Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-36835643

RESUMO

SNARE proteins reside between opposing membranes and facilitate vesicle fusion, a physiological process ubiquitously required for secretion, endocytosis and autophagy. With age, neurosecretory SNARE activity drops and is pertinent to age-associated neurological disorders. Despite the importance of SNARE complex assembly and disassembly in membrane fusion, their diverse localization hinders the complete understanding of their function. Here, we revealed a subset of SNARE proteins, the syntaxin SYX-17, the synaptobrevins VAMP-7, SNB-6 and the tethering factor USO-1, to be either localized or in close proximity to mitochondria, in vivo. We term them mitoSNAREs and show that animals deficient in mitoSNAREs exhibit increased mitochondria mass and accumulation of autophagosomes. The SNARE disassembly factor NSF-1 seems to be required for the effects of mitoSNARE depletion. Moreover, we find mitoSNAREs to be indispensable for normal aging in both neuronal and non-neuronal tissues. Overall, we uncover a previously unrecognized subset of SNAREs that localize to mitochondria and propose a role of mitoSNARE assembly and disassembly factors in basal autophagy regulation and aging.


Assuntos
Envelhecimento , Autofagia , Caenorhabditis elegans , Proteínas SNARE , Animais , Caenorhabditis elegans/fisiologia , Endocitose , Fusão de Membrana , Proteínas SNARE/fisiologia
2.
Sci Rep ; 11(1): 10955, 2021 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-34040104

RESUMO

The primary hallmark of Parkinson's disease (PD) is the generation of Lewy bodies of which major component is α-synuclein (α-Syn). Because of increasing evidence of the fundamental roles of α-Syn oligomers in disease progression, α-Syn oligomers have become potential targets for therapeutic interventions for PD. One of the potential toxicities of α-Syn oligomers is their inhibition of SNARE-mediated vesicle fusion by specifically interacting with vesicle-SNARE protein synaptobrevin-2 (Syb2), which hampers dopamine release. Here, we show that α-Syn monomers and oligomers cooperatively inhibit neuronal SNARE-mediated vesicle fusion. α-Syn monomers at submicromolar concentrations increase the fusion inhibition by α-Syn oligomers. This cooperative pathological effect stems from the synergically enhanced vesicle clustering. Based on this cooperative inhibition mechanism, we reverse the fusion inhibitory effect of α-Syn oligomers using small peptide fragments. The small peptide fragments, derivatives of α-Syn, block the binding of α-Syn oligomers to Syb2 and dramatically reverse the toxicity of α-Syn oligomers in vesicle fusion. Our findings demonstrate a new strategy for therapeutic intervention in PD and related diseases based on this specific interaction of α-Syn.


Assuntos
Fusão de Membrana/efeitos dos fármacos , Proteínas SNARE/antagonistas & inibidores , alfa-Sinucleína/farmacologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Dopamina/metabolismo , Dopamina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Lipossomos , Lipídeos de Membrana/metabolismo , Modelos Moleculares , Mutação de Sentido Incorreto , Fragmentos de Peptídeos/farmacologia , Mutação Puntual , Ligação Proteica , Multimerização Proteica , Proteolipídeos/química , Proteínas Recombinantes de Fusão/farmacologia , Proteínas SNARE/fisiologia , Proteína 2 Associada à Membrana da Vesícula/antagonistas & inibidores , Proteína 2 Associada à Membrana da Vesícula/fisiologia , alfa-Sinucleína/química , alfa-Sinucleína/genética , alfa-Sinucleína/toxicidade
3.
J Comp Neurol ; 529(5): 1066-1080, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32783205

RESUMO

BMAL1 is a core component of the mammalian circadian clockwork. Removal of BMAL1 from the retina significantly affects visual information processing in both rod and cone pathways. To identify potential pathways and/or molecules through which BMAL1 alters signal transmission at the cone pedicle, we performed an RNA-seq differential expression analysis between cone-specific Bmal1 knockout cones (cone-Bmal1-/- ) and wild-type (WT) cones. We found 88 genes differentially expressed. Among these, Complexin3 (Cplx3), a SNARE regulator at ribbon synapses, was downregulated fivefold in the mutant cones. The purpose of this work was to determine whether BMAL1 and/or the cone clock controls CPLX3 protein expression at cone pedicles. We found that CPLX3 expression level was decreased twofold in cone-Bmal1-/- cones. Furthermore, CPLX3 expression was downregulated at night compared to the day in WT cones but remained constitutively low in mutant cones both day and night. The transcript and protein expression levels of Cplx4-the other complexin expressed in cones-were similar in WT and mutant cones; in WT cones, CPLX4 protein level did not change with the time of day. In silico analysis revealed four potential BMAL1:CLOCK binding sites upstream from exon one of Cplx3 and none upstream of exon one of Cplx4. Our results suggest that CPLX3 expression is regulated at the transcriptional level by the cone clock. The modulation of CPLX3 may be a mechanism by which the clock controls the cone synaptic transfer function to second-order cells and thereby impacts retinal signal processing during the day/night cycle.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Relógios Circadianos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Proteínas SNARE/fisiologia , Fatores de Transcrição ARNTL/deficiência , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/biossíntese , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Regulação para Baixo , Feminino , Masculino , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA-Seq , Pigmentos da Retina/genética , Rodopsinas Sensoriais/genética , Transdução de Sinais/fisiologia
4.
Plant Cell Physiol ; 61(10): 1750-1759, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32706881

RESUMO

SNAREs (soluble N-ethyl maleimide-sensitive factor attachment protein receptors) mediate membrane fusion of vesicle transport in eukaryotic cells. LjSYP132s are the members of Qa-SNAREs in Lotus japonicus. Two isoforms, LjSYP132a and LjSYP132b, are generated by alternative splicing. Immunoblot analysis detected strong expression of LjSYP132s in infected root nodules and seeds by posttranscriptional modification. In either LjSYP132a or LjSYP132b silenced roots (RNAi-LjSYP132a, RNAi-LjSYP132b), the infection thread (IT) was not elongated, suggesting that both LjSYP132a and LjSYP132b have a role in IT progression. The results were consistent with the data of qRT-PCR showing that both genes were expressed at the early stage of infection. However, during the nodulation, only LjSYP132a was induced. LjSYP132s protein was observed in the Mesorhizobium loti-inoculated roots of mutants, nfr1, castor and pollux, suggesting that LjSYP132s can be induced without Nod factor signaling. Accumulation of LjSYP132s in the peribacteroid membrane suggests the function of not only IT formation but also nutrient transport. In contrast, qRT-PCR showed that LjSYP132b was expressed in the seeds. A stable transgenic plant of LjSYP132b, R132b, was produced by RNAi silencing. In the R132b plants, small pods with a few seeds and abnormal tip growth of the pollen tubes were observed, suggesting that LjSYP132b has a role in pollen tube growth and nutrient transport in the plasma membrane of seeds.


Assuntos
Lotus/crescimento & desenvolvimento , Proteínas de Plantas/fisiologia , Nodulação , Proteínas SNARE/fisiologia , Sementes/crescimento & desenvolvimento , Processamento Alternativo , Regulação da Expressão Gênica de Plantas , Lotus/metabolismo , Proteínas de Plantas/metabolismo , Raízes de Plantas/metabolismo , Plantas Geneticamente Modificadas , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Interferência de RNA , Proteínas SNARE/metabolismo , Sementes/metabolismo
5.
Neuron ; 107(1): 22-37, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32559416

RESUMO

Neuronal SNAREs and their key regulators together drive synaptic vesicle exocytosis and synaptic transmission as a single integrated membrane fusion machine. Human pathogenic mutations have now been reported for all eight core components, but patients are diagnosed with very different neurodevelopmental syndromes. We propose to unify these syndromes, based on etiology and mechanism, as "SNAREopathies." Here, we review the strikingly diverse clinical phenomenology and disease severity and the also remarkably diverse genetic mechanisms. We argue that disease severity generally scales with functional redundancy and, conversely, that the large effect of mutations in some SNARE genes is the price paid for extensive integration and exceptional specialization. Finally, we discuss how subtle differences in components being rate limiting in different types of neurons helps to explain the main symptoms.


Assuntos
Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/fisiopatologia , Proteínas SNARE/fisiologia , Humanos , Mutação
6.
Brain ; 143(6): 1780-1797, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32428221

RESUMO

Transportation of key proteins via extracellular vesicles has been recently implicated in various neurodegenerative disorders, including Parkinson's disease, as a new mechanism of disease spreading and a new source of biomarkers. Extracellular vesicles likely to be derived from the brain can be isolated from peripheral blood and have been reported to contain higher levels of α-synuclein (α-syn) in Parkinson's disease patients. However, very little is known about extracellular vesicles in multiple system atrophy, a disease that, like Parkinson's disease, involves pathological α-syn aggregation, though the process is centred around oligodendrocytes in multiple system atrophy. In this study, a novel immunocapture technology was developed to isolate blood CNPase-positive, oligodendrocyte-derived enriched microvesicles (OEMVs), followed by fluorescent nanoparticle tracking analysis and assessment of α-syn levels contained within the OEMVs. The results demonstrated that the concentrations of OEMVs were significantly lower in multiple system atrophy patients, compared to Parkinson's disease patients and healthy control subjects. It is also noted that the population of OEMVs involved was mainly in the size range closer to that of exosomes, and that the average α-syn concentrations (per vesicle) contained in these OEMVs were not significantly different among the three groups. The phenomenon of reduced OEMVs was again observed in a transgenic mouse model of multiple system atrophy and in primary oligodendrocyte cultures, and the mechanism involved was likely related, at least in part, to an α-syn-mediated interference in the interaction between syntaxin 4 and VAMP2, leading to the dysfunction of the SNARE complex. These results suggest that reduced OEMVs could be an important mechanism related to pathological α-syn aggregation in oligodendrocytes, and the OEMVs found in peripheral blood could be further explored for their potential as multiple system atrophy biomarkers.


Assuntos
Atrofia de Múltiplos Sistemas/fisiopatologia , Oligodendroglia/metabolismo , Proteínas SNARE/metabolismo , Idoso , Animais , Secreções Corporais/metabolismo , Encéfalo/patologia , Micropartículas Derivadas de Células/imunologia , Micropartículas Derivadas de Células/metabolismo , Modelos Animais de Doenças , Exossomos/metabolismo , Exossomos/fisiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Neurônios/metabolismo , Doença de Parkinson/patologia , Proteínas SNARE/fisiologia , alfa-Sinucleína/metabolismo
7.
Elife ; 92020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32391794

RESUMO

Vesicle fusion is mediated by assembly of SNARE proteins between opposing membranes. While previous work suggested an active role of SNARE transmembrane domains (TMDs) in promoting membrane merger (Dhara et al., 2016), the underlying mechanism remained elusive. Here, we show that naturally-occurring v-SNARE TMD variants differentially regulate fusion pore dynamics in mouse chromaffin cells, indicating TMD flexibility as a mechanistic determinant that facilitates transmitter release from differentially-sized vesicles. Membrane curvature-promoting phospholipids like lysophosphatidylcholine or oleic acid profoundly alter pore expansion and fully rescue the decelerated fusion kinetics of TMD-rigidifying VAMP2 mutants. Thus, v-SNARE TMDs and phospholipids cooperate in supporting membrane curvature at the fusion pore neck. Oppositely, slowing of pore kinetics by the SNARE-regulator complexin-2 withstands the curvature-driven speeding of fusion, indicating that pore evolution is tightly coupled to progressive SNARE complex formation. Collectively, TMD-mediated support of membrane curvature and SNARE force-generated membrane bending promote fusion pore formation and expansion.


Assuntos
Exocitose , Fusão de Membrana , Complexos Multiproteicos/fisiologia , Neurotransmissores/fisiologia , Fosfolipídeos/metabolismo , Proteínas SNARE/fisiologia , Proteína 2 Associada à Membrana da Vesícula/fisiologia , Animais , Cálcio/fisiologia , Membrana Celular/metabolismo , Células Cultivadas , Células Cromafins , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mutantes/fisiologia , Ligação Proteica , Domínios Proteicos , Vesículas Secretórias/fisiologia
8.
J Neurosci ; 40(15): 3052-3062, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32132265

RESUMO

Maintenance of cardiorespiratory homeostasis depends on autonomic reflexes controlled by neuronal circuits of the brainstem. The neurophysiology and neuroanatomy of these reflex pathways are well understood, however, the mechanisms and functional significance of autonomic circuit modulation by glial cells remain largely unknown. In the experiments conducted in male laboratory rats we show that astrocytes of the nucleus of the solitary tract (NTS), the brain area that receives and integrates sensory information from the heart and blood vessels, respond to incoming afferent inputs with [Ca2+]i elevations. Astroglial [Ca2+]i responses are triggered by transmitters released by vagal afferents, glutamate acting at AMPA receptors and 5-HT acting at 5-HT2A receptors. In conscious freely behaving animals blockade of Ca2+-dependent vesicular release mechanisms in NTS astrocytes by virally driven expression of a dominant-negative SNARE protein (dnSNARE) increased baroreflex sensitivity by 70% (p < 0.001). This effect of compromised astroglial function was specific to the NTS as expression of dnSNARE in astrocytes of the ventrolateral brainstem had no effect. ATP is considered the principle gliotransmitter and is released by vesicular mechanisms blocked by dnSNARE expression. Consistent with this hypothesis, in anesthetized rats, pharmacological activation of P2Y1 purinoceptors in the NTS decreased baroreflex gain by 40% (p = 0.031), whereas blockade of P2Y1 receptors increased baroreflex gain by 57% (p = 0.018). These results suggest that glutamate and 5-HT, released by NTS afferent terminals, trigger Ca2+-dependent astroglial release of ATP to modulate baroreflex sensitivity via P2Y1 receptors. These data add to the growing body of evidence supporting an active role of astrocytes in brain information processing.SIGNIFICANCE STATEMENT Cardiorespiratory reflexes maintain autonomic balance and ensure cardiovascular health. Impaired baroreflex may contribute to the development of cardiovascular disease and serves as a robust predictor of cardiovascular and all-cause mortality. The data obtained in this study suggest that astrocytes are integral components of the brainstem mechanisms that process afferent information and modulate baroreflex sensitivity via the release of ATP. Any condition associated with higher levels of "ambient" ATP in the NTS would be expected to decrease baroreflex gain by the mechanism described here. As ATP is the primary signaling molecule of glial cells (astrocytes, microglia), responding to metabolic stress and inflammatory stimuli, our study suggests a plausible mechanism of how the central component of the baroreflex is affected in pathological conditions.


Assuntos
Astrócitos/fisiologia , Barorreflexo/fisiologia , Núcleo Solitário/fisiologia , Trifosfato de Adenosina/fisiologia , Animais , Sinalização do Cálcio/fisiologia , Masculino , Neurônios Aferentes/metabolismo , Neurotransmissores/metabolismo , Neurotransmissores/fisiologia , Agonistas do Receptor Purinérgico P2Y/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor 5-HT2A de Serotonina/efeitos dos fármacos , Receptores de AMPA/efeitos dos fármacos , Receptores Purinérgicos P2Y1/efeitos dos fármacos , Proteínas SNARE/fisiologia , Serotonina/farmacologia , Estimulação do Nervo Vago
9.
Dialogues Clin Neurosci ; 21(3): 271-279, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31749651

RESUMO

Prospective, community-based studies allow evaluation of associations between cognitive functioning and synaptic measures, controlled for age-related pathologies. Findings from >400 community-based participants are reviewed. Levels of two presynaptic proteins, complexin-I (inhibitory terminals), and complexin-II (excitatory terminals) contributed to cognitive variation from normal to dementia. Adding the amount of protein-protein interaction between two others, synaptosome-associated protein-25 and syntaxin, explained 6% of overall variance. The presynaptic protein Munc18-1 long variant was localized to inhibitory terminals, and like complexin-I, was positively associated with cognition. Associations depended on Braak stage, with the level of complexin-I contributing nearly 15% to cognitive variation in stages 0-II, while complexin-II contributed 7% in stages V-VI. Non-denaturing gels identified multiple soluble N-ethylmaleimide-sensitive factor attachment protein receptor protein-protein (SNARE) complexes in frontal and in temporal lobes, making specific contributions to cognitive functions. Multiple mechanisms of presynaptic plasticity contribute to cognitive function during aging.
.


Los estudios prospectivos realizados en la comunidad permiten evaluar las asociaciones entre el funcionamiento cognitivo y las mediciones sinápticas, controladas por patologías relacionadas con la edad. Se revisan los hallazgos de más de 400 participantes de la comunidad. Los niveles de dos proteínas presinápticas, complexina-I (terminales inhibitorios) y complexina-II (terminales excitatorios) contribuyeron a la variación cognitiva entre la situación normal y la demencia. Al agregar la interacción proteína-proteína entre la proteína 25 asociada al sinaptosoma y la sintaxina, se explicó el 6% de la varianza general. La variante larga de la proteína presináptica Munc 18-1 se localizó en terminales inhibitorios y, al igual que la complexina I, se asoció positivamente con la cognición. Las asociaciones dependían de la etapa de Braak, siendo el nivel de complexina-I responsable de casi el 15% de la variación cognitiva para las etapas 0-II, mientras que la complexina-II contribuyó con el 7% en las etapas V-VI. Los geles no desnaturalizantes identificaron múltiples complejos de proteína-proteína del receptor de proteína de unión al factor sensible a N-etilmaleimida soluble (SNARE) en los lóbulos frontales y temporales, los cuales contribuyen de manera específica a las funciones cognitivas. Durante el envejecimiento participan múltiples mecanismos de plasticidad presináptica en la función cognitiva.


Des études prospectives communautaires permettent d'évaluer des associations entre le fonctionnement cognitif et des mesures synaptiques, contrôlées pour des pathologies liées à l'âge. Des résultats issus de plus de 400 participants communautaires sont analysés. Les taux de complexine-I (terminaisons inhibitrices) et de complexine-II (terminaisons excitatrices), deux protéines présynaptiques, contribuent aux variations cognitives de la normalité à la démence. S'y ajoute l'interaction protéine-protéine entre la protéine SNAP25 (protéine 25 associée au synaptosome) et la syntaxine, expliquant 6 % de la variance totale. Le variant long de la protéine pré-synaptique Munc18-1 est localisé sur les terminaisons inhibitrices et associé positivement à la cognition, comme la complexine-I. Les associations dépendent du stade Braak, le taux de complexine-I étant responsable de presque 15 % de la variation cognitive pour les stades 0-II alors que la complexine-II contribue pour 7 % aux stades V-VI. Des gels non dénaturants identifient des complexes multiples protéine-protéine SNARE (Soluble N-ethylmaleimide-sensitive factor Attachment protein REceptor) dans les lobes frontaux et temporaux, contribuant spécifiquement aux fonctions cognitives. Au cours du vieillissement, de nombreux mécanismes de plasticité présynaptique participent à la fonction cognitive.


Assuntos
Cognição/fisiologia , Disfunção Cognitiva/patologia , Proteínas SNARE/fisiologia , Sinapses/patologia , Sinapses/fisiologia , Envelhecimento/patologia , Envelhecimento/psicologia , Animais , Disfunção Cognitiva/psicologia , Humanos , Estudos Prospectivos
10.
Nat Commun ; 10(1): 4326, 2019 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-31548544

RESUMO

Munc18-1 and Munc13-1 orchestrate assembly of the SNARE complex formed by syntaxin-1, SNAP-25 and synaptobrevin, allowing exquisite regulation of neurotransmitter release. Non-regulated neurotransmitter release might be prevented by αSNAP, which inhibits exocytosis and SNARE-dependent liposome fusion. However, distinct mechanisms of inhibition by αSNAP were suggested, and it is unknown how such inhibition is overcome. Using liposome fusion assays, FRET and NMR spectroscopy, here we provide a comprehensive view of the mechanisms underlying the inhibitory functions of αSNAP, showing that αSNAP potently inhibits liposome fusion by: binding to syntaxin-1, hindering Munc18-1 binding; binding to syntaxin-1-SNAP-25 heterodimers, precluding SNARE complex formation; and binding to trans-SNARE complexes, preventing fusion. Importantly, inhibition by αSNAP is avoided only when Munc18-1 binds first to syntaxin-1, leading to Munc18-1-Munc13-1-dependent liposome fusion. We propose that at least some of the inhibitory activities of αSNAP ensure that neurotransmitter release occurs through the highly-regulated Munc18-1-Munc13-1 pathway at the active zone.


Assuntos
Proteínas Munc18/fisiologia , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/fisiologia , Vesículas Sinápticas/metabolismo , Animais , Bovinos , Cricetulus , Escherichia coli/genética , Fusão de Membrana , Proteínas Munc18/química , Proteínas Munc18/metabolismo , Conformação Proteica , Ratos , Proteínas SNARE/metabolismo , Proteínas SNARE/fisiologia , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/genética , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo , Sintaxina 1/química , Sintaxina 1/metabolismo
12.
Cells ; 8(4)2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30974782

RESUMO

Protein synthesis begins at free ribosomes or ribosomes attached with the endoplasmic reticulum (ER). Newly synthesized proteins are transported to the plasma membrane for secretion through conventional or unconventional pathways. In conventional protein secretion, proteins are transported from the ER lumen to Golgi lumen and through various other compartments to be secreted at the plasma membrane, while unconventional protein secretion bypasses the Golgi apparatus. Soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) proteins are involved in cargo vesicle trafficking and membrane fusion. The ER localized vesicle associated SNARE (v-SNARE) protein Sec22 plays a major role during anterograde and retrograde transport by promoting efficient membrane fusion and assisting in the assembly of higher order complexes by homodimer formation. Sec22 is not only confined to ER-Golgi intermediate compartments (ERGIC) but also facilitates formation of contact sites between ER and plasma membranes. Sec22 mutation is responsible for the development of atherosclerosis and symptoms in the brain in Alzheimer's disease and aging in humans. In the fruit fly Drosophila melanogaster, Sec22 is essential for photoreceptor morphogenesis, the wingless signaling pathway, and normal ER, Golgi, and endosome morphology. In the plant Arabidopsis thaliana, it is involved in development, and in the nematode Caenorhabditis elegans, it is in involved in the RNA interference (RNAi) pathway. In filamentous fungi, it affects cell wall integrity, growth, reproduction, pathogenicity, regulation of reactive oxygen species (ROS), expression of extracellular enzymes, and transcriptional regulation of many development related genes. This review provides a detailed account of Sec22 function, summarizes its domain structure, discusses its genetic redundancy with Ykt6, discusses what is known about its localization to discrete membranes, its contributions in conventional and unconventional autophagy, and a variety of other roles across different cellular systems ranging from higher to lower eukaryotes, and highlights some of the surprises that have originated from research on Sec22.


Assuntos
Proteínas SNARE/classificação , Proteínas SNARE/fisiologia , Animais , Autofagia/fisiologia , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Fungos/metabolismo , Complexo de Golgi/metabolismo , Humanos , Fusão de Membrana/fisiologia , Plantas/metabolismo , Transporte Proteico/fisiologia
13.
Sci Rep ; 9(1): 1211, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718891

RESUMO

Homozygous mutations in SNAP29, encoding a SNARE protein mainly involved in membrane fusion, cause CEDNIK (Cerebral Dysgenesis, Neuropathy, Ichthyosis and Keratoderma), a rare congenital neurocutaneous syndrome associated with short life expectancy, whose pathogenesis is unclear. Here, we report the analysis of the first genetic model of CEDNIK in zebrafish. Strikingly, homozygous snap29 mutant larvae display CEDNIK-like features, such as microcephaly and skin defects. Consistent with Snap29 role in membrane fusion during autophagy, we observe accumulation of the autophagy markers p62 and LC3, and formation of aberrant multilamellar organelles and mitochondria. Importantly, we find high levels of apoptotic cell death during early development that might play a yet uncharacterized role in CEDNIK pathogenesis. Mutant larvae also display mouth opening problems, feeding impairment and swimming difficulties. These alterations correlate with defective trigeminal nerve formation and excess axonal branching. Since the paralog Snap25 is known to promote axonal branching, Snap29 might act in opposition with, or modulate Snap25 activity during neurodevelopment. Our vertebrate genetic model of CEDNIK extends the description in vivo of the multisystem defects due to loss of Snap29 and could provide the base to test compounds that might ameliorate traits of the disease.


Assuntos
Ceratodermia Palmar e Plantar/metabolismo , Síndromes Neurocutâneas/metabolismo , Proteínas SNARE/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Autofagia , Humanos , Ceratodermia Palmar e Plantar/genética , Ceratodermia Palmar e Plantar/fisiopatologia , Fusão de Membrana , Modelos Genéticos , Mutação , Malformações do Sistema Nervoso/metabolismo , Síndromes Neurocutâneas/genética , Síndromes Neurocutâneas/fisiopatologia , Fenótipo , Ligação Proteica , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteínas SNARE/fisiologia , Proteína 25 Associada a Sinaptossoma/metabolismo , Proteína 25 Associada a Sinaptossoma/fisiologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia
14.
Proc Natl Acad Sci U S A ; 116(7): 2435-2442, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30700546

RESUMO

SNARE proteins zipper to form complexes (SNAREpins) that power vesicle fusion with target membranes in a variety of biological processes. A single SNAREpin takes about 1 s to fuse two bilayers, yet a handful can ensure release of neurotransmitters from synaptic vesicles much faster: in a 10th of a millisecond. We propose that, similar to the case of muscle myosins, the ultrafast fusion results from cooperative action of many SNAREpins. The coupling originates from mechanical interactions induced by confining scaffolds. Each SNAREpin is known to have enough energy to overcome the fusion barrier of 25-[Formula: see text]; however, the fusion barrier only becomes relevant when the SNAREpins are nearly completely zippered, and from this state, each SNAREpin can deliver only a small fraction of this energy as mechanical work. Therefore, they have to act cooperatively, and we show that at least three of them are needed to ensure fusion in less than a millisecond. However, to reach the prefusion state collectively, starting from the experimentally observed half-zippered metastable state, the SNAREpins have to mechanically synchronize, which takes more time as the number of SNAREpins increases. Incorporating this somewhat counterintuitive idea in a simple coarse-grained model results in the prediction that there should be an optimum number of SNAREpins for submillisecond fusion: three to six over a wide range of parameters. Interestingly, in situ cryoelectron microscope tomography has very recently shown that exactly six SNAREpins participate in the fusion of each synaptic vesicle. This number is in the range predicted by our theory.


Assuntos
Proteínas SNARE/fisiologia , Animais , Microscopia Crioeletrônica , Fusão de Membrana , Modelos Biológicos , Ligação Proteica , Proteínas SNARE/metabolismo
15.
Nat Struct Mol Biol ; 25(10): 911-917, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30291360

RESUMO

The regulated exocytotic release of neurotransmitter and hormones is accomplished by a complex protein machinery whose core consists of SNARE proteins and the calcium sensor synaptotagmin-1. We propose a mechanism in which the lipid membrane is intimately involved in coupling calcium sensing to release. We found that fusion of dense core vesicles, derived from rat PC12 cells, was strongly linked to the angle between the cytoplasmic domain of the SNARE complex and the plane of the target membrane. We propose that, as this tilt angle increases, force is exerted on the SNARE transmembrane domains to drive the merger of the two bilayers. The tilt angle markedly increased following calcium-mediated binding of synaptotagmin to membranes, strongly depended on the surface electrostatics of the membrane, and was strictly coupled to the lipid order of the target membrane.


Assuntos
Exocitose , Modelos Moleculares , Sinaptotagminas/fisiologia , Vesículas Transportadoras/química , Animais , Sinalização do Cálcio , Metabolismo dos Lipídeos/fisiologia , Células PC12 , Domínios Proteicos , Proteínas Qa-SNARE/química , Proteínas Qa-SNARE/metabolismo , Proteínas Qa-SNARE/fisiologia , Ratos , Proteínas SNARE/química , Proteínas SNARE/metabolismo , Proteínas SNARE/fisiologia , Sinaptotagminas/química , Sinaptotagminas/metabolismo , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/fisiologia
16.
J Neurosci ; 38(32): 7179-7191, 2018 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-30012692

RESUMO

The soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins synaptobrevin (Syb), syntaxin, and SNAP-25 function in Ca2+-triggered exocytosis in both endocrine cells and neurons. The transmembrane domains (TMDs) of Syb and syntaxin span the vesicle and plasma membrane, respectively, and influence flux through fusion pores in endocrine cells as well as fusion pores formed during SNARE-mediated fusion of reconstituted membranes. These results support a model for exocytosis in which SNARE TMDs form the initial fusion pore. The present study sought to test this model in synaptic terminals. Patch-clamp recordings of miniature EPSCs (mEPSCs) were used to probe fusion pore properties in cultured hippocampal neurons from mice of both sexes. Mutants harboring tryptophan at four different sites in the Syb TMD reduced the rate-of-rise of mEPSCs. A computer model that simulates glutamate diffusion and receptor activation kinetics could account for this reduction in mEPSC rise rate by slowing the flux of glutamate through synaptic fusion pores. TMD mutations introducing positive charge also reduced the mEPSC rise rate, but negatively charged residues and glycine, which should have done the opposite, had no effect. The sensitivity of mEPSCs to pharmacological blockade of receptor desensitization was enhanced by a mutation that slowed the mEPSC rate-of-rise, suggesting that the mutation prolonged the residence of glutamate in the synaptic cleft. The same four Syb TMD residues found here to influence synaptic release were found previously to influence endocrine release, leading us to propose that a similar TMD-lined fusion pore functions widely in Ca2+-triggered exocytosis in mammalian cells.SIGNIFICANCE STATEMENT SNARE proteins function broadly in biological membrane fusion. Evidence from non-neuronal systems suggests that SNARE proteins initiate fusion by forming a fusion pore lined by transmembrane domains, but this model has not yet been tested in synapses. The present study addressed this question by testing mutations in the synaptic vesicle SNARE synaptobrevin for an influence on the rise rate of miniature synaptic currents. These results indicate that synaptobrevin's transmembrane domain interacts with glutamate as it passes through the fusion pore. The sites in synaptobrevin that influence this flux are identical to those shown previously to influence flux through endocrine fusion pores. Thus, SNARE transmembrane domains may function in the fusion pores of Ca2+-triggered exocytosis of both neurotransmitters and hormones.


Assuntos
Exocitose/fisiologia , Ácido Glutâmico/metabolismo , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Neurônios/fisiologia , Proteína 2 Associada à Membrana da Vesícula/fisiologia , Substituição de Aminoácidos , Animais , Transporte Biológico , Cálcio/fisiologia , Simulação por Computador , Difusão , Feminino , Técnicas de Inativação de Genes , Hipocampo/citologia , Cinética , Masculino , Fusão de Membrana , Camundongos , Modelos Biológicos , Técnicas de Patch-Clamp , Domínios Proteicos , Proteínas SNARE/fisiologia , Triptofano/análise , Proteína 2 Associada à Membrana da Vesícula/química , Proteína 2 Associada à Membrana da Vesícula/genética
17.
Cell Calcium ; 73: 53-54, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29880197

RESUMO

The focus of this special issue (SI) ¼Membrane Merger in Conventional and Unconventional Vesicle Secretion« is regulated exocytosis, a universally conserved mechanism, consisting of a merger between the vesicle and the plasma membranes. Although this process evolved with eukaryotic organisms some three billion years ago (Spang et al., 2015), the understanding of physiology and patobiology of this process, especially at elementary vesicle level, remains unclear. Exocytotic fusion consists of several stages, starting by vesicle delivery to the plasma membrane, initially establishing a very narrow and stable fusion pore, that can reversibly open and close several times before it can fully widen. This allows vesicle cargo to be completely discharged from the vesicle lumen and permits vesicle-membrane resident proteins including channels, transporters, receptors and other signalling molecules, to be incorporated into the plasma membrane. The contributions in this SI bring new insights on the complexity of vesicle-based secretion, including discussion that vesicle anatomy appears to modulate exocytotic fusion pore properties and that the soluble N-ethylmaleimide-sensitive-factor attachment protein receptor proteins (SNARE-proteins), not only facilitate pre- and post-fusion stages of exocytosis, but also serve in vesicle navigation within the cytoplasm.


Assuntos
Membrana Celular/fisiologia , Exocitose/fisiologia , Fusão de Membrana/fisiologia , Proteínas SNARE/fisiologia , Animais , Humanos , Saccharomyces cerevisiae/metabolismo , Vesículas Secretórias/fisiologia
18.
Plant Cell Environ ; 41(11): 2668-2677, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29940699

RESUMO

Vesicle traffic is tightly coordinated with ion transport for plant cell expansion through physical interactions between subsets of vesicle-trafficking (so-called SNARE) proteins and plasma membrane Kv channels, including the archetypal inward-rectifying K+ channel, KAT1 of Arabidopsis. Ion channels open and close rapidly over milliseconds, whereas vesicle fusion events require many seconds. Binding has been mapped to conserved motifs of both the Kv channels and the SNAREs, but knowledge of the temporal kinetics of their interactions, especially as it might relate to channel gating and its coordination with vesicle fusion remains unclear. Here, we report that the SNARE SYP121 promotes KAT1 gating through a persistent interaction that alters the stability of the channel, both in its open and closed states. We show, too, that SYP121 action on the channel open state requires SNARE anchoring in the plasma membrane. Our findings indicate that SNARE binding confers a conformational bias that encompasses the microscopic kinetics of channel gating, with leverage applied through the SNARE anchor in favour of the open channel.


Assuntos
Proteínas de Arabidopsis/fisiologia , Arabidopsis/metabolismo , Ativação do Canal Iônico , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Potássio/metabolismo , Proteínas Qa-SNARE/fisiologia , Arabidopsis/fisiologia , Proteínas de Arabidopsis/metabolismo , Transporte Biológico , Ativação do Canal Iônico/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas SNARE/metabolismo , Proteínas SNARE/fisiologia
19.
PLoS Pathog ; 14(5): e1007028, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29746582

RESUMO

Positive-strand RNA viruses assemble numerous membrane-bound viral replicase complexes within large replication compartments to support their replication in infected cells. Yet the detailed mechanism of how given subcellular compartments are subverted by viruses is incompletely understood. Although, Tomato bushy stunt virus (TBSV) uses peroxisomal membranes for replication, in this paper, we show evidence that the ER-resident SNARE (soluble NSF attachment protein receptor) proteins play critical roles in the formation of active replicase complexes in yeast model host and in plants. Depletion of the syntaxin 18-like Ufe1 and Use1, which are components of the ER SNARE complex in the ERAS (ER arrival site) subdomain, in yeast resulted in greatly reduced tombusvirus accumulation. Over-expression of a dominant-negative mutant of either the yeast Ufe1 or the orthologous plant Syp81 syntaxin greatly interferes with tombusvirus replication in yeast and plants, thus further supporting the role of this host protein in tombusvirus replication. Moreover, tombusvirus RNA replication was low in cell-free extracts from yeast with repressed Ufe1 or Use1 expression. We also present evidence for the mislocalization of the tombusviral p33 replication protein to the ER membrane in Ufe1p-depleted yeast cells. The viral p33 replication protein interacts with both Ufe1p and Use1p and co-opts them into the TBSV replication compartment in yeast and plant cells. The co-opted Ufe1 affects the virus-driven membrane contact site formation, sterol-enrichment at replication sites, recruitment of several pro-viral host factors and subversion of the Rab5-positive PE-rich endosomes needed for robust TBSV replication. In summary, we demonstrate a critical role for Ufe1 and Use1 SNARE proteins in TBSV replication and propose that the pro-viral functions of Ufe1 and Use1 are to serve as assembly hubs for the formation of the extensive TBSV replication compartments in cells. Altogether, these findings point clearly at the ERAS subdomain of ER as a critical site for the biogenesis of the TBSV replication compartment.


Assuntos
Proteínas SNARE/metabolismo , Proteínas SNARE/fisiologia , Tombusvirus/fisiologia , Replicação do DNA , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/fisiologia , Endossomos/metabolismo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Membranas Mitocondriais/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas Qa-SNARE/fisiologia , Proteínas Qc-SNARE/metabolismo , Proteínas Qc-SNARE/fisiologia , RNA Viral/genética , RNA Polimerase Dependente de RNA/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Tombusvirus/genética , Tombusvirus/metabolismo , Tombusvirus/patogenicidade , Proteínas Virais/genética , Replicação Viral/fisiologia
20.
Curr Biol ; 28(8): R397-R401, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29689222

RESUMO

A fundamental hallmark of eukaryotic cells is their compartmentalization into functionally distinct organelles, including those of the secretory and endocytic pathways. Transport of cargo between these compartments and to/from the cell surface is mediated by membrane-bound vesicles and tubules. Delivery of cargo is facilitated by SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor)-mediated membrane fusion of vesicles with their target compartments. Vesicles contain a variety of cargos, including lipids, membrane proteins, signaling molecules, biosynthetic and hydrolytic enzymes, and the trafficking machinery itself. Proper function of membrane trafficking is required for cellular growth, division, movement, and cell-cell communication. Defects in these processes have been implicated in a variety of human diseases, such as cancer, diabetes, neurodegenerative disorders, ciliopathies, and infections. The elucidation of the mechanisms of SNARE assembly and disassembly is key to understanding how membrane fusion is regulated throughout eukaryotes. Here, we introduce the SNARE proteins, their structures and functions in eukaryotic cells, and discuss recent breakthroughs in elucidating the regulation of SNARE assembly and disassembly through the use of high-resolution structural biology and biophysical techniques.


Assuntos
Fusão de Membrana/fisiologia , Proteínas SNARE/biossíntese , Proteínas SNARE/metabolismo , Animais , Transporte Biológico/fisiologia , Membrana Celular/metabolismo , Humanos , Ligação Proteica , Transporte Proteico/fisiologia , Proteínas SNARE/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...