Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Exp Neurol ; 373: 114668, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38147972

RESUMO

The pathogenesis of degeneration in Parkinson's disease (PD) remains poorly understood but multiple lines of evidence have converged on the presynaptic protein α-synuclein (αsyn). αSyn has been shown to regulate several cellular processes, however, its normal function remains poorly understood. In this review, we will specifically focus on its role in exocytosis.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Humanos , alfa-Sinucleína/metabolismo , Doença de Parkinson/patologia , Exocitose/fisiologia
2.
Proc Natl Acad Sci U S A ; 120(42): e2309843120, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37812725

RESUMO

The burst firing of midbrain dopamine neurons releases a phasic dopamine signal that mediates reinforcement learning. At many synapses, however, high firing rates deplete synaptic vesicles (SVs), resulting in synaptic depression that limits release. What accounts for the increased release of dopamine by stimulation at high frequency? We find that adaptor protein-3 (AP-3) and its coat protein VPS41 promote axonal dopamine release by targeting vesicular monoamine transporter VMAT2 to the axon rather than dendrites. AP-3 and VPS41 also produce SVs that respond preferentially to high-frequency stimulation, independent of their role in axonal polarity. In addition, conditional inactivation of VPS41 in dopamine neurons impairs reinforcement learning, and this involves a defect in the frequency dependence of release rather than the amount of dopamine released. Thus, AP-3 and VPS41 promote the axonal polarity of dopamine release but enable learning by producing a distinct population of SVs tuned specifically to high firing frequency that confers the phasic release of dopamine.


Assuntos
Dopamina , Vesículas Sinápticas , Dopamina/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/genética , Proteínas Vesiculares de Transporte de Monoamina/metabolismo , Axônios/metabolismo , Mesencéfalo/metabolismo
3.
Nat Neurosci ; 26(10): 1685-1700, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37723322

RESUMO

Neural systems encode information in the frequency of action potentials, which is then decoded by synaptic transmission. However, the rapid, synchronous release of neurotransmitters depletes synaptic vesicles (SVs), limiting release at high firing rates. How then do synapses convey information about frequency? Here, we show in mouse hippocampal neurons and slices that the adaptor protein AP-3 makes a subset of SVs that respond specifically to high-frequency stimulation. Neurotransmitter transporters slot onto these SVs in different proportions, contributing to the distinct properties of release observed at different excitatory synapses. Proteomics reveals that AP-3 targets the phospholipid flippase ATP8A1 to SVs; loss of ATP8A1 recapitulates the defect in SV mobilization at high frequency observed with loss of AP-3. The mechanism involves recruitment of synapsin by the cytoplasmically oriented phosphatidylserine translocated by ATP8A1. Thus, ATP8A1 enables the subset of SVs made by AP-3 to release at high frequency.


Assuntos
Complexo 3 de Proteínas Adaptadoras , Adenosina Trifosfatases , Fosfolipídeos , Transmissão Sináptica , Vesículas Sinápticas , Animais , Camundongos , Fosfolipídeos/metabolismo , Sinapses/metabolismo , Sinapsinas/metabolismo , Vesículas Sinápticas/metabolismo , Complexo 3 de Proteínas Adaptadoras/metabolismo , Adenosina Trifosfatases/metabolismo
4.
bioRxiv ; 2023 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-37609166

RESUMO

The burst firing of midbrain dopamine neurons releases a phasic dopamine signal that mediates reinforcement learning. At many synapses, however, high firing rates deplete synaptic vesicles (SVs), resulting in synaptic depression that limits release. What accounts for the increased release of dopamine by stimulation at high frequency? We find that adaptor protein-3 (AP-3) and its coat protein VPS41 promote axonal dopamine release by targeting vesicular monoamine transporter VMAT2 to the axon rather than dendrites. AP-3 and VPS41 also produce SVs that respond preferentially to high frequency stimulation, independent of their role in axonal polarity. In addition, conditional inactivation of VPS41 in dopamine neurons impairs reinforcement learning, and this involves a defect in the frequency dependence of release rather than the amount of dopamine released. Thus, AP-3 and VPS41 promote the axonal polarity of dopamine release but enable learning by producing a novel population of SVs tuned specifically to high firing frequency that confers the phasic release of dopamine.

5.
NPJ Parkinsons Dis ; 9(1): 47, 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36977661
6.
J Biol Chem ; 299(5): 104646, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36965620

RESUMO

The solute carrier 17 family transports diverse organic anions using two distinct modes of coupling to a source of energy. Transporters that package glutamate and nucleotide into secretory vesicles for regulated release by exocytosis are driven by membrane potential but subject to allosteric regulation by H+ and Cl-. Other solute carrier 17 members including the lysosomal sialic acid exporter couple the flux of organic anion to cotransport of H+. To begin to understand how similar proteins can perform such different functions, we have studied Escherichia coli DgoT, a H+/galactonate cotransporter. A recent structure of DgoT showed many residues contacting D-galactonate, and we now find that they do not tolerate even conservative substitutions. In contrast, the closely related lysosomal H+/sialic acid cotransporter Sialin tolerates similar mutations, consistent with its recognition of diverse substrates with relatively low affinity. We also find that despite coupling to H+, DgoT transports more rapidly but with lower apparent affinity at high pH. Indeed, membrane potential can drive uptake, indicating electrogenic transport and suggesting a H+:galactonate stoichiometry >1. Located in a polar pocket of the N-terminal helical bundle, Asp46 and Glu133 are each required for net flux by DgoT, but the E133Q mutant exhibits robust exchange activity and rescues exchange by D46N, suggesting that these two residues operate in series to translocate protons. E133Q also shifts the pH sensitivity of exchange by DgoT, supporting a central role for the highly conserved TM4 glutamate in H+ coupling by DgoT.


Assuntos
Proteínas de Escherichia coli , Prótons , Simportadores , Ânions/metabolismo , Transporte Biológico , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Mutação , Simportadores/genética , Simportadores/metabolismo
7.
Proc Natl Acad Sci U S A ; 120(1): e2214897120, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36574702

RESUMO

During exocytosis, the fusion of secretory vesicle with plasma membrane forms a pore that regulates release of neurotransmitter and peptide. Heterogeneity of fusion pore behavior has been attributed to stochastic variation in a common exocytic mechanism, implying a lack of biological control. Using a fluorescent false neurotransmitter (FFN), we imaged dense core vesicle (DCV) exocytosis in primary mouse adrenal chromaffin cells by total internal reflection fluorescence microscopy at millisecond resolution and observed strikingly divergent modes of release, with fast events lasting <30 ms and slow events persisting for seconds. Dual imaging of slow events shows a delay in the entry of external dye relative to FFN release, suggesting exclusion by an extremely narrow pore <1 nm in diameter. Unbiased comprehensive analysis shows that the observed variation cannot be explained by stochasticity alone, but rather involves distinct mechanisms, revealing the bimodal nature of DCV exocytosis. Further, loss of calcium sensor synaptotagmin 7 increases the proportion of slow events without changing the intrinsic properties of either class, indicating the potential for independent regulation. The identification of two distinct mechanisms for release capable of independent regulation suggests a biological basis for the diversity of fusion pore behavior.


Assuntos
Células Cromafins , Vesículas de Núcleo Denso , Camundongos , Animais , Sinaptotagminas/metabolismo , Exocitose/fisiologia , Membrana Celular/metabolismo , Células Cromafins/metabolismo , Vesículas Secretórias/metabolismo , Fusão de Membrana/fisiologia , Cálcio/metabolismo
8.
Curr Opin Struct Biol ; 75: 102399, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35660266

RESUMO

Originally identified as transporters for inorganic phosphate, solute carrier 17 (SLC17) family proteins subserve diverse physiological roles. The vesicular glutamate transporters (VGLUTs) package the principal excitatory neurotransmitter glutamate into synaptic vesicles (SVs). In contrast, the closely related sialic acid transporter sialin mediates the flux of sialic acid in the opposite direction, from lysosomes to the cytoplasm. The two proteins couple in different ways to the H+ electrochemical gradient driving force, and high-resolution structures of the Escherichia coli homolog d-galactonate transporter (DgoT) and more recently rat VGLUT2 now begin to suggest the mechanisms involved as well as the basis for substrate specificity.


Assuntos
Transportadores de Ânions Orgânicos , Vesículas Sinápticas , Animais , Escherichia coli/metabolismo , Ácido Glutâmico/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Transportadores de Ânions Orgânicos/química , Transportadores de Ânions Orgânicos/metabolismo , Ratos , Especificidade por Substrato , Vesículas Sinápticas/metabolismo
9.
Proc Natl Acad Sci U S A ; 119(20): e2118430119, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35533272

RESUMO

The assembly of functional neuronal circuits requires appropriate numbers of distinct classes of neurons, but the mechanisms through which their relative proportions are established remain poorly defined. Investigating the mouse striatum, we found that the two most prominent subtypes of striatal interneurons, parvalbumin-expressing (PV+) GABAergic and cholinergic (ChAT+) interneurons, undergo extensive programmed cell death between the first and second postnatal weeks. Remarkably, the survival of PV+ and ChAT+ interneurons is regulated by distinct mechanisms mediated by their specific afferent connectivity. While long-range cortical inputs control PV+ interneuron survival, ChAT+ interneuron survival is regulated by local input from the medium spiny neurons. Our results identify input-specific circuit mechanisms that operate during the period of programmed cell death to establish the final number of interneurons in nascent striatal networks.


Assuntos
Corpo Estriado , Interneurônios , Córtex Cerebral/fisiologia , Corpo Estriado/fisiologia , Neurônios GABAérgicos/fisiologia , Interneurônios/fisiologia , Parvalbuminas
10.
J Cell Biol ; 221(5)2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35426896

RESUMO

The time course of signaling by peptide hormones, neural peptides, and other neuromodulators depends on their storage inside dense core vesicles (DCVs). Adaptor protein 3 (AP-3) assembles the membrane proteins that confer regulated release of DCVs and is thought to promote their trafficking from endosomes directly to maturing DCVs. We now find that regulated monoamine release from DCVs requires sorting nexin 5 (SNX5). Loss of SNX5 disrupts trafficking of the vesicular monoamine transporter (VMAT) to DCVs. The mechanism involves a role for SNX5 in retrograde transport of VMAT from endosomes to the TGN. However, this role for SNX5 conflicts with the proposed function of AP-3 in trafficking from endosomes directly to DCVs. We now identify a transient role for AP-3 at the TGN, where it associates with DCV cargo. Thus, retrograde transport from endosomes by SNX5 enables DCV assembly at the TGN by AP-3, resolving the apparent antagonism. A novel role for AP-3 at the TGN has implications for other organelles that also depend on this adaptor.


Assuntos
Complexo 3 de Proteínas Adaptadoras , Vesículas de Núcleo Denso , Endossomos , Nexinas de Classificação , Complexo 3 de Proteínas Adaptadoras/metabolismo , Transporte Biológico , Proteínas de Transporte/metabolismo , Vesículas de Núcleo Denso/metabolismo , Endossomos/metabolismo , Neurotransmissores/metabolismo , Transporte Proteico , Nexinas de Classificação/metabolismo
11.
Methods Mol Biol ; 2417: 29-44, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35099789

RESUMO

The analysis of organellar membrane transporters presents many technical problems. In general, their activity depends on a H+ electrochemical driving force (ΔµH+). However, transport itself influences the expression of ΔµH+ in standard radiotracer flux assays, making it difficult to disentangle the role of the chemical component ΔpH and the membrane potential Δψ. Whole endosome recording in voltage clamp circumvents many of these problems, controlling ionic conditions as well as membrane potential inside and outside the organelle . This approach has been used primarily to study the properties of endolysosomal channels, which generate substantial currents (Saito et al., J Biol Chem 282(37):27327-27333, 2007; Cang et al., Nat Chem Biol 10(6):463-469, 2014; Cang et al., Cell 152(4):778-790, 2013; Chen et al., Nat Protoc 12(8):1639-1658, 2017; Samie et al., Dev Cell 26(5):511-524, 2013; Wang et al., Cell 151(2):372-383, 2012). Electrogenic transport produces much smaller currents, but we have recently reported the detection of transport currents and an uncoupled Cl- conductance associated with the vesicular glutamate transporters (VGLUTs) that fill synaptic vesicles with glutamate (Chang et al., eLife 7:e34896, 2018). In this protocol, we will focus on the measurement of transport currents on enlarged endosomes of heterologous mammalian cells.


Assuntos
Vesículas Sinápticas , Proteínas Vesiculares de Transporte de Glutamato , Animais , Endossomos/metabolismo , Ácido Glutâmico/metabolismo , Mamíferos/metabolismo , Potenciais da Membrana , Vesículas Sinápticas/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/metabolismo
12.
Biochemistry ; 60(32): 2463-2470, 2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34319067

RESUMO

The role of glutamate in excitatory neurotransmission depends on its transport into synaptic vesicles by the vesicular glutamate transporters (VGLUTs). The three VGLUT isoforms exhibit a complementary distribution in the nervous system, and the knockout of each produces severe, pleiotropic neurological effects. However, the available pharmacology lacks sensitivity and specificity, limiting the analysis of both transport mechanism and physiological role. To develop new molecular probes for the VGLUTs, we raised six mouse monoclonal antibodies to VGLUT2. All six bind to a structured region of VGLUT2, five to the luminal face, and one to the cytosolic. Two are specific to VGLUT2, whereas the other four bind to both VGLUT1 and 2; none detect VGLUT3. Antibody 8E11 recognizes an epitope spanning the three extracellular loops in the C-domain that explains the recognition of both VGLUT1 and 2 but not VGLUT3. 8E11 also inhibits both glutamate transport and the VGLUT-associated chloride conductance. Since the antibody binds outside the substrate recognition site, it acts allosterically to inhibit function, presumably by restricting conformational changes. The isoform specificity also shows that allosteric inhibition provides a mechanism to distinguish between closely related transporters.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas Vesiculares de Transporte de Glutamato/imunologia , Regulação Alostérica/imunologia , Animais , Cloretos/metabolismo , Epitopos/química , Epitopos/imunologia , Ácido Glutâmico/metabolismo , Células HEK293 , Humanos , Isoformas de Proteínas/imunologia , Proteína Vesicular 1 de Transporte de Glutamato/química , Proteína Vesicular 1 de Transporte de Glutamato/imunologia , Proteína Vesicular 2 de Transporte de Glutamato/química , Proteína Vesicular 2 de Transporte de Glutamato/imunologia , Proteínas Vesiculares de Transporte de Glutamato/química , Xenopus laevis
13.
Annu Rev Pathol ; 16: 465-485, 2021 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-33497259

RESUMO

Specific proteins accumulate in neurodegenerative disease, and human genetics has indicated a causative role for many. In most cases, however, the mechanisms remain poorly understood. Degeneration is thought to involve a gain of abnormal function, although we do not know the normal function of many proteins implicated. The protein α-synuclein accumulates in the Lewy pathology of Parkinson's disease and related disorders, and mutations in α-synuclein cause degeneration, but we have not known its normal function or how it triggers disease. α-Synuclein localizes to presynaptic boutons and interacts with membranes in vitro. Overexpression slows synaptic vesicle exocytosis, and recent data suggest a normal role for the endogenous synucleins in dilation of the exocytic fusion pore. Disrupted membranes also appear surprisingly prominent in Lewy pathology. Synuclein thus interacts with membranes under both physiological and pathological conditions, suggesting that the normal function of synuclein may illuminate its role in degeneration.


Assuntos
Doenças Neurodegenerativas , Sinucleínas/genética , Sinucleínas/metabolismo , Animais , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/fisiopatologia
14.
Mol Psychiatry ; 26(9): 4795-4812, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-32398719

RESUMO

Serotonin and dopamine are associated with multiple psychiatric disorders. How they interact during development to affect subsequent behavior remains unknown. Knockout of the serotonin transporter or postnatal blockade with selective serotonin reuptake inhibitors (SSRIs) leads to novelty-induced exploration deficits in adulthood, potentially involving the dopamine system. Here, we show in the mouse that raphe nucleus serotonin neurons activate ventral tegmental area dopamine neurons via glutamate co-transmission and that this co-transmission is reduced in animals exposed postnatally to SSRIs. Blocking serotonin neuron glutamate co-transmission mimics this SSRI-induced hypolocomotion, while optogenetic activation of dopamine neurons reverses this hypolocomotor phenotype. Our data demonstrate that serotonin neurons modulate dopamine neuron activity via glutamate co-transmission and that this pathway is developmentally malleable, with high serotonin levels during early life reducing co-transmission, revealing the basis for the reduced novelty-induced exploration in adulthood due to postnatal SSRI exposure.


Assuntos
Ácido Glutâmico , Área Tegmentar Ventral , Animais , Neurônios Dopaminérgicos , Feminino , Camundongos , Camundongos Knockout , Gravidez , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
15.
Proc Natl Acad Sci U S A ; 117(51): 32701-32710, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33273122

RESUMO

α-Synuclein is expressed at high levels at presynaptic terminals, but defining its role in the regulation of neurotransmission under physiologically relevant conditions has proven elusive. We report that, in vivo, α-synuclein is responsible for the facilitation of dopamine release triggered by action potential bursts separated by short intervals (seconds) and a depression of release with longer intervals between bursts (minutes). These forms of presynaptic plasticity appear to be independent of the presence of ß- and γ-synucleins or effects on presynaptic calcium and are consistent with a role for synucleins in the enhancement of synaptic vesicle fusion and turnover. These results indicate that the presynaptic effects of α-synuclein depend on specific patterns of neuronal activity.


Assuntos
Dopamina/metabolismo , Neurônios/metabolismo , Substância Negra/metabolismo , alfa-Sinucleína/metabolismo , Anestésicos Inalatórios/farmacologia , Animais , Sinalização do Cálcio , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Feminino , Isoflurano/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurotransmissores/metabolismo , Substância Negra/citologia , Vesículas Sinápticas/metabolismo , alfa-Sinucleína/genética , gama-Sinucleína/metabolismo
16.
Science ; 368(6493): 893-897, 2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32439795

RESUMO

Synaptic vesicles accumulate neurotransmitters, enabling the quantal release by exocytosis that underlies synaptic transmission. Specific neurotransmitter transporters are responsible for this activity and therefore are essential for brain function. The vesicular glutamate transporters (VGLUTs) concentrate the principal excitatory neurotransmitter glutamate into synaptic vesicles, driven by membrane potential. However, the mechanism by which they do so remains poorly understood owing to a lack of structural information. We report the cryo-electron microscopy structure of rat VGLUT2 at 3.8-angstrom resolution and propose structure-based mechanisms for substrate recognition and allosteric activation by low pH and chloride. A potential permeation pathway for chloride intersects with the glutamate binding site. These results demonstrate how the activity of VGLUTs can be coordinated with large shifts in proton and chloride concentrations during the synaptic vesicle cycle to ensure normal synaptic transmission.


Assuntos
Canais de Cloreto/química , Canais de Cloreto/metabolismo , Cloretos/metabolismo , Ácido Glutâmico/metabolismo , Vesículas Sinápticas/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/química , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Animais , Sítios de Ligação , Microscopia Crioeletrônica , Concentração de Íons de Hidrogênio , Transporte de Íons , Potenciais da Membrana , Domínios Proteicos , Ratos
17.
Biochim Biophys Acta Biomembr ; 1862(12): 183259, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32147354

RESUMO

The transport of classical neurotransmitters into synaptic vesicles generally relies on a H+ electrochemical gradient (∆µH+). Synaptic vesicle uptake of glutamate depends primarily on the electrical component ∆ψ as the driving force, rather than the chemical component ∆pH. However, the vesicular glutamate transporters (VGLUTs) belong to the solute carrier 17 (SLC17) family, which includes closely related members that function as H+ cotransporters. Recent work has also shown that the VGLUTs undergo allosteric regulation by H+ and Cl-, and exhibit an associated Cl- conductance. These properties appear to coordinate VGLUT activity with the large ionic shifts that accompany the rapid recycling of synaptic vesicles driven by neural activity. Recent structural information also suggests common mechanisms that underlie the apparently divergent function of SLC17 family members, and that confer allosteric regulation.


Assuntos
Ácido Glutâmico/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/metabolismo , Regulação Alostérica , Animais , Transporte Biológico , Cloretos/metabolismo , Ligantes , Especificidade por Substrato , Proteínas Vesiculares de Transporte de Glutamato/química
18.
Mol Psychiatry ; 25(4): 732-749, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-30127471

RESUMO

Astrocytes orchestrate neural development by powerfully coordinating synapse formation and function and, as such, may be critically involved in the pathogenesis of neurodevelopmental abnormalities and cognitive deficits commonly observed in psychiatric disorders. Here, we report the identification of a subset of cortical astrocytes that are competent for regulating dopamine (DA) homeostasis during postnatal development of the prefrontal cortex (PFC), allowing for optimal DA-mediated maturation of excitatory circuits. Such control of DA homeostasis occurs through the coordinated activity of astroglial vesicular monoamine transporter 2 (VMAT2) together with organic cation transporter 3 and monoamine oxidase type B, two key proteins for DA uptake and metabolism. Conditional deletion of VMAT2 in astrocytes postnatally produces loss of PFC DA homeostasis, leading to defective synaptic transmission and plasticity as well as impaired executive functions. Our findings show a novel role for PFC astrocytes in the DA modulation of cognitive performances with relevance to psychiatric disorders.


Assuntos
Astrócitos/metabolismo , Disfunção Cognitiva/metabolismo , Dopamina/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Encéfalo/metabolismo , Disfunção Cognitiva/fisiopatologia , Dopamina/farmacologia , Homeostase , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/fisiologia
19.
J Neurochem ; 150(5): 475-486, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31269263

RESUMO

The protein α-synuclein has a central role in the pathogenesis of Parkinson's disease (PD). In this review, we discuss recent results concerning its primary function, which appears to be on cell membranes. The pre-synaptic location of synuclein has suggested a role in neurotransmitter release and it apparently associates with synaptic vesicles because of their high curvature. Indeed, synuclein over-expression inhibits synaptic vesicle exocytosis. However, loss of synuclein has not yet been shown to have a major effect on synaptic transmission. Consistent with work showing that synuclein can promote as well as sense membrane curvature, recent analysis of synuclein triple knockout mice now shows that synuclein accelerates dilation of the exocytic fusion pore. This form of regulation affects primarily the release of slowly discharged lumenal cargo such as neural peptides, but presumably also contributes to maintenance of the release site. This article is part of the Special Issue "Synuclein".


Assuntos
Doença de Parkinson/metabolismo , alfa-Sinucleína/fisiologia , Animais , Axônios/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Exocitose/fisiologia , Humanos , Fusão de Membrana/fisiologia , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/patologia , Mutação de Sentido Incorreto , Terminações Pré-Sinápticas/química , Domínios Proteicos , Dobramento de Proteína , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiologia , Proteínas Recombinantes/metabolismo , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura , alfa-Sinucleína/química , alfa-Sinucleína/deficiência , alfa-Sinucleína/genética
20.
PLoS Biol ; 17(5): e3000260, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31083648

RESUMO

Members of the solute carrier 17 (SLC17) family use divergent mechanisms to concentrate organic anions. Membrane potential drives uptake of the principal excitatory neurotransmitter glutamate into synaptic vesicles, whereas closely related proteins use proton cotransport to drive efflux from the lysosome. To delineate the divergent features of ionic coupling by the SLC17 family, we determined the structure of Escherichia coli D-galactonate/H+ symporter D-galactonate transporter (DgoT) in 2 states: one open to the cytoplasmic side and the other open to the periplasmic side with substrate bound. The structures suggest a mechanism that couples H+ flux to substrate recognition. A transition in the role of H+ from flux coupling to allostery may confer regulation by trafficking to and from the plasma membrane.


Assuntos
Metabolismo Energético , Escherichia coli/metabolismo , Transportadores de Ânions Orgânicos/química , Transportadores de Ânions Orgânicos/metabolismo , Transporte Biológico , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Conformação Proteica , Prótons , Açúcares Ácidos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA