptic vesicle trafficking implicate a core complex of proteins comprised of N-ethylmaleimide-sensitive factor (NSF), soluble NSF attachment proteins (SNAPs), and SNAREs in synaptic vesicle fusion and neurotransmitter release. Despite this progress, major challenges remain in establishing the in vivo functions of these proteins and their roles in determining the physiological properties of synapses. The present study employs glutamatergic adult neuromuscular synapses of Drosophila, which exhibit conserved properties of short-term synaptic plasticity with respect to mammalian glutamatergic synapses, to address these issues through genetic analysis. Our findings establish an in vivo role for SNAP-25 in synaptic vesicle priming, and support a zippering model of SNARE function in this process. Moreover, these studies define the contribution of SNAP-25-dependent vesicle priming to the detailed properties of short-term depression elicited by paired-pulse (PP) and train stimulation. In contrast, NSF is shown here not to be required for WT PP depression, but to be critical for maintaining neurotransmitter release during sustained stimulation. In keeping with this role, disruption of NSF function results in activity-dependent redistribution of the t-SNARE proteins, SYNTAXIN and SNAP-25, away from neurotransmitter release sites (active zones). These findings support a role for NSF in replenishing active zone t-SNAREs for subsequent vesicle priming, and provide new insight into the spatial organization of SNARE protein cycling during synaptic activity. Together, the results reported here establish in vivo contributions of SNAP-25 and NSF to synaptic vesicle trafficking and define molecular mechanisms determining conserved functional properties of short-term depression.


Assuntos
Proteínas de Drosophila/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Sinapses/fisiologia , Vesículas Sinápticas/fisiologia , Proteína 25 Associada a Sinaptossoma/fisiologia , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Eletrofisiologia , Potenciais Evocados/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Imuno-Histoquímica , Mutação , Proteínas Sensíveis a N-Etilmaleimida/genética , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Junção Neuromuscular/fisiologia , Proteínas Qa-SNARE/metabolismo , Proteínas SNARE/metabolismo , Sinapses/metabolismo , Vesículas Sinápticas/metabolismo , Proteína 25 Associada a Sinaptossoma/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Fatores de Tempo
12.
Mol Cells ; 26(6): 517-29, 2008 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-19011361

RESUMO

Porosomes are supramolecular, lipoprotein structures at the cell plasma membrane, where membrane-bound secretory vesicles transiently dock and fuse to release inravesicular contents to the outside during cell secretion. The mouth of the porosome opening to the outside, range in size from 150 nm in diameter in acinar cells of the exocrine pancreas, to 12 nm in neurons, which dilates during cell secretion, returning to its resting size following completion of the process. In the past decade, the composition of the porosome, its structure and dynamics at nm resolution and in real time, and its functional reconstitution into artificial lipid membrane, have all been elucidated. In this mini review, the discovery of the porosome, its structure, function, isolation, chemistry, and reconstitution into lipid membrane, the molecular mechanism of secretory vesicle swelling and fusion at the base of porosomes, and how this new information provides a paradigm shift in our understanding of cell secretion, is discussed.


Assuntos
Membrana Celular/fisiologia , Células/metabolismo , Animais , Humanos , Bicamadas Lipídicas/metabolismo , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Organelas/fisiologia , Pâncreas Exócrino/metabolismo , Proteínas SNARE/metabolismo , Proteínas SNARE/ultraestrutura , Vesículas Secretórias/fisiologia , Vesículas Secretórias/ultraestrutura , Sinaptossomos/fisiologia , Sinaptossomos/ultraestrutura
13.
J Neurosci ; 28(31): 7820-7, 2008 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-18667614

RESUMO

Although the maintenance mechanism of late long-term potentiation (LTP) is critical for the storage of long-term memory, the expression mechanism of synaptic enhancement during late-LTP is unknown. The autonomously active protein kinase C isoform, protein kinase Mzeta (PKMzeta), is a core molecule maintaining late-LTP. Here we show that PKMzeta maintains late-LTP through persistent N-ethylmaleimide-sensitive factor (NSF)/glutamate receptor subunit 2 (GluR2)-dependent trafficking of AMPA receptors (AMPARs) to the synapse. Intracellular perfusion of PKMzeta into CA1 pyramidal cells causes potentiation of postsynaptic AMPAR responses; this synaptic enhancement is mediated through NSF/GluR2 interactions but not vesicle-associated membrane protein-dependent exocytosis. PKMzeta may act through NSF to release GluR2-containing receptors from a reserve pool held at extrasynaptic sites by protein interacting with C-kinase 1 (PICK1), because disrupting GluR2/PICK1 interactions mimic and occlude PKMzeta-mediated AMPAR potentiation. During LTP maintenance, PKMzeta directs AMPAR trafficking, as measured by NSF/GluR2-dependent increases of GluR2/3-containing receptors in synaptosomal fractions from tetanized slices. Blocking this trafficking mechanism reverses established late-LTP and persistent potentiation at synapses that have undergone synaptic tagging and capture. Thus, PKMzeta maintains late-LTP by persistently modifying NSF/GluR2-dependent AMPAR trafficking to favor receptor insertion into postsynaptic sites.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciação de Longa Duração/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Proteína Quinase C/fisiologia , Receptores de AMPA/fisiologia , Sequência de Aminoácidos , Animais , Isoenzimas/fisiologia , Masculino , Dados de Sequência Molecular , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores de AMPA/metabolismo
14.
Dev Neurobiol ; 68(3): 379-91, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18161855

RESUMO

The precise role of actin and actin-binding proteins in synaptic development is unclear. In Drosophila, overexpression of a dominant-negative NSF2 construct perturbs filamentous actin, which is associated with overgrowth of the NMJ, while co-expression of moesin, which encodes an actin binding protein, suppresses this overgrowth phenotype. These data suggest that Moesin may play a role in synaptic development at the Drosophila NMJ. To further investigate this possibility, we examined the influence of loss-of-function moesin alleles on the NSF2-induced overgrowth phenotype. We found that flies carrying P-element insertions that reduce moesin expression enhanced the NMJ overgrowth phenotype, indicating a role for Moesin in normal NMJ morphology. In addition to the NMJ overgrowth phenotype, expression of dominant-negative NSF2 is known to reduce the frequency of miniature excitatory junctional potentials and the amplitude of excitatory junctional potentials. We found that moesin coexpression did not restore the physiology of the mutant NSF2 phenotype. Together, our results demonstrate a role for moesin in regulating synaptic growth in the Drosophila NMJ and suggest that the effect of dominant-negative NSF2 on NMJ morphology and physiology may have different underlying molecular origins.


Assuntos
Proteínas dos Microfilamentos/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Junção Neuromuscular/fisiologia , Actinas/metabolismo , Análise de Variância , Animais , Animais Geneticamente Modificados , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Embrião não Mamífero , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Modelos Lineares , Potenciais da Membrana/genética , Potenciais da Membrana/efeitos da radiação , Músculos/citologia , Mutação/fisiologia
15.
Circ Res ; 101(12): 1247-54, 2007 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-17932325

RESUMO

Exocytosis of endothelial granules promotes thrombosis and inflammation and may contribute to the pathophysiology of early reperfusion injury following myocardial ischemia. TAT-NSF700 is a novel peptide that reduces endothelial exocytosis by inhibiting the ATPase activity and disassembly activity of N-ethylmaleimide-sensitive factor (NSF), a critical component of the exocytic machinery. We hypothesized that TAT-NSF700 would limit myocardial injury in an in vivo murine model of myocardial ischemia/reperfusion injury. Mice were subjected to 30 minutes of ischemia followed by 24 hours of reperfusion. TAT-NSF700 or the scrambled control peptide TAT-NSF700scr was administered intravenously 20 minutes before the onset of ischemia. Myocardial ischemia/reperfusion caused endothelial exocytosis, myocardial infarction, and left ventricular dysfunction. However, TAT-NSF700 decreased von Willebrand factor levels after myocardial ischemia/reperfusion, attenuated myocardial infarct size by 47%, and preserved left ventricular structure and function. These data suggest that drugs targeting endothelial exocytosis may be useful in the treatment of myocardial injury following ischemia/reperfusion.


Assuntos
Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteínas Sensíveis a N-Etilmaleimida/antagonistas & inibidores , Animais , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/enzimologia , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Peptídeos/fisiologia , Peptídeos/uso terapêutico , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Esquerda/fisiologia
16.
J Cell Sci ; 120(Pt 14): 2318-27, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17606987

RESUMO

Starving, highly motile Dictyostelium cells maintain an active endocytic cycle, taking up their surface about every 11 minutes. Cell motility depends on a functional NSF (N-ethylmaleimide sensitive factor) protein--also essential for endocytosis and membrane trafficking generally--and we, therefore, investigated possible ways in which the endocytic cycle might be required for cell movement. First, NSF, and presumably membrane trafficking, are not required for the initial polarization of the leading edge in a cyclic-AMP gradient. Second, we can detect no evidence for membrane flow from the leading edge, as photobleached or photoactivated marks in the plasma membrane move forward roughly in step with the leading edge, rather than backwards from it. Third, we find that the surface area of a cell--measured from confocal reconstructions--constantly fluctuates during movement as it projects pseudopodia and otherwise changes shape; increases of 20-30% can often occur over a few minutes. These fluctuations cannot be explained by reciprocal changes in filopodial surface area and they substantially exceed the 2-3% by which membranes can stretch. We propose that the endocytic cycle has a key function in motility by allowing adjustment of cell surface area to match changes in shape and that, without this function, movement is severely impaired.


Assuntos
Movimento Celular/fisiologia , Dictyostelium/fisiologia , Endocitose/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Polaridade Celular , Clatrina/metabolismo , Clatrina/fisiologia , Dictyostelium/citologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia
17.
Mol Pharmacol ; 72(2): 429-39, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17510209

RESUMO

Recycling of G protein-coupled receptors determines the functional resensitization of receptors and is implicated in switching beta2 adrenoceptor (beta2AR) G protein specificity in cardiomyocytes. The human beta2AR carboxyl end binds to the N-ethylmaleimide-sensitive factor (NSF), an ATPase integral to membrane trafficking machinery. It is interesting that the human beta2AR (hbeta2AR) carboxyl end pulled down NSF from mouse heart lysates, whereas the murine one did not. Despite this difference, both beta2ARs exhibited substantial agonist-induced internalization, recycling, and Gi coupling in cardiomyocytes. The hbeta2AR, however, displayed faster rates of agonist-induced internalization and recycling compared with the murine beta2AR (mbeta2AR) and a more profound Gi component in its contraction response. Replacing the mbeta2AR proline (-1) with a leucine generated a gain-of-function mutation, mbeta2AR-P417L, with a rescued ability to bind NSF, faster internalization and recycling than the mbeta2AR, and a significant enhancement in Gi signaling, which mimics the hbeta2AR. Selective disruption of the mbeta2AR-P417L binding to NSF inhibited the receptor coupling to Gi. Mean-while, inhibiting NSF with N-ethylmaleimide blocked the mbeta2AR recycling after agonist-induced endocytosis. Expressing the NSF-E329Q mutant lacking ATPase activity inhibited the mbeta2AR coupling to Gi in cardiomyocytes. Our results revealed a dual regulation on hbeta2AR trafficking and signaling by NSF through direct binding to cargo receptor and its ATPase activity and uncovered an unprecedented role for the receptor binding to NSF in regulating G protein specificity that has diverged between mouse and human beta2ARs.


Assuntos
Miócitos Cardíacos/metabolismo , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Humanos , Camundongos , Contração Miocárdica , Transporte Proteico
18.
J Neurophysiol ; 96(3): 1053-60, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16760338

RESUMO

Peptides that inhibit the SNAP-stimulated ATPase activity of N-ethylmaleimide-sensitive fusion protein (NSF-2, NSF-3) were injected intra-axonally to study the role of this protein in the release of glutamate at the crayfish neuromuscular junction. Macropatch recording was used to establish the quantal content and to construct synaptic delay histograms. NSF-2 or NSF-3 injection reduced the quantal content, evoked by either direct depolarization of a single release bouton or by axonal action potentials, on average by 66 +/- 12% (mean +/- SD; n = 32), but had no effect on the time course of release. NSF-2 had no effect on the amplitude or shape of the presynaptic action potential nor on the excitatory nerve terminal current. Neither NSF-2 nor NSF-3 affected the shape or amplitude of single quantal currents. Injection of a peptide with the same composition as NSF-2, but with a scrambled amino acid sequence, failed to alter the quantal content. We conclude that, at the crayfish neuromuscular junction, NSF-dependent reactions regulate quantal content without contributing to the presynaptic mechanisms that control the time course of release.


Assuntos
Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Junção Neuromuscular/fisiologia , Transmissão Sináptica/fisiologia , Animais , Astacoidea , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Cricetinae , Estimulação Elétrica , Extremidades/inervação , Microinjeções , Proteínas Sensíveis a N-Etilmaleimida/administração & dosagem , Proteínas Sensíveis a N-Etilmaleimida/genética , Proteínas Sensíveis a N-Etilmaleimida/farmacologia , Junção Neuromuscular/efeitos dos fármacos , Ratos , Proteínas Recombinantes , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Caminhada
19.
Curr Biol ; 16(7): 636-48, 2006 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-16581508

RESUMO

BACKGROUND: Myelinated axons are essential for rapid conduction of action potentials in the vertebrate nervous system. Of particular importance are the nodes of Ranvier, sites of voltage-gated sodium channel clustering that allow action potentials to be propagated along myelinated axons by saltatory conduction. Despite their critical role in the function of myelinated axons, little is known about the mechanisms that organize the nodes of Ranvier. RESULTS: Starting with a forward genetic screen in zebrafish, we have identified an essential requirement for nsf (N-ethylmaleimide sensitive factor) in the organization of myelinated axons. Previous work has shown that NSF is essential for membrane fusion in eukaryotes and has a critical role in vesicle fusion at chemical synapses. Zebrafish nsf mutants are paralyzed and have impaired response to light, reflecting disrupted nsf function in synaptic transmission and neural activity. In addition, nsf mutants exhibit defects in Myelin basic protein expression and in localization of sodium channel proteins at nodes of Ranvier. Analysis of chimeric larvae indicates that nsf functions autonomously in neurons, such that sodium channel clusters are evident in wild-type neurons transplanted into the nsf mutant hosts. Through pharmacological analyses, we show that neural activity and function of chemical synapses are not required for sodium channel clustering and myelination in the larval nervous system. CONCLUSIONS: Zebrafish nsf mutants provide a novel vertebrate system to investigate Nsf function in vivo. Our results reveal a previously unknown role for nsf, independent of its function in synaptic vesicle fusion, in the formation of the nodes of Ranvier in the vertebrate nervous system.


Assuntos
Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Nós Neurofibrosos/ultraestrutura , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/metabolismo , Potenciais de Ação/fisiologia , Animais , Morte Celular/fisiologia , Quimera/metabolismo , Marcadores Genéticos , Células Ciliadas Auditivas/fisiologia , Larva/anatomia & histologia , Larva/genética , Larva/metabolismo , Movimento/fisiologia , Mutação , Proteína Básica da Mielina/genética , Proteínas Sensíveis a N-Etilmaleimida/genética , Fenótipo , RNA Mensageiro/metabolismo , Nós Neurofibrosos/metabolismo , Canais de Sódio/fisiologia , Transmissão Sináptica/fisiologia , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética
20.
J Biol Chem ; 281(15): 9852-8, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16461345

RESUMO

Pctaire1, a member of the cyclin-dependent kinase (Cdk)-related family, has recently been shown to be phosphorylated and regulated by Cdk5/p35. Although Pctaire1 is expressed in both neuronal and non-neuronal cells, its precise functions remain elusive. We performed a yeast two-hybrid screen to identify proteins that interact with Pctaire1. N-Ethylmaleimide-sensitive fusion protein (NSF), a crucial factor in vesicular transport and membrane fusion, was identified as one of the Pctaire1 interacting proteins. We demonstrate that the D2 domain of NSF, which is required for the oligomerization of NSF subunits, binds directly to and is phosphorylated by Pctaire1 on serine 569. Mutation of this phosphorylation site on NSF (S569A) augments its ability to oligomerize. Moreover, inhibition of Pctaire1 activity by transfecting its kinase-dead (KD) mutant into COS-7 cells enhances the self-association of NSF. Interestingly, Pctaire1 associates with NSF and synaptic vesicle-associated proteins in adult rat brain. To investigate whether Pctaire1 phosphorylation of NSF is involved in regulation of Ca(2+)-dependent exocytosis, we examined the effect of expressing Pctaire1 or NSF phosphorylation mutants on the regulated secretion of growth hormone from PC12 cells. Interestingly, expression of either Pctaire1-KD or NSF-S569A in PC12 cells significantly increases high K(+)-stimulated growth hormone release. Taken together, our findings provide the first demonstration that Pctaire1 phosphorylation of NSF regulates the ability of NSF to oligomerize, implicating an unexpected role of this kinase in modulating exocytosis. These findings open a new avenue of research in studying the functional roles of Pctaire1 in the nervous system.


Assuntos
Quinases Ciclina-Dependentes/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Western Blotting , Células COS , Cálcio/metabolismo , Sistema Nervoso Central/metabolismo , Chlorocebus aethiops , Clonagem Molecular , Quinases Ciclina-Dependentes/química , DNA Complementar/metabolismo , Dimerização , Relação Dose-Resposta a Droga , Exocitose , Humanos , Imunoprecipitação , Proteínas Sensíveis a N-Etilmaleimida/química , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Células PC12 , Fosforilação , Plasmídeos/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/química , Estrutura Terciária de Proteína , Ratos , Serina/química , Transfecção , Técnicas do Sistema de Duplo-Híbrido , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...