Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Neurosci ; 38(9): 2207-2225, 2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29311141

RESUMO

mTORC1-dependent translational control plays a key role in several enduring forms of synaptic plasticity such as long term potentiation (LTP) and mGluR-dependent long term depression. Recent evidence demonstrates an additional role in regulating synaptic homeostasis in response to inactivity, where dendritic mTORC1 serves to modulate presynaptic function via retrograde signaling. Presently, it is unclear whether LTP and homeostatic plasticity use a common route to mTORC1-dependent signaling or whether each engage mTORC1 through distinct pathways. Here, we report a unique signaling pathway that specifically couples homeostatic signaling to postsynaptic mTORC1 after loss of excitatory synaptic input. We find that AMPAR blockade, but not LTP-inducing stimulation, induces phospholipase D (PLD)-dependent synthesis of the lipid second messenger phosphatidic acid (PA) in rat cultured hippocampal neurons of either sex. Pharmacological blockade of PLD1/2 or pharmacogenetic disruption of PA interactions with mTOR eliminates mTORC1 signaling and presynaptic compensation driven by AMPAR blockade, but does not alter mTORC1 activation or functional changes during chemical LTP (cLTP). Overexpression of PLD1, but not PLD2, recapitulates both functional synaptic changes as well as signature cellular adaptations associated with homeostatic plasticity. Finally, transient application of exogenous PA is sufficient to drive rapid presynaptic compensation requiring mTORC1-dependent translation of BDNF in the postsynaptic compartment. These results thus define a unique homeostatic signaling pathway coupling mTORC1 activation to changes in excitatory synaptic drive. Our results further imply that more than one canonical mTORC1 activation pathway may be relevant for the design of novel therapeutic approaches against neurodevelopmental disorders associated with mTORC1 dysregulation.SIGNIFICANCE STATEMENT Homeostatic and Hebbian forms of synaptic plasticity are thought to play complementary roles in regulating neural circuit function, but we know little about how these forms of plasticity are distinguished at the single neuron level. Here, we define a signaling pathway that uniquely links mTORC1 with homeostatic signaling in neurons.


Assuntos
Homeostase/fisiologia , Potenciação de Longa Duração/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Animais , Feminino , Hipocampo/metabolismo , Masculino , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley
2.
J Biol Chem ; 293(7): 2232-2246, 2018 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-29269412

RESUMO

Altering the expression of Tomosyn-1 (Tomo-1), a soluble, R-SNARE domain-containing protein, significantly affects behavior in mice, Drosophila, and Caenorhabditis elegans Yet, the mechanisms that modulate Tomo-1 expression and its regulatory activity remain poorly defined. Here, we found that Tomo-1 expression levels influence postsynaptic spine density. Tomo-1 overexpression increased dendritic spine density, whereas Tomo-1 knockdown (KD) decreased spine density. These findings identified a novel action of Tomo-1 on dendritic spines, which is unique because it occurs independently of Tomo-1's C-terminal R-SNARE domain. We also demonstrated that the ubiquitin-proteasome system (UPS), which is known to influence synaptic strength, dynamically regulates Tomo-1 protein levels. Immunoprecipitated and affinity-purified Tomo-1 from cultured rat hippocampal neurons was ubiquitinated, and the levels of ubiquitinated Tomo-1 dramatically increased upon pharmacological proteasome blockade. Moreover, Tomo-1 ubiquitination appeared to be mediated through an interaction with the E3 ubiquitin ligase HRD1, as immunoprecipitation of Tomo-1 from neurons co-precipitated HRD1, and this interaction increases upon proteasome inhibition. Further, in vitro reactions indicated direct, HRD1 concentration-dependent Tomo-1 ubiquitination. We also noted that the UPS regulates both Tomo-1 expression and functional output, as HRD1 KD in hippocampal neurons increased Tomo-1 protein level and dendritic spine density. Notably, the effect of HRD1 KD on spine density was mitigated by additional KD of Tomo-1, indicating a direct HRD1/Tomo-1 effector relationship. In summary, our results indicate that the UPS is likely to participate in tuning synaptic efficacy and spine dynamics by precise regulation of neuronal Tomo-1 levels.


Assuntos
Espinhas Dendríticas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas R-SNARE/metabolismo , Ubiquitina/metabolismo , Animais , Células Cultivadas , Espinhas Dendríticas/enzimologia , Espinhas Dendríticas/genética , Feminino , Hipocampo/citologia , Hipocampo/enzimologia , Masculino , Proteínas do Tecido Nervoso/genética , Neurônios/enzimologia , Densidade Pós-Sináptica/genética , Densidade Pós-Sináptica/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Ligação Proteica , Proteínas R-SNARE/genética , Ratos , Ratos Sprague-Dawley , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
3.
J Neurosci ; 36(44): 11208-11222, 2016 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-27807164

RESUMO

Neural networks engaged in high-frequency activity rely on sustained synaptic vesicle recycling and coordinated recruitment from functionally distinct synaptic vesicle (SV) pools. However, the molecular pathways matching neural activity to SV dynamics and release requirements remain unclear. Here we identify unique roles of SNARE-binding Tomosyn1 (Tomo1) proteins as activity-dependent substrates that regulate dynamics of SV pool partitioning at rat hippocampal synapses. Our analysis is based on monitoring changes in distinct functionally defined SV pools via V-Glut1-pHluorin fluorescence in cultured hippocampal neurons in response to alterations in presynaptic protein expression. Specifically, we find knockdown of Tomo1 facilitates release efficacy from the Readily Releasable Pool (RRP), and regulates SV distribution to the Total Recycling Pool (TRP), which is matched by a decrease in the SV Resting Pool. Notably, these effects were reversed by Tomo1 rescue and overexpression. Further, we identify that these actions of Tomo1 are regulated via activity-dependent phosphorylation by cyclin-dependent kinase 5 (Cdk5). Assessment of molecular interactions that may contribute to these actions identified Tomo1 interaction with the GTP-bound state of Rab3A, an SV GTPase involved in SV targeting and presynaptic membrane tethering. In addition, Tomo1 via Rab3A-GTP was also observed to interact with Synapsin 1a/b cytoskeletal interacting proteins. Finally, our data indicate that Tomo1 regulation of SV pool sizes serves to adapt presynaptic neurotransmitter release to chronic silencing of network activity. Overall, the results establish Tomo1 proteins as central mediators in neural activity-dependent changes in SV distribution among SV pools. SIGNIFICANCE STATEMENT: Although information transfer at central synapses via sustained high-frequency neural activity requires coordinated synaptic vesicle (SV) recycling, the mechanism(s) by which synapses sense and dynamically modify SV pools to match network demands remains poorly defined. To advance understanding, we quantified SV pool sizes and their sensitivity to neural activity while altering Tomo1 expression, a putative regulator of the presynaptic Readily Releasable Pool. Remarkably, we find Tomo1 actions to extend beyond the Readily Releasable Pool to mediate the Total Recycling Pool and SV Resting Pool distribution, and this action is sensitive to neural activity through Cdk5 phosphorylation of Tomo1. Moreover, Tomo1 appears to exert these actions through interaction with Rab3A-GTP and synapsin proteins. Together, our results argue that Tomo1 is a central mediator of SV availability for neurotransmission.


Assuntos
Guanosina Trifosfato/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Terminações Pré-Sinápticas/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/metabolismo , Proteína rab3A de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Feminino , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Masculino , Ratos , Sinapses
4.
Traffic ; 15(9): 997-1015, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24909540

RESUMO

Rab GTPases associated with insulin-containing secretory granules (SGs) are key in targeting, docking and assembly of molecular complexes governing pancreatic ß-cell exocytosis. Four Rab3 isoforms along with Rab27A are associated with insulin granules, yet elucidation of the distinct roles of these Rab families on exocytosis remains unclear. To define specific actions of these Rab families we employ Rab3GAP and/or EPI64A GTPase-activating protein overexpression in ß-cells from wild-type or Ashen mice to selectively transit the entire Rab3 family or Rab27A to a GDP-bound state. Ashen mice carry a spontaneous mutation that eliminates Rab27A expression. Using membrane capacitance measurements we find that GTP/GDP nucleotide cycling of Rab27A is essential for generation of the functionally defined immediately releasable pool (IRP) and central to regulating the size of the readily releasable pool (RRP). By comparison, nucleotide cycling of Rab3 GTPases, but not of Rab27A, is essential for a kinetically rapid filling of the RRP with SGs. Aside from these distinct functions, Rab3 and Rab27A GTPases demonstrate considerable functional overlap in building the readily releasable granule pool. Hence, while Rab3 and Rab27A cooperate to generate release-ready SGs in ß-cells, they also direct unique kinetic and functional properties of the exocytotic pathway.


Assuntos
Exocitose/fisiologia , GTP Fosfo-Hidrolases/metabolismo , Insulina/metabolismo , Proteínas rab3 de Ligação ao GTP/metabolismo , Animais , Nucléolo Celular/metabolismo , Grânulos Citoplasmáticos/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Transporte Proteico/fisiologia , Vesículas Secretórias/metabolismo
5.
J Biol Chem ; 289(24): 17087-99, 2014 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-24782308

RESUMO

Neuronal exocytosis depends on efficient formation of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes and is regulated by tomosyn, a SNARE-binding protein. To gain new information about tomosyn's activity, we characterized its mobility and organization on the plasma membrane (PM) in relation to other SNARE proteins and inhibition of exocytosis. By using direct stochastic optical reconstruction microscopy (dSTORM), we found tomosyn to be organized in small clusters adjacent to syntaxin clusters. In addition, we show that tomosyn is present in both syntaxin-tomosyn complexes and syntaxin-SNAP25-tomosyn complexes. Tomosyn mutants that lack residues 537-578 or 897-917 from its ß-propeller core diffused faster on the PM and exhibited reduced binding to SNAP25, suggesting that these mutants shift the equilibrium between tomosyn-syntaxin-SNAP25 complexes on the PM to tomosyn-syntaxin complexes. As these deletion mutants impose less inhibition on exocytosis, we suggest that tomosyn inhibition is mediated via tomosyn-syntaxin-SNAP25 complexes and not tomosyn-syntaxin complexes. These findings characterize, for the first time, tomosyn's dynamics at the PM and its relation to its inhibition of exocytosis.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Proteínas R-SNARE/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo , Sintaxina 1/metabolismo , Animais , Sítios de Ligação , Membrana Celular/metabolismo , Exocitose , Deleção de Genes , Células HEK293 , Humanos , Camundongos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Células PC12 , Ligação Proteica , Transporte Proteico , Proteínas R-SNARE/química , Proteínas R-SNARE/genética , Ratos , Proteína 25 Associada a Sinaptossoma/química , Proteína 25 Associada a Sinaptossoma/genética , Sintaxina 1/química , Sintaxina 1/genética
6.
J Neurosci ; 32(48): 17128-42, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23197706

RESUMO

Mutations that alter signaling through the mammalian target of rapamycin complex 1 (mTORC1), a well established regulator of neuronal protein synthesis, have been linked to autism and cognitive dysfunction. Although previous studies have established a role for mTORC1 as necessary for enduring changes in postsynaptic function, here we demonstrate that dendritic mTORC1 activation in rat hippocampal neurons also drives a retrograde signaling mechanism promoting enhanced neurotransmitter release from apposed presynaptic terminals. This novel mode of synaptic regulation conferred by dendritic mTORC1 is locally implemented, requires downstream synthesis of brain-derived neurotrophic factor as a retrograde messenger, and is engaged in an activity-dependent fashion to support homeostatic trans-synaptic control of presynaptic function. Our findings thus reveal that mTORC1-dependent translation in dendrites subserves a unique mode of synaptic regulation, highlighting an alternative regulatory pathway that could contribute to the social and cognitive dysfunction that accompanies dysregulated mTORC1 signaling.


Assuntos
Dendritos/metabolismo , Hipocampo/metabolismo , Complexos Multiproteicos/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Animais Recém-Nascidos , Dendritos/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Complexos Multiproteicos/genética , Ratos , Transdução de Sinais/fisiologia , Transmissão Sináptica/fisiologia , Serina-Treonina Quinases TOR/genética
7.
J Biol Chem ; 286(16): 14542-53, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21330375

RESUMO

Tomosyn is a 130-kDa cytosolic R-SNARE protein that associates with Q-SNAREs and reduces exocytotic activity. Two paralogous genes, tomosyn-1 and -2, occur in mammals and produce seven different isoforms via alternative splicing. Here, we map the structural differences between the yeast homologue of m-tomosyn-1, Sro7, and tomosyn genes/isoforms to identify domains critical to the regulation of exocytotic activity to tomosyn that are outside the soluble N-ethylmaleimide-sensitive attachment receptor motif. Homology modeling of m-tomosyn-1 based on the known structure of yeast Sro7 revealed a highly conserved functional conformation but with tomosyn containing three additional loop domains that emanate from a ß-propeller core. Notably, deletion of loops 1 and 3 eliminates tomosyn inhibitory activity on secretion without altering its soluble N-ethylmaleimide-sensitive attachment receptor pairing with syntaxin1A. By comparison, deletion of loop 2, which contains the hypervariable splice region, did not reduce the ability of tomosyn to inhibit regulated secretion. However, exon variation within the hypervariable splice region resulted in significant differences in protein accumulation of tomosyn-2 isoforms. Functional analysis of s-tomosyn-1, m-tomosyn-1, m-tomosyn-2, and xb-tomosyn-2 demonstrated that they exert similar inhibitory effects on elevated K(+)-induced secretion in PC12 cells, although m-tomosyn-2 was novel in strongly augmenting basal secretion. Finally, we report that m-tomosyn-1 is a target substrate for SUMO 2/3 conjugation and that mutation of this small ubiquitin-related modifier target site (Lys-730) enhances m-tomosyn-1 inhibition of secretion without altering interaction with syntaxin1A. Together these results suggest that multiple domains outside the R-SNARE of tomosyn are critical to the efficacy of inhibition by tomosyn on exocytotic secretion.


Assuntos
Proteínas do Tecido Nervoso/química , Proteínas R-SNARE/química , Processamento Alternativo , Motivos de Aminoácidos , Animais , Membrana Celular/metabolismo , Exocitose , Hormônio do Crescimento Humano/metabolismo , Humanos , Células PC12 , Estrutura Terciária de Proteína , Ratos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sintaxina 1/química
8.
Front Endocrinol (Lausanne) ; 13: 875865, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35795145

RESUMO

The adrenal medulla plays a critical role in mammalian homeostasis and the stress response. It is populated by clustered chromaffin cells that secrete epinephrine or norepinephrine along with peptides into the bloodstream affecting distant target organs. Despite been heavily studied, the central control of adrenal medulla and in-situ spatiotemporal responsiveness remains poorly understood. For this work, we continuously monitored the electrical activity of individual adrenomedullary chromaffin cells in the living anesthetized rat using multielectrode arrays. We measured the chromaffin cell activity under basal and physiological stress conditions and characterized the functional micro-architecture of the adrenal medulla. Under basal conditions, chromaffin cells fired action potentials with frequencies between ~0.2 and 4 Hz. Activity was almost completely driven by sympathetic inputs coming through the splanchnic nerve. Chromaffin cells were organized into independent local networks in which cells fired in a specific order, with latencies from hundreds of microseconds to a few milliseconds. Electrical stimulation of the splanchnic nerve evoked almost exactly the same spatiotemporal firing patterns that occurred spontaneously. Hypoglycemic stress, induced by insulin administration resulted in increased activity of a subset of the chromaffin cells. In contrast, respiratory arrest induced by lethal anesthesia resulted in an increase in the activity of virtually all chromaffin cells before cessation of all activity. These results suggest a stressor-specific activation of adrenomedullary chromaffin cell networks and revealed a surprisingly complex electrical organization that likely reflects the dynamic nature of the adrenal medulla's neuroendocrine output during basal conditions and during different types of physiological stress.


Assuntos
Medula Suprarrenal , Células Cromafins , Medula Suprarrenal/inervação , Medula Suprarrenal/metabolismo , Animais , Células Cromafins/metabolismo , Epinefrina , Mamíferos/metabolismo , Norepinefrina , Ratos , Nervos Esplâncnicos/metabolismo
9.
Biophys J ; 99(4): 1311-20, 2010 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-20713017

RESUMO

Biological processes are governed by extensive networks of dynamic molecular interactions. Yet, establishing a spatial and temporal map of these interactions and their direct relationship to specific cell functions has remained a challenge. Here, we implement sensitized emission Förster resonance energy transfer (FRET) stoichiometry under total internal reflection fluorescence (TIRF) microscopy. We demonstrate through quantitative analysis and modeling that evanescent fields must be precisely matched between FRET excitation wavelengths to isolate dynamic interactions between bimolecular FRET pairs that are not entirely membrane-delimited. We then use TIRF-FRET to monitor the behavior of individual insulin-containing secretory granules at the plasma membrane of living cells, while simultaneously tracking the dynamic interaction between the GTPase Rab27A and its effector Slp4A, on those same granules. Notably, insulin granules that underwent exocytosis demonstrated a specific increase in Rab27A-GTP/Slp4A FRET in the 5 s before membrane fusion, which coincided temporally with an increase in granule displacement and mobility. These results demonstrate an initial spatiotemporal mapping of a dynamic protein-protein interaction on individual secretory granules that is linked to a specific granule behavior in living cells.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Insulina/metabolismo , Microscopia de Fluorescência/métodos , Mapeamento de Interação de Proteínas/métodos , Vesículas Secretórias/metabolismo , Animais , Calibragem , Linhagem Celular , Corantes Fluorescentes/metabolismo , Secreção de Insulina , Camundongos , Ligação Proteica , Solubilidade , Transfecção , Proteínas de Transporte Vesicular/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP
10.
Physiol Rep ; 8(9): e14428, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32358861

RESUMO

Members of the Rab3 gene family are considered central to membrane trafficking of synaptic vesicles at mammalian central excitatory synapses. Recent evidence, however, indicates that the Rab27B-GTPase, which is highly homologous to the Rab3 family, is also enriched on SV membranes and co-localize with Rab3A and Synaptotagmin at presynaptic terminals. While functional roles of Rab3A have been well-established, little functional information exists on the role of Rab27B in synaptic transmission. Here we report on functional effects of Rab27B at SC-CA1 and DG-MF hippocampal synapses. The data establish distinct functional actions of Rab27B and demonstrate functions of Rab27B that differ between SC-CA1 and DG-MF synapses. Rab27B knockout reduced frequency facilitation compared to wild-type (WT) controls at the DG/MF-CA3 synaptic region, while increasing facilitation at the SC-CA1 synaptic region. Remarkably, Rab27B KO resulted in a complete elimination of LTP at the MF-CA3 synapse with no effect at the SC-CA1 synapse. These actions are similar to those previously reported for Rab3A KO. Specificity of action on LTP to Rab27B was confirmed as LTP was rescued in response to lentiviral infection and expression of human Rab27B, but not to GFP, in the DG in the Rab27B KO mice. Notably, the effect of Rab27B KO on MF-CA3 LTP occurred in spite of continued expression of Rab3A in the Rab27B KO. Overall, the results provide a novel perspective in suggesting that Rab27B and Rab3A act synergistically, perhaps via sequential effector recruitment or signaling for presynaptic LTP expression in this hippocampal synaptic region.


Assuntos
Hipocampo/metabolismo , Terminações Pré-Sinápticas/metabolismo , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia , Animais , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína rab3A de Ligação ao GTP/metabolismo
11.
J Physiol ; 586(22): 5367-81, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18801842

RESUMO

The small GTPase Rab27a, along with the isoforms of Rab3, is present on insulin secretory granules and has been implicated in regulation of Ca(2+)-triggered exocytosis. We have used membrane capacitance measurements to define the role of Rab27a in regulating the size and refilling of distinct pools of insulin granules by comparison of evoked secretory responses from Rab27a-null ashen and strain-matched wild-type control pancreatic beta-cells. We find that ashen beta-cells display a kinetic defect in refilling of readily releasable and immediately releasable vesicle pools (RRP and IRP, respectively) in response to depolarization-evoked Ca(2+) influx. The deficit in IRP refilling was not observed in the presence of stimulatory glucose concentrations (16.7 mm), though incomplete refilling of the RRP persisted. Comparatively, beta-cells from Rab3a(-/-) mice exhibited complete refilling of the IRP and RRP, demonstrating that Rab27a and Rab3a exert distinct roles in the insulin granule secretory pathway. Further, depletion of the RRP in ashen beta-cells was twofold faster than that of control beta-cells. These deficits in refilling and exocytotic rate in ashen beta-cells were absent when cAMP-regulatory pathways were activated. Elevated cAMP increased the RRP pool size, and complete refilling of the RRP occurred in ashen beta-cells; responses were comparable to wild-type controls. These effects of cAMP were largely eliminated by Rp-cAMP inhibition of PKA, indicating that PKA acts on vesicle priming downstream or via pathways independent of Rab27a. In summary, Rab27a exerts dual roles in glucose-mediated insulin granule exocytosis, facilitating refilling of releasable granule pools while also limiting the rate of release from these pools.


Assuntos
Células Secretoras de Insulina/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Proteínas de Transporte/metabolismo , AMP Cíclico/metabolismo , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Feminino , Glucose/farmacologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Transdução de Sinais , Proteínas rab de Ligação ao GTP/deficiência , Proteínas rab de Ligação ao GTP/genética , Proteínas rab27 de Ligação ao GTP , Proteína rab3A de Ligação ao GTP/deficiência , Proteína rab3A de Ligação ao GTP/genética , Proteína rab3A de Ligação ao GTP/metabolismo
12.
Brain Res ; 1043(1-2): 76-86, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15862520

RESUMO

Stress stimulates the adrenal medulla to rapidly secrete catecholamines (CAs), and the adrenal cortex to release progesterone (PROG), which may locally regulate stress-induced CA release. We used bovine chromaffin cells to investigate the effects of PROG on CA secretion. PROG dose-dependently inhibited CA secretion induced by nicotinic acetylcholine receptor (nAChR) agonist 1,1-dimethyl-4-phenlypiperazinium iodide (DMPP) up to 77%. Pre-incubation with PROG up to 1 h increased this inhibition. 3alpha,5alpha-Tetrahydroprogesterone (3alpha,5alpha-THP) and dexamethasone were less potent inhibitors. Patch-clamp techniques revealed that PROG co-applied with DMPP inhibited peak DMPP-induced current up to 68% and with 3 min pre-incubation inhibited both peak and integrated current up to approximately 95%. Monitoring of FURA-2 showed that PROG similarly inhibited parallel changes in intracellular-free Ca(++) concentration. PROG also inhibited CA secretion elicited by elevated K(+) (38%), and, in single cells, suppressed Ca(++) current evoked by step depolarization, inhibiting amplitude by 15%, and reducing the time constant of current decay during depolarization by 57%. In contrast to the immediate inhibition of nicotinic current, inhibition of Ca(++) current became statistically significant only after 1 min exposure to PROG. PROG did not inhibit secretion stimulated by high Ca(++) perfusion of permeabilized cells. These data suggest that PROG inhibits CA secretion from chromaffin cells predominantly by rapidly inhibiting nAChRs, and by gradually enhancing the inactivation of voltage-dependent Ca(++) channels (VDCCs), but not by affecting secretory processes downstream of Ca(++) influx. This study supports a role for adrenocortical PROG in the regulation of CA secretion during stress.


Assuntos
Catecolaminas/metabolismo , Células Cromafins/efeitos dos fármacos , Células Cromafins/metabolismo , Progesterona/farmacologia , Animais , Cálcio/metabolismo , Canais de Cálcio/fisiologia , Bovinos , Células Cultivadas , Células Cromafins/citologia , Iodeto de Dimetilfenilpiperazina/farmacologia , Relação Dose-Resposta a Droga , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Agonistas Nicotínicos/farmacologia , Receptores Nicotínicos/fisiologia
13.
Cytotechnology ; 67(3): 573-83, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-24549789

RESUMO

We have developed an improved procedure for isolating and transfecting a chromaffin cell-enriched population of primary cells from adult mouse adrenal glands. Significantly, the parameters of a novel electroporation transfection technique were optimized to achieve an average transfection efficiency of 45 % on the small number of cells derived from the mouse glands. Such transfection efficiency was previously unachievable with the electroporation protocols conventionally used with bovine chromaffin cells, even with use of large cell numbers. Our small scale technique now makes feasible the use of genetically homogenous inbred mouse models for investigations on the exocytotic pathway without the time, expense, and cellular changes associated with viral approaches. High fidelity co-expression of multiple plasmids in individual cells is a further advantage of the procedure. To assess whether the biophysical characteristics of mouse adrenal chromaffin cells were altered by this process, we examined structural integrity using immunocytochemistry and functional response to stimuli using calcium imaging, amperometry, and whole-cell capacitance and current clamp recordings. We conclude these parameters are minimally affected. Finally, we demonstrate that high transfection efficiency makes possible the use of primary mouse adrenal chromaffin cells, rather than a cell line, in human growth hormone secretion assays for high throughput evaluation of secretion.

14.
PLoS One ; 10(5): e0125596, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25951179

RESUMO

The small G-protein Rab27A has been shown to regulate the intracellular trafficking of secretory granules in various cell types. However, the presence, subcellular localization and functional impact of Rab27A on digestive enzyme secretion by mouse pancreatic acinar cells are poorly understood. Ashen mice, which lack the expression of Rab27A due to a spontaneous mutation, were used to investigate the function of Rab27A in pancreatic acinar cells. Isolated pancreatic acini were prepared from wild-type or ashen mouse pancreas by collagenase digestion, and CCK- or carbachol-induced amylase secretion was measured. Secretion occurring through the major-regulated secretory pathway, which is characterized by zymogen granules secretion, was visualized by Dextran-Texas Red labeling of exocytotic granules. The minor-regulated secretory pathway, which operates through the endosomal/lysosomal pathway, was characterized by luminal cell surface labeling of lysosomal associated membrane protein 1 (LAMP1). Compared to wild-type, expression of Rab27B was slightly increased in ashen mouse acini, while Rab3D and digestive enzymes (amylase, lipase, chymotrypsin and elastase) were not affected. Localization of Rab27B, Rab3D and amylase by immunofluorescence was similar in both wild-type and ashen acinar cells. The GTP-bound states of Rab27B and Rab3D in wild-type and ashen mouse acini also remained similar in amount. In contrast, acini from ashen mice showed decreased amylase release induced by CCK- or carbachol. Rab27A deficiency reduced the apical cell surface labeling of LAMP1, but did not affect that of Dextran-Texas Red incorporation into the fusion pockets at luminal surface. These results show that Rab27A is present in mouse pancreatic acinar cells and mainly regulates secretion through the minor-regulated pathway.


Assuntos
Amilases/metabolismo , Pâncreas/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Exocitose , Masculino , Camundongos , Camundongos Endogâmicos ICR , Pâncreas/citologia , Frações Subcelulares/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab27 de Ligação ao GTP
15.
Cell Rep ; 12(3): 396-404, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26166572

RESUMO

Neurotransmitter release probability (P(r)) largely determines the dynamic properties of synapses. While much is known about the role of presynaptic proteins in transmitter release, their specific contribution to synaptic plasticity is unclear. One such protein, tomosyn, is believed to reduce P(r) by interfering with the SNARE complex formation. Tomosyn is enriched at hippocampal mossy fiber-to-CA3 pyramidal cell synapses (MF-CA3), which characteristically exhibit low P(r), strong synaptic facilitation, and pre-synaptic protein kinase A (PKA)-dependent long-term potentiation (LTP). To evaluate tomosyn's role in MF-CA3 function, we used a combined knockdown (KD)-optogenetic strategy whereby presynaptic neurons with reduced tomosyn levels were selectively activated by light. Using this approach in mouse hippocampal slices, we found that facilitation, LTP, and PKA-induced potentiation were significantly impaired at tomosyn-deficient synapses. These findings not only indicate that tomosyn is a key regulator of MF-CA3 plasticity but also highlight the power of a combined KD-optogenetic approach to determine the role of presynaptic proteins.


Assuntos
Fibras Musgosas Hipocampais/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Plasticidade Neuronal/fisiologia , Proteínas R-SNARE/fisiologia , RNA Interferente Pequeno/metabolismo , Animais , Técnicas de Silenciamento de Genes/métodos , Humanos , Camundongos , Fibras Musgosas Hipocampais/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Optogenética/métodos , Proteínas R-SNARE/genética , Proteínas R-SNARE/metabolismo
16.
Brain Res ; 992(1): 30-42, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-14604770

RESUMO

The neurotoxin alpha-latrotoxin elicits spontaneous exocytosis of neurotransmitter from neurons and peptide hormones from endocrine cells. While the mechanism of action is not fully understood, both Ca(2+)-dependent and Ca(2+)-independent pathways participate in the facilitation of release, with the relative contribution of the pathways differing among neuronal and endocrine cell types. Here, we investigate the actions of alpha-latrotoxin on neuroendocrine nerve endings that emanate from central nervous system neurons and, therefore, are unique in that they possess properties of central nerve endings and endocrine cells. Using intracellular [Ca(2+)] measurements both calcium-independent receptors for latrotoxin (CIRL or latrophilin) and neurexin 1 alpha receptors were found to be functionally present. Interaction of alpha-latrotoxin with these receptors stimulated secretion of vasopressin and oxytocin neuropeptide. The secretory response was entirely dependent upon toxin-mediated extracellular Ca(2+) influx, although alpha-latrotoxin also consistently triggered mobilization of Ca(2+) from an intracellular store. The mobilization of intracellular Ca(2+) relied on alpha-latrotoxin-mediated Na(+) influx and was blocked by the protonophore FCCP, thereby implicating mitochondria as the Ca(2+) store being mobilized. Using the whole cell recording configuration of the patch clamp, we report that alpha-latrotoxin interaction with the CIRL receptor on these nerve endings resulted in ionic pore formation, generating unitary inward current steps of 20 pA and a channel conductance of approximately 220 pS in Ca(2+)-free saline. Thus, alpha-latrotoxin stimulates Ca(2+)-dependent exocytosis in neurohypophysial nerve endings through receptor interaction and insertion of Ca(2+) permeable membrane pores. While alpha-latrotoxin mobilizes intracellular Ca(2+) stores the elevation in [Ca(2+)] reached is insufficient to trigger measurable exocytosis.


Assuntos
Cálcio/metabolismo , Terminações Nervosas/efeitos dos fármacos , Neurossecreção/efeitos dos fármacos , Receptores de Peptídeos/metabolismo , Venenos de Aranha/farmacologia , Animais , Células Cultivadas , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Glicoproteínas , Líquido Intracelular/química , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mitocôndrias/metabolismo , Terminações Nervosas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos , Neurossecreção/fisiologia , Ocitocina/metabolismo , Técnicas de Patch-Clamp , Neuro-Hipófise/efeitos dos fármacos , Neuro-Hipófise/fisiologia , Ratos , Ratos Sprague-Dawley , Vasopressinas/metabolismo
17.
Nat Commun ; 5: 4834, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25189398

RESUMO

Autophagy deregulation during obesity contributes to the pathogenesis of diverse metabolic disorders. However, without understanding the molecular mechanism of obesity interference in autophagy, development of therapeutic strategies for correcting such defects in obese individuals is challenging. Here we show that a chronic increase of the cytosolic calcium concentration in hepatocytes during obesity and lipotoxicity attenuates autophagic flux by preventing the fusion between autophagosomes and lysosomes. As a pharmacological approach to restore cytosolic calcium homeostasis in vivo, we administered the clinically approved calcium channel blocker verapamil to obese mice. Such treatment successfully increases autophagosome-lysosome fusion in liver, preventing accumulation of protein inclusions and lipid droplets and suppressing inflammation and insulin resistance. As calcium channel blockers have been safely used in clinics for the treatment of hypertension for more than 30 years, our results suggest they may be a safe therapeutic option for restoring autophagic flux and treating metabolic pathologies in obese patients.


Assuntos
Autofagia/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Lisossomos/metabolismo , Doenças Metabólicas/tratamento farmacológico , Obesidade/complicações , Fagossomos/metabolismo , Verapamil/farmacologia , Animais , Autofagia/efeitos dos fármacos , Cálcio/metabolismo , Citosol/metabolismo , Ecocardiografia , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Doenças Metabólicas/etiologia , Doenças Metabólicas/fisiopatologia , Camundongos
18.
Mol Biol Cell ; 22(11): 1907-18, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21460182

RESUMO

Dynamin is a master regulator of membrane fission in endocytosis. However, a function for dynamin immediately upon fusion has also been suspected from a variety of experiments that measured release of granule contents. The role of dynamin guanosine triphosphate hydrolase (GTPase) activity in controlling fusion pore expansion and postfusion granule membrane topology was investigated using polarization optics and total internal reflection fluorescence microscopy (pTIRFM) and amperometry. A dynamin-1 (Dyn1) mutant with increased GTPase activity resulted in transient deformations consistent with rapid fusion pore widening after exocytosis; a Dyn1 mutant with decreased activity slowed fusion pore widening by stabilizing postfusion granule membrane deformations. The experiments indicate that, in addition to its role in endocytosis, GTPase activity of dynamin regulates the rapidity of fusion pore expansion from tens of milliseconds to seconds after fusion. These findings expand the membrane-sculpting repertoire of dynamin to include the regulation of immediate postfusion events in exocytosis that control the rate of release of soluble granule contents.


Assuntos
Dinamina I/metabolismo , Exocitose , GTP Fosfo-Hidrolases/metabolismo , Proteínas Recombinantes/metabolismo , Vesículas Secretórias/metabolismo , Animais , Catecolaminas/metabolismo , Bovinos , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Células Cultivadas , Células Cromafins , Dinamina I/genética , Elasticidade , GTP Fosfo-Hidrolases/genética , Humanos , Fusão de Membrana/genética , Mutação de Sentido Incorreto , Neuropeptídeo Y/metabolismo , Transporte Proteico , Proteínas Recombinantes/genética , Vesículas Secretórias/ultraestrutura
19.
Front Neuroanat ; 4: 149, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21191478

RESUMO

The protein tomosyn decreases synaptic transmission and release probability of vesicles, and is essential for modulating synaptic transmission in neurons. In this study, we provide a detailed description of the expression and localization patterns of tomosyn1 and tomosyn2 in the subareas of the mouse hippocampus. Using confocal and two-photon high-resolution microscopy we demonstrate that tomosyn colocalizes with several pre- and postsynaptic markers and is found mainly in glutamatergic synapses. Specifically, we show that tomosyn1 is differentially distributed in the mouse hippocampus and concentrated mainly in the hilus and mossy fibers. Surprisingly, we found that tomosyn2 is expressed in the subiculum, CA1 and CA2 pyramidal cell bodies, dendrites and spines, and colocalizes with PSD95, suggesting a postsynaptic role. These results suggest that in addition to the well-characterized presynaptic function of tomosyn in neurotransmitter release, tomosyn2 might have a postsynaptic function, and place tomosyn as a more general regulator of synaptic transmission and plasticity.

20.
Mol Biol Cell ; 19(2): 485-97, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18003982

RESUMO

Membrane fusion is a process that intimately involves both proteins and lipids. Although the SNARE proteins, which ultimately overcome the energy barrier for fusion, have been extensively studied, regulation of the energy barrier itself, determined by specific membrane lipids, has been largely overlooked. Our findings reveal a novel function for SNARE proteins in reducing the energy barrier for fusion, by directly binding and sequestering fusogenic lipids to sites of fusion. We demonstrate a specific interaction between Syntaxin1A and the fusogenic lipid phosphatidic acid, in addition to multiple polyphosphoinositide lipids, and define a polybasic juxtamembrane region within Syntaxin1A as its lipid-binding domain. In PC-12 cells, Syntaxin1A mutations that progressively reduced lipid binding resulted in a progressive reduction in evoked secretion. Moreover, amperometric analysis of fusion events driven by a lipid-binding-deficient Syntaxin1A mutant (5RK/A) demonstrated alterations in fusion pore dynamics, suggestive of an energetic defect in secretion. Overexpression of the phosphatidic acid-generating enzyme, phospholipase D1, completely rescued the secretory defect seen with the 5RK/A mutant. Moreover, knockdown of phospholipase D1 activity drastically reduced control secretion, while leaving 5RK/A-mediated secretion relatively unaffected. Altogether, these data suggest that Syntaxin1A-lipid interactions are a critical determinant of the energetics of SNARE-catalyzed fusion events.


Assuntos
Metabolismo dos Lipídeos , Fusão de Membrana , Sintaxina 1/metabolismo , Sequência de Aminoácidos , Animais , Toxinas Botulínicas/metabolismo , Catálise , Membrana Celular/metabolismo , Sobrevivência Celular , Humanos , Espaço Intracelular/metabolismo , Dados de Sequência Molecular , Proteínas Munc18/metabolismo , Mutação/genética , Células PC12 , Fenótipo , Ácidos Fosfatídicos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Ratos , Proteína 25 Associada a Sinaptossoma/metabolismo , Sintaxina 1/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA