Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Biol Cell ; 32(20): ar7, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34347508

RESUMEN

Polarized morphogenesis is achieved by targeting or inhibiting growth in distinct regions. Rod-shaped fission yeast cells grow exclusively at their ends by restricting exocytosis and secretion to these sites. This growth pattern implies the existence of mechanisms that prevent exocytosis and growth along nongrowing cell sides. We previously identified a set of 50-100 megadalton-sized node structures along the sides of fission yeast cells that contained the interacting proteins Skb1 and Slf1. Here, we show that Skb1-Slf1 nodes contain the syntaxin-like soluble N-ethylmaleimide-sensitive factor attachment protein receptor Psy1, which mediates exocytosis in fission yeast. Psy1 localizes in a diffuse pattern at cell tips, where it likely promotes exocytosis and growth, but is sequestered in Skb1-Slf1 nodes at cell sides where growth does not occur. Mutations that prevent node assembly or inhibit Psy1 localization to nodes lead to aberrant exocytosis at cell sides and increased cell width. Genetic results indicate that this Psy1 node mechanism acts in parallel to actin cables and Cdc42 regulation. Our work suggests that sequestration of syntaxin-like Psy1 at nongrowing regions of the cell cortex reinforces cell morphology by restricting exocytosis to proper sites of polarized growth.


Asunto(s)
Metiltransferasas/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Actinas/metabolismo , Ciclo Celular/fisiología , Membrana Celular/metabolismo , Polaridad Celular/fisiología , Exocitosis , Fusión de Membrana , Morfogénesis , Transporte de Proteínas , Proteínas Qa-SNARE/fisiología , Proteínas SNARE/metabolismo , Schizosaccharomyces/citología , Proteínas de Schizosaccharomyces pombe/fisiología
2.
J Pharmacol Sci ; 145(4): 297-307, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33712280

RESUMEN

Herein, we investigated the functional association of the serotonin transporter (SERT) with syntaxin-3 (STX3). We first overexpressed SERT and STX3 in various cells and examined their interaction, localization, and functional association. Immunoprecipitation studies revealed that STX3 interacted with SERT when expressed in COS-7 cells. Immunocytochemical studies revealed that SERT and STX3 were colocalized in the endoplasmic reticulum (ER) and Golgi apparatus. STX3 overexpression significantly reduced the uptake activity of SERT by attenuating its plasma membrane expression, suggesting that overexpressed STX3 anchors SERT in the ER and Golgi apparatus. STX3 knockdown did not affect the uptake activity of SERT but altered its glycosylation state. To elucidate the association of STX3 with SERT under physiological conditions, rather than overexpressing cells, we investigated this interaction in polarized Caco-2 cells, which endogenously express both proteins. Immunocytochemical studies revealed that SERT and STX3 were localized in microvilli-like structures at the apical plasma membrane. STX3 knockdown marginally but significantly decreased the serotonin uptake activity of Caco-2 cells, suggesting that STX3 positively regulates SERT function in Caco-2 cells, as opposed to SERT regulation by STX3 in overexpressing cells. Collectively, STX3 may colocalize with SERT during SERT membrane trafficking and regulate SERT function in an STX3-expressing lesion-dependent manner.


Asunto(s)
Epistasis Genética/genética , Expresión Génica/genética , Proteínas Qa-SNARE/metabolismo , Proteínas Qa-SNARE/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Glicosilación , Aparato de Golgi/metabolismo , Microvellosidades/metabolismo , Proteínas Qa-SNARE/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
3.
Sci Rep ; 10(1): 709, 2020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31959797

RESUMEN

Recent evidence suggests that SNARE fusion machinery play critical roles in postsynaptic neurotransmitter receptor trafficking, which is essential for synaptic plasticity. However, the key SNAREs involved remain highly controversial; syntaxin-3 and syntaxin-4 are leading candidates for the syntaxin isoform underlying postsynaptic plasticity. In a previous study, we showed that pyramidal-neuron specific conditional knockout (cKO) of syntaxin-4 significantly reduces basal transmission, synaptic plasticity and impairs postsynaptic receptor trafficking. However, this does not exclude a role for syntaxin-3 in such processes. Here, we generated and analyzed syntaxin-3 cKO mice. Extracellular field recordings in hippocampal slices showed that syntaxin-3 cKO did not exhibit significant changes in CA1 basal neurotransmission or in paired-pulse ratios. Importantly, there were no observed differences during LTP in comparison to control mice. Syntaxin-3 cKO mice performed similarly as the controls in spatial and contextual learning tasks. Consistent with the minimal effects of syntaxin-3 cKO, syntaxin-3 mRNA level was very low in hippocampal and cortex pyramidal neurons, but strongly expressed in the corpus callosum and caudate axon fibers. Together, our data suggest that syntaxin-3 is dispensable for hippocampal basal neurotransmission and synaptic plasticity, and further supports the notion that syntaxin-4 is the major isoform mediating these processes.


Asunto(s)
Región CA1 Hipocampal/fisiología , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Proteínas Qa-SNARE/fisiología , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología , Animales , Región CA1 Hipocampal/metabolismo , Cuerpo Calloso/metabolismo , Expresión Génica , Técnicas In Vitro , Potenciación a Largo Plazo/fisiología , Ratones Noqueados , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , ARN Mensajero/metabolismo
4.
PLoS Pathog ; 15(7): e1007982, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31356625

RESUMEN

To colonize phagocytes, Leishmania subverts microbicidal processes through components of its surface coat that include lipophosphoglycan and the GP63 metalloprotease. How these virulence glycoconjugates are shed, exit the parasitophorous vacuole (PV), and traffic within host cells is poorly understood. Here, we show that lipophosphoglycan and GP63 are released from the parasite surface following phagocytosis and redistribute to the endoplasmic reticulum (ER) of macrophages. Pharmacological disruption of the trafficking between the ER and the Golgi hindered the exit of these molecules from the PV and dampened the cleavage of host proteins by GP63. Silencing by RNA interference of the soluble N-ethylmaleimide-sensitive-factor attachment protein receptors Sec22b and syntaxin-5, which regulate ER-Golgi trafficking, identified these host proteins as components of the machinery that mediates the spreading of Leishmania effectors within host cells. Our findings unveil a mechanism whereby a vacuolar pathogen takes advantage of the host cell's secretory pathway to promote egress of virulence factors beyond the PV.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Leishmania/fisiología , Leishmania/patogenicidad , Proteínas Protozoarias/fisiología , Factores de Virulencia/fisiología , Animales , Retículo Endoplásmico/parasitología , Femenino , Glicoesfingolípidos/fisiología , Humanos , Leishmania/crecimiento & desarrollo , Leishmaniasis/parasitología , Metaloendopeptidasas/fisiología , Ratones , Ratones Endogámicos C57BL , Fagocitos/parasitología , Fagocitosis , Fagosomas/parasitología , Proteínas Qa-SNARE/fisiología , Proteínas R-SNARE/fisiología , Vías Secretoras , Vacuolas/parasitología , Virulencia
5.
Nat Struct Mol Biol ; 25(10): 911-917, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30291360

RESUMEN

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.


Asunto(s)
Exocitosis , Modelos Moleculares , Sinaptotagminas/fisiología , Vesículas Transportadoras/química , Animales , Señalización del Calcio , Metabolismo de los Lípidos/fisiología , Células PC12 , Dominios Proteicos , Proteínas Qa-SNARE/química , Proteínas Qa-SNARE/metabolismo , Proteínas Qa-SNARE/fisiología , Ratas , Proteínas SNARE/química , Proteínas SNARE/metabolismo , Proteínas SNARE/fisiología , Sinaptotagminas/química , Sinaptotagminas/metabolismo , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/fisiología
6.
Plant Cell Environ ; 41(11): 2668-2677, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29940699

RESUMEN

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.


Asunto(s)
Proteínas de Arabidopsis/fisiología , Arabidopsis/metabolismo , Activación del Canal Iónico , Canales de Potasio de Rectificación Interna/fisiología , Potasio/metabolismo , Proteínas Qa-SNARE/fisiología , Arabidopsis/fisiología , Proteínas de Arabidopsis/metabolismo , Transporte Biológico , Activación del Canal Iónico/fisiología , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas SNARE/metabolismo , Proteínas SNARE/fisiología
7.
PLoS Genet ; 14(6): e1007432, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29912942

RESUMEN

Axonal growth and guidance rely on correct growth cone responses to guidance cues. Unlike the signaling cascades that link axonal growth to cytoskeletal dynamics, little is known about the crosstalk mechanisms between guidance and membrane dynamics and turnover. Recent studies indicate that whereas axonal attraction requires exocytosis, chemorepulsion relies on endocytosis. Indeed, our own studies have shown that Netrin-1/Deleted in Colorectal Cancer (DCC) signaling triggers exocytosis through the SNARE Syntaxin-1 (STX1). However, limited in vivo evidence is available about the role of SNARE proteins in axonal guidance. To address this issue, here we systematically deleted SNARE genes in three species. We show that loss-of-function of STX1 results in pre- and post-commissural axonal guidance defects in the midline of fly, chick, and mouse embryos. Inactivation of VAMP2, Ti-VAMP, and SNAP25 led to additional abnormalities in axonal guidance. We also confirmed that STX1 loss-of-function results in reduced sensitivity of commissural axons to Slit-2 and Netrin-1. Finally, genetic interaction studies in Drosophila show that STX1 interacts with both the Netrin-1/DCC and Robo/Slit pathways. Our data provide evidence of an evolutionarily conserved role of STX1 and SNARE proteins in midline axonal guidance in vivo, by regulating both pre- and post-commissural guidance mechanisms.


Asunto(s)
Neurogénesis/genética , Sintaxina 1/genética , Sintaxina 1/fisiología , Animales , Axones/metabolismo , Quimiotaxis/genética , Embrión de Pollo , Drosophila/genética , Proteínas de Drosophila/genética , Exocitosis/genética , Regulación del Desarrollo de la Expresión Génica/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Ratones , Ratones Noqueados , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/embriología , Netrina-1/genética , Netrina-1/metabolismo , Neurogénesis/fisiología , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/fisiología , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Transducción de Señal/genética , Médula Espinal/embriología , Médula Espinal/metabolismo
8.
PLoS Pathog ; 14(5): e1007028, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29746582

RESUMEN

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.


Asunto(s)
Proteínas SNARE/metabolismo , Proteínas SNARE/fisiología , Tombusvirus/fisiología , Replicación del ADN , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/fisiología , Endosomas/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Membranas Mitocondriales/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas Qa-SNARE/fisiología , Proteínas Qc-SNARE/metabolismo , Proteínas Qc-SNARE/fisiología , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Tombusvirus/genética , Tombusvirus/metabolismo , Tombusvirus/patogenicidad , Proteínas Virales/genética , Replicación Viral/fisiología
9.
J Cell Biol ; 217(8): 2633-2645, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29789439

RESUMEN

Macroautophagy is an evolutionarily conserved catabolic mechanism that delivers intracellular constituents to lysosomes using autophagosomes. To achieve degradation, lysosomes must fuse with closed autophagosomes. We previously reported that the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein syntaxin (STX) 17 translocates to autophagosomes to mediate fusion with lysosomes. In this study, we report an additional mechanism. We found that autophagosome-lysosome fusion is retained to some extent even in STX17 knockout (KO) HeLa cells. By screening other human SNAREs, we identified YKT6 as a novel autophagosomal SNARE protein. Depletion of YKT6 inhibited autophagosome-lysosome fusion partially in wild-type and completely in STX17 KO cells, suggesting that YKT6 and STX17 are independently required for fusion. YKT6 formed a SNARE complex with SNAP29 and lysosomal STX7, both of which are required for autophagosomal fusion. Recruitment of YKT6 to autophagosomes depends on its N-terminal longin domain but not on the C-terminal palmitoylation and farnesylation that are essential for its Golgi localization. These findings suggest that two independent SNARE complexes mediate autophagosome-lysosome fusion.


Asunto(s)
Autofagosomas/fisiología , Lisosomas/fisiología , Proteínas Qa-SNARE/fisiología , Proteínas R-SNARE/fisiología , Animales , Autofagosomas/metabolismo , Línea Celular , Técnicas de Inactivación de Genes , Células HEK293 , Células HeLa , Humanos , Lisosomas/metabolismo , Ratones , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteínas R-SNARE/genética , Proteínas R-SNARE/metabolismo
10.
PLoS Genet ; 14(4): e1007359, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29694367

RESUMEN

The autophagosomal SNARE Syntaxin17 (Syx17) forms a complex with Snap29 and Vamp7/8 to promote autophagosome-lysosome fusion via multiple interactions with the tethering complex HOPS. Here we demonstrate that, unexpectedly, one more SNARE (Ykt6) is also required for autophagosome clearance in Drosophila. We find that loss of Ykt6 leads to large-scale accumulation of autophagosomes that are unable to fuse with lysosomes to form autolysosomes. Of note, loss of Syx5, the partner of Ykt6 in ER-Golgi trafficking does not prevent autolysosome formation, pointing to a more direct role of Ykt6 in fusion. Indeed, Ykt6 localizes to lysosomes and autolysosomes, and forms a SNARE complex with Syx17 and Snap29. Interestingly, Ykt6 can be outcompeted from this SNARE complex by Vamp7, and we demonstrate that overexpression of Vamp7 rescues the fusion defect of ykt6 loss of function cells. Finally, a point mutant form with an RQ amino acid change in the zero ionic layer of Ykt6 protein that is thought to be important for fusion-competent SNARE complex assembly retains normal autophagic activity and restores full viability in mutant animals, unlike palmitoylation or farnesylation site mutant Ykt6 forms. As Ykt6 and Vamp7 are both required for autophagosome-lysosome fusion and are mutually exclusive subunits in a Syx17-Snap29 complex, these data suggest that Vamp7 is directly involved in membrane fusion and Ykt6 acts as a non-conventional, regulatory SNARE in this process.


Asunto(s)
Autofagosomas/fisiología , Proteínas de Drosophila/fisiología , Lisosomas/fisiología , Fusión de Membrana/fisiología , Proteínas R-SNARE/fisiología , Animales , Animales Modificados Genéticamente , Sitios de Unión , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Fusión de Membrana/genética , Modelos Biológicos , Complejos Multiproteicos/genética , Complejos Multiproteicos/fisiología , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/fisiología , Proteínas R-SNARE/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas SNARE/genética , Proteínas SNARE/fisiología
11.
Proc Natl Acad Sci U S A ; 113(50): E8031-E8040, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27911771

RESUMEN

Synaptic soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) couple their stepwise folding to fusion of synaptic vesicles with plasma membranes. In this process, three SNAREs assemble into a stable four-helix bundle. Arguably, the first and rate-limiting step of SNARE assembly is the formation of an activated binary target (t)-SNARE complex on the target plasma membrane, which then zippers with the vesicle (v)-SNARE on the vesicle to drive membrane fusion. However, the t-SNARE complex readily misfolds, and its structure, stability, and dynamics are elusive. Using single-molecule force spectroscopy, we modeled the synaptic t-SNARE complex as a parallel three-helix bundle with a small frayed C terminus. The helical bundle sequentially folded in an N-terminal domain (NTD) and a C-terminal domain (CTD) separated by a central ionic layer, with total unfolding energy of ∼17 kBT, where kB is the Boltzmann constant and T is 300 K. Peptide binding to the CTD activated the t-SNARE complex to initiate NTD zippering with the v-SNARE, a mechanism likely shared by the mammalian uncoordinated-18-1 protein (Munc18-1). The NTD zippering then dramatically stabilized the CTD, facilitating further SNARE zippering. The subtle bidirectional t-SNARE conformational switch was mediated by the ionic layer. Thus, the t-SNARE complex acted as a switch to enable fast and controlled SNARE zippering required for synaptic vesicle fusion and neurotransmission.


Asunto(s)
Proteínas SNARE/química , Secuencia de Aminoácidos , Animales , Fusión de Membrana , Ratones , Microscopía de Fuerza Atómica , Simulación de Dinámica Molecular , Proteínas Munc18/química , Proteínas Munc18/fisiología , Pinzas Ópticas , Conformación Proteica , Dominios Proteicos , Pliegue de Proteína , Estabilidad Proteica , Proteínas Qa-SNARE/química , Proteínas Qa-SNARE/fisiología , Proteínas SNARE/genética , Proteínas SNARE/fisiología , Transmisión Sináptica/fisiología , Proteína 25 Asociada a Sinaptosomas/química , Proteína 25 Asociada a Sinaptosomas/fisiología , Proteína 2 de Membrana Asociada a Vesículas/química , Proteína 2 de Membrana Asociada a Vesículas/fisiología
12.
Curr Psychiatry Rep ; 18(8): 77, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27371030

RESUMEN

Schizophrenia is a serious psychiatric illness which is experienced by about 1 % of individuals worldwide and has a debilitating impact on perception, cognition, and social function. Over the years, several models/hypotheses have been developed which link schizophrenia to dysregulations of the dopamine, glutamate, and serotonin receptor pathways. An important segment of these pathways that have been extensively studied for the pathophysiology of schizophrenia is the presynaptic neurotransmitter release mechanism. This set of molecular events is an evolutionarily well-conserved process that involves vesicle recruitment, docking, membrane fusion, and recycling, leading to efficient neurotransmitter delivery at the synapse. Accumulated evidence indicate dysregulation of this mechanism impacting postsynaptic signal transduction via different neurotransmitters in key brain regions implicated in schizophrenia. In recent years, after ground-breaking work that elucidated the operations of this mechanism, research efforts have focused on the alterations in the messenger RNA (mRNA) and protein expression of presynaptic neurotransmitter release molecules in schizophrenia and other neuropsychiatric conditions. In this review article, we present recent evidence from schizophrenia human postmortem studies that key proteins involved in the presynaptic release mechanism are dysregulated in the disorder. We also discuss the potential impact of dysfunctional presynaptic neurotransmitter release on the various neurotransmitter systems implicated in schizophrenia.


Asunto(s)
Encéfalo/fisiopatología , Esquizofrenia/fisiopatología , Psicología del Esquizofrénico , Vesículas Sinápticas/fisiología , Animales , Encéfalo/patología , Humanos , Proteínas Munc18/fisiología , Neurotransmisores/metabolismo , Proteínas Qa-SNARE/fisiología , Proteínas R-SNARE/fisiología , ARN Mensajero/genética , Receptores Presinapticos/fisiología , Proteínas SNARE/fisiología , Esquizofrenia/patología , Transducción de Señal/fisiología , Sinapsinas/fisiología , Vesículas Sinápticas/genética , Sinaptofisina/fisiología , Proteína 25 Asociada a Sinaptosomas/fisiología
13.
PLoS One ; 11(3): e0151658, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26986836

RESUMEN

The SNARE complex composed of VAMP727, SYP22, VTI11 and SYP51 is critical for protein trafficking and PSV biogenesis in Arabidopsis. This SNARE complex directs the fusion between the prevacuolar compartment (PVC) and the vacuole, and thus mediates protein trafficking to the vacuole. In this study, we examined the role of AtNHX5 and AtNHX6 in regulating this SNARE complex and its function in protein trafficking. We found that AtNHX5 and AtNHX6 were required for seed production, protein trafficking and PSV biogenesis. We further found that the nhx5 nhx6 syp22 triple mutant showed severe defects in seedling growth and seed development. The triple mutant had short siliques and reduced seed sets, but larger seeds. In addition, the triple mutant had numerous smaller protein storage vacuoles (PSVs) and accumulated precursors of the seed storage proteins in seeds. The PVC localization of SYP22 and VAMP727 was repressed in nhx5 nhx6, while a significant amount of SYP22 and VAMP727 was trapped in the Golgi or TGN in nhx5 nhx6. AtNHX5 and AtNHX6 were co-localized with SYP22 and VAMP727. Three conserved acidic residues, D164, E188, and D193 in AtNHX5 and D165, E189, and D194 in AtNHX6, were essential for the transport of the storage proteins, indicating the importance of exchange activity in protein transport. AtNHX5 or AtNHX6 did not interact physically with the SNARE complex. Taken together, AtNHX5 and AtNHX6 are required for the PVC localization of the SNARE complex and hence its function in protein transport. AtNHX5 and AtNHX6 may regulate the subcellular localization of the SNARE complex by their transport activity.


Asunto(s)
Proteínas de Arabidopsis/fisiología , Arabidopsis/metabolismo , Transporte de Proteínas/fisiología , Proteínas SNARE/fisiología , Semillas/metabolismo , Proteínas Qa-SNARE/fisiología , Proteínas Qb-SNARE/fisiología , Fracciones Subcelulares/fisiología , Vacuolas/fisiología
14.
Blood ; 127(17): 2113-21, 2016 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-26864340

RESUMEN

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory disease. Inherited forms of HLH are caused by biallelic mutations in several effectors of granule-dependent lymphocyte-mediated cytotoxicity. A small proportion of patients with a so-called "secondary" form of HLH, which develops in the aftermath of infection, autoimmunity, or cancer, carry a monoallelic mutation in one or more HLH-associated genes. Although this observation suggests that HLH may have a polygenic mode of inheritance, the latter is very difficult to prove in humans. In order to determine whether the accumulation of partial genetic defects in lymphocyte-mediated cytotoxicity can contribute to the development of HLH, we generated mice that were doubly or triply heterozygous for mutations in HLH-associated genes, those coding for perforin, Rab27a, and syntaxin-11. We found that the accumulation of monoallelic mutations did indeed increase the risk of developing HLH immunopathology after lymphocytic choriomeningitis virus infection. In mechanistic terms, the accumulation of heterozygous mutations in the two degranulation genes Rab27a and syntaxin-11, impaired the dynamics and secretion of cytotoxic granules at the immune synapse of T lymphocytes. In addition, the accumulation of heterozygous mutations within the three genes impaired natural killer lymphocyte cytotoxicity in vivo. The genetic defects can be ranked in terms of the severity of the resulting HLH manifestations. Our results form the basis of a polygenic model of the occurrence of secondary HLH.


Asunto(s)
Linfohistiocitosis Hemofagocítica/genética , Herencia Multifactorial , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Qa-SNARE/genética , Proteínas de Unión al GTP rab/genética , Animales , Degranulación de la Célula/genética , Cruzamientos Genéticos , Citotoxicidad Inmunológica/genética , Dosificación de Gen , Predisposición Genética a la Enfermedad , Heterocigoto , Sinapsis Inmunológicas/inmunología , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Mutación , Proteínas Citotóxicas Formadoras de Poros/fisiología , Proteínas Qa-SNARE/fisiología , Organismos Libres de Patógenos Específicos , Linfocitos T Citotóxicos/inmunología , Proteínas de Unión al GTP rab/fisiología , Proteínas rab27 de Unión a GTP
15.
Cancer Sci ; 106(10): 1455-62, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26176172

RESUMEN

Peripheral T-cell lymphomas (PTCL) are a heterogeneous group of non-Hodgkin lymphomas with poor prognosis. Their molecular pathogenesis has not been entirely elucidated. We previously showed that 6q24 is one of the most frequently deleted regions in primary thyroid T-cell lymphoma. In this study, we extended the analysis to other subtypes of PTCL and performed functional assays to identify the causative genes of PTCL that are located on 6q24. Genomic loss of 6q24 was observed in 14 of 232 (6%) PTCL cases. The genomic loss regions identified at 6q24 always involved only two known genes, STX11 and UTRN. The expression of STX11, but not UTRN, was substantially lower in PTCL than in normal T-cells. STX11 sequence analysis revealed mutations in two cases (one clinical sample and one T-cell line). We further analyzed the function of STX11 in 14 cell lines belonging to different lineages. STX11 expression only suppressed the proliferation of T-cell lines bearing genomic alterations at the STX11 locus. Interestingly, expression of a novel STX11 mutant (p.Arg78Cys) did not exert suppressive effects on the induced cell lines, suggesting that this mutant is a loss-of-function mutation. In addition, STX11-altered PTCL not otherwise specified cases were characterized by the presence of hemophagocytic syndrome (67% vs 8%, P = 0.04). They also tended to have a poor prognosis compared with those without STX11 alteration. These results suggest that STX11 plays an important role in the pathogenesis of PTCL and they may contribute to the future development of new drugs for the treatment of PTCL.


Asunto(s)
Linfoma de Células T Periférico/metabolismo , Proteínas Qa-SNARE/fisiología , Eliminación de Secuencia/genética , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Cromosomas Humanos Par 6/genética , Hibridación Genómica Comparativa , Genes Supresores de Tumor , Predisposición Genética a la Enfermedad , Células HEK293 , Haploinsuficiencia/genética , Células HeLa , Humanos , Células Jurkat , Linfoma de Células T Periférico/patología , Células MCF-7 , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Proteínas Qa-SNARE/genética , Linfocitos T/patología , Utrofina/genética
16.
Anesthesiology ; 122(5): 1060-74, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25738637

RESUMEN

BACKGROUND: Recent evidence suggests that general anesthetics activate endogenous sleep pathways, yet this mechanism cannot explain the entirety of general anesthesia. General anesthetics could disrupt synaptic release processes, as previous work in Caenorhabditis elegans and in vitro cell preparations suggested a role for the soluble NSF attachment protein receptor protein, syntaxin1A, in mediating resistance to several general anesthetics. The authors questioned whether the syntaxin1A-mediated effects found in these reductionist systems reflected a common anesthetic mechanism distinct from sleep-related processes. METHODS: Using the fruit fly model, Drosophila melanogaster, the authors investigated the relevance of syntaxin1A manipulations to general anesthesia. The authors used different behavioral and electrophysiological endpoints to test the effect of syntaxin1A mutations on sensitivity to isoflurane. RESULTS: The authors found two syntaxin1A mutations that confer opposite general anesthesia phenotypes: syxH3-C, a 14-amino acid deletion mutant, is resistant to isoflurane (n = 40 flies), and syxKARRAA, a strain with two amino acid substitutions, is hypersensitive to the drug (n = 40 flies). Crucially, these opposing effects are maintained across different behavioral endpoints and life stages. The authors determined the isoflurane sensitivity of syxH3-C at the larval neuromuscular junction to assess effects on synaptic release. The authors find that although isoflurane slightly attenuates synaptic release in wild-type animals (n = 8), syxH3-C preserves synaptic release in the presence of isoflurane (n = 8). CONCLUSION: The study results are evidence that volatile general anesthetics target synaptic release mechanisms; in addition to first activating sleep pathways, a major consequence of these drugs may be to decrease the efficacy of neurotransmission.


Asunto(s)
Anestésicos por Inhalación/farmacología , Proteínas de Drosophila/fisiología , Resistencia a Medicamentos/genética , Hipersensibilidad/genética , Isoflurano/farmacología , Proteínas Qa-SNARE/fisiología , Animales , Conducta Animal/efectos de los fármacos , Proteínas de Drosophila/genética , Drosophila melanogaster , Larva , Locomoción/efectos de los fármacos , Mutación , Unión Neuromuscular/efectos de los fármacos , Neurotransmisores/metabolismo , Proteínas Qa-SNARE/genética , Reflejo de Sobresalto , Sueño/efectos de los fármacos
17.
J Biol Chem ; 290(3): 1536-45, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25404741

RESUMEN

Platelet secretion not only drives thrombosis and hemostasis, but also mediates a variety of other physiological and pathological processes. The ubiquitous SNARE machinery and a number of accessory proteins have been implicated in regulating secretion in platelet. Although several platelet SNAREs have been identified, further members of the SNARE family may be needed to fine-tune platelet secretion. In this study we identified expression of the t-SNARE syntaxin 8 (STX8) (Qc SNARE) in mouse and human platelets. In mouse studies, whereas STX8 was not essential for α-granule or lysosome secretion, Stx8(-/-) platelets showed a significant defect in dense granule secretion in response to thrombin and CRP. This was most pronounced at intermediate concentrations of agonists. They also showed an aggregation defect that could be rescued with exogenous ADP and increased embolization in Stx8(-/-) mice in vivo consistent with an important autocrine and paracrine role for ADP in aggregation and thrombus stabilization. STX8 therefore specifically contributes to dense granule secretion and represents another member of a growing family of genes that play distinct roles in regulating granule release from platelets and thus platelet function in thrombosis and hemostasis.


Asunto(s)
Plaquetas/metabolismo , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/fisiología , Trombosis/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Digitonina/química , Exocitosis , Citometría de Flujo , Hemostasis , Humanos , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Activación Plaquetaria , Proteínas SNARE/metabolismo , Vesículas Secretoras/metabolismo
18.
Enzyme Microb Technol ; 68: 33-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25435503

RESUMEN

Limitations in protein production and secretion have been attributed to the inefficient folding rate of overexpressed proteins and the cellular response to the presence of overexpressed proteins in the endoplasmic reticulum (ER). In this study, we improved the yield of glucose oxidase (GOD) by manipulating genes involved in protein folding machinery and abnormal folding stress responses. First, genes with folding and secretion functions were used to modulate the folding rate of GOD in the ER and its secretion level in the cytoplasm. Next, the potential benefits of the ERAD elements were determined. Cellular resistance to ER derived stress was then strengthened by overexpressing the stress response gene GCN4. Furthermore, a module combination strategy, which co-expressed the SEC53, CNE1 and GCN4 genes, was employed to construct the Pichia pastoris strain S17. This increased the yield of GOD to 21.81g/L, with an activity of 1972.9U/mL, which were 2.53- and 5.11-fold higher, respectively, than the control strain. The work described here improved GOD production significantly, and the strategies employed in this study provide novel information for the large-scale production of heterologous proteins.


Asunto(s)
Aspergillus niger/enzimología , Proteínas Fúngicas/biosíntesis , Glucosa Oxidasa/química , Pichia/metabolismo , Ingeniería de Proteínas , Aspergillus niger/genética , Electroporación , Degradación Asociada con el Retículo Endoplásmico/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Genes Fúngicos , Glucosa Oxidasa/biosíntesis , Glucosa Oxidasa/genética , Glucosa Oxidasa/metabolismo , Pichia/clasificación , Pichia/genética , Plásmidos/genética , Pliegue de Proteína , Transporte de Proteínas , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/fisiología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Estrés Fisiológico/genética , Transformación Genética
19.
J Cell Sci ; 127(Pt 8): 1712-25, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24496451

RESUMEN

Acquisition of an invasive phenotype is prerequisite for tumor metastasis. Degradation of the extracellular matrix (ECM), and subsequent invasion by tumor cells, is mediated, in part, through subcellular structures called invadopodia. Src-dependent cytoskeletal rearrangements are required to form invadopodia, and here we identify an association between Src, epidermal growth factor receptor (EGFR), and ß1 integrin that facilitates invadopodia formation. The association of Src, EGFR and ß1 integrin is dependent upon membrane traffic that is mediated by syntaxin13 (officially known as STX12) and SNAP23; a similar dependence on these two SNARE proteins was observed for invadopodium-based matrix degradation and cell invasion. Inhibition of SNARE function impaired the delivery of Src and EGFR to developing invadopodia, as well as the ß1-integrin-dependent activation of Src and phosphorylation of EGFR on Tyr residue 845. We also identified an association between SNAP23 and ß1 integrin, and inhibition of ß1 integrin increased this association, whereas the interaction between syntaxin13 and SNAP23 was reduced. The results suggest that SNARE-dependent trafficking is regulated, in part, by ß1 integrin and is required for the delivery of Src and EGFR to sites of invadopodia formation in order to support tumor cell invasion.


Asunto(s)
Receptores ErbB/metabolismo , Integrina beta1/metabolismo , Proteínas Qa-SNARE/fisiología , Proteínas Qb-SNARE/fisiología , Proteínas Qc-SNARE/fisiología , Familia-src Quinasas/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular , Matriz Extracelular/metabolismo , Humanos , Invasividad Neoplásica , Fosforilación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Seudópodos/metabolismo
20.
Plant Mol Biol ; 85(1-2): 107-21, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24452833

RESUMEN

A Glycine max syntaxin 31 homolog (Gm-SYP38) was identified as being expressed in nematode-induced feeding structures known as syncytia undergoing an incompatible interaction with the plant parasitic nematode Heterodera glycines. The observed Gm-SYP38 expression was consistent with prior gene expression analyses that identified the alpha soluble NSF attachment protein (Gm-α-SNAP) resistance gene because homologs of these genes physically interact and function together in other genetic systems. Syntaxin 31 is a protein that resides on the cis face of the Golgi apparatus and binds α-SNAP-like proteins, but has no known role in resistance. Experiments presented here show Gm-α-SNAP overexpression induces Gm-SYP38 transcription. Overexpression of Gm-SYP38 rescues G. max [Williams 82/PI 518671], genetically rhg1 (-/-), by suppressing H. glycines parasitism. In contrast, Gm-SYP38 RNAi in the rhg1 (+/+) genotype G. max [Peking/PI 548402] increases susceptibility. Gm-α-SNAP and Gm-SYP38 overexpression induce the transcriptional activity of the cytoplasmic receptor-like kinase BOTRYTIS INDUCED KINASE 1 (Gm-BIK1-6) which is a family of defense proteins known to anchor to membranes through a 5' MGXXXS/T(R) N-myristoylation sequence. Gm-BIK1-6 had been identified previously by RNA-seq experiments as expressed in syncytia undergoing an incompatible reaction. Gm-BIK1-6 overexpression rescues the resistant phenotype. In contrast, Gm-BIK1-6 RNAi increases parasitism. The analysis demonstrates a role for syntaxin 31-like genes in resistance that until now was not known.


Asunto(s)
Genes de Plantas , Glycine max/parasitología , Interacciones Huésped-Parásitos , Nematodos/patogenicidad , Proteínas Qa-SNARE/fisiología , Animales , Clonación Molecular , Proteínas Qa-SNARE/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Glycine max/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...