Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Circ Res ; 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39011635

RESUMEN

BACKGROUND: Cardiac hypertrophy compensates for increased biomechanical stress of the heart induced by prevalent cardiovascular pathologies but can result in cardiac failure if left untreated. We hypothesized that the tail-anchored protein dysferlin with multiple Ca2+-binding C2-domains is critical for the integrity of the transverse-axial tubule (TAT) network inside cardiomyocytes and contributes to the proliferation of TAT endomembranes during pressure overload-induced cardiac hypertrophy. OBJECTIVE: To reveal the impact of the membrane fusion and repair protein dysferlin on TAT network stabilization and proliferation necessary for the hypertrophic growth of cardiomyocytes. METHODS AND RESULTS: Super-resolution light and electron microscopy of mouse cardiomyocytes identified a specific localization of dysferlin in a vesicular compartment in nanometric proximity to contact sites of the TAT network with the sarcoplasmic reticulum, a.k.a. junctional complexes for Ca2+-induced Ca2+ release. Mass spectrometry was used to characterize the cardiac dysferlin interactome, thereby identifying a novel protein interaction with the membrane-tethering sarcoplasmic reticulum protein juncophilin-2, a putative interactor of L-type Ca2+ channels and ryanodine receptor Ca2+ release channels in junctional complexes. While the dysferlin knockout caused a mild progressive phenotype of dilated cardiomyopathy in the mouse heart, global proteome analysis revealed changes preceding systolic failure. Following transverse aortic constriction, dysferlin protein expression was significantly increased in hypertrophied wild-type myocardium, while dysferlin knockout animals presented markedly reduced left-ventricular hypertrophy. Live-cell membrane imaging demonstrated a profound reorganization of the TAT network in wild-type left-ventricular myocytes post-transverse aortic constriction with robust proliferation of axial tubules, which critically depended on the increased expression of dysferlin within newly emerging tubule components. CONCLUSIONS: Dysferlin represents a new molecular target in cardiac disease that protects the integrity of tubule-sarcoplasmic reticulum junctional complexes for regulated excitation-contraction coupling and controls TAT network reorganization and tubular membrane proliferation in cardiomyocyte hypertrophy induced by pressure overload.

3.
Nature ; 611(7937): 827-834, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36418452

RESUMEN

Vacuolar-type adenosine triphosphatases (V-ATPases)1-3 are electrogenic rotary mechanoenzymes structurally related to F-type ATP synthases4,5. They hydrolyse ATP to establish electrochemical proton gradients for a plethora of cellular processes1,3. In neurons, the loading of all neurotransmitters into synaptic vesicles is energized by about one V-ATPase molecule per synaptic vesicle6,7. To shed light on this bona fide single-molecule biological process, we investigated electrogenic proton-pumping by single mammalian-brain V-ATPases in single synaptic vesicles. Here we show that V-ATPases do not pump continuously in time, as suggested by observing the rotation of bacterial homologues8 and assuming strict ATP-proton coupling. Instead, they stochastically switch between three ultralong-lived modes: proton-pumping, inactive and proton-leaky. Notably, direct observation of pumping revealed that physiologically relevant concentrations of ATP do not regulate the intrinsic pumping rate. ATP regulates V-ATPase activity through the switching probability of the proton-pumping mode. By contrast, electrochemical proton gradients regulate the pumping rate and the switching of the pumping and inactive modes. A direct consequence of mode-switching is all-or-none stochastic fluctuations in the electrochemical gradient of synaptic vesicles that would be expected to introduce stochasticity in proton-driven secondary active loading of neurotransmitters and may thus have important implications for neurotransmission. This work reveals and emphasizes the mechanistic and biological importance of ultraslow mode-switching.


Asunto(s)
Encéfalo , Mamíferos , ATPasas de Translocación de Protón Vacuolares , Animales , Adenosina Trifosfato/metabolismo , Encéfalo/enzimología , Encéfalo/metabolismo , Mamíferos/metabolismo , Protones , Vesículas Sinápticas/enzimología , Vesículas Sinápticas/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Neurotransmisores/metabolismo , Transmisión Sináptica , Factores de Tiempo , Cinética
5.
Eur Biophys J ; 51(6): 465-482, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35904588

RESUMEN

The size, polydispersity, and electron density profile of synaptic vesicles (SVs) can be studied by small-angle X-ray scattering (SAXS), i.e. by X-ray diffraction from purified SV suspensions in solution. Here we show that size and shape transformations, as they appear in the functional context of these important synaptic organelles, can also be monitored by SAXS. In particular, we have investigated the active uptake of neurotransmitters, and find a mean vesicle radius increase of about 12% after the uptake of glutamate, which indicates an unusually large extensibility of the vesicle surface, likely to be accompanied by conformational changes of membrane proteins and rearrangements of the bilayer. Changes in the electron density profile (EDP) give first indications for such a rearrangement. Details of the protein structure are screened, however, by SVs polydispersity. To overcome the limitations of large ensemble averages and heterogeneous structures, we therefore propose serial X-ray diffraction by single free electron laser pulses. Using simulated data for realistic parameters, we show that this is in principle feasible, and that even spatial distances between vesicle proteins could be assessed by this approach.


Asunto(s)
Ácido Glutámico , Vesículas Sinápticas , Transporte Biológico , Proteínas/metabolismo , Dispersión del Ángulo Pequeño , Vesículas Sinápticas/química , Vesículas Sinápticas/metabolismo , Difracción de Rayos X
6.
Neuron ; 110(9): 1483-1497.e7, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35263617

RESUMEN

Vesicular transporters (VTs) define the type of neurotransmitter that synaptic vesicles (SVs) store and release. While certain mammalian neurons release multiple transmitters, it is not clear whether the release occurs from the same or distinct vesicle pools at the synapse. Using quantitative single-vesicle imaging, we show that a vast majority of SVs in the rodent brain contain only one type of VT, indicating specificity for a single neurotransmitter. Interestingly, SVs containing dual transporters are highly diverse (27 types) but small in proportion (2% of all SVs), excluding the largest pool that carries VGLUT1 and ZnT3 (34%). Using VGLUT1-ZnT3 SVs, we demonstrate that the transporter colocalization influences the SV content and synaptic quantal size. Thus, the presence of diverse transporters on the same vesicle is bona fide, and depending on the VT types, this may act to regulate neurotransmitter type, content, and release in space and time.


Asunto(s)
Proteínas de Transporte de Neurotransmisores , Vesículas Sinápticas , Animales , Mamíferos , Proteínas de Transporte de Membrana , Neurotransmisores , Sinapsis , Vesículas Sinápticas/fisiología , Proteína 1 de Transporte Vesicular de Glutamato
7.
Cell Rep ; 34(2): 108623, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33440152

RESUMEN

Vesicular glutamate transporters (VGLUTs) fill synaptic vesicles with glutamate. VGLUTs were originally identified as sodium-dependent transporters of inorganic phosphate (Pi), but the physiological relevance of this activity remains unclear. Heterologous expression of all three VGLUTs greatly augments intracellular Pi levels. Using neuronal models, we show that translocation of VGLUTs to the plasma membrane during exocytosis results in highly increased Pi uptake. VGLUT-mediated Pi influx is counteracted by Pi efflux. Synaptosomes prepared from perinatal VGLUT2-/- mice that are virtually free of VGLUTs show drastically reduced cytosolic Pi levels and fail to import Pi. Glutamate partially competes with sodium (Na+)/Pi (NaPi)-uptake mediated by VGLUTs but does not appear to be transported. A nanobody that blocks glutamate transport by binding to the cytoplasmic domain of VGLUT1 abolishes Pi transport when co-expressed with VGLUT1. We conclude that VGLUTs have a dual function that is essential for both vesicular glutamate loading and Pi restoration in neurons.


Asunto(s)
Transporte Biológico/fisiología , Fosfatos/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Animales , Humanos , Ratas , Transfección
8.
Elife ; 92020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33164744

RESUMEN

Insulin secretion from ß-cells is reduced at the onset of type-1 and during type-2 diabetes. Although inflammation and metabolic dysfunction of ß-cells elicit secretory defects associated with type-1 or type-2 diabetes, accompanying changes to insulin granules have not been established. To address this, we performed detailed functional analyses of insulin granules purified from cells subjected to model treatments that mimic type-1 and type-2 diabetic conditions and discovered striking shifts in calcium affinities and fusion characteristics. We show that this behavior is correlated with two subpopulations of insulin granules whose relative abundance is differentially shifted depending on diabetic model condition. The two types of granules have different release characteristics, distinct lipid and protein compositions, and package different secretory contents alongside insulin. This complexity of ß-cell secretory physiology establishes a direct link between granule subpopulation and type of diabetes and leads to a revised model of secretory changes in the diabetogenic process.


Diabetes is a disease that occurs when sugar levels in the blood can no longer be controlled by a hormone called insulin. People with type 1 diabetes lose the ability to produce insulin after their immune system attacks the ß-cells in their pancreas that make this hormone. People with type 2 diabetes develop the disease when ß-cells become exhausted from increased insulin demand and stop producing insulin. ß-cells store insulin in small compartments called granules. When blood sugar levels rise, these granules fuse with the cell membrane allowing ß-cells to release large quantities of insulin at once. This fusion is disrupted early in type 1 diabetes, but later in type 2: the underlying causes of these disruptions are unclear. In the laboratory, signals that trigger inflammation and molecules called fatty acids can mimic type 1 or type 2 diabetes respectively when applied to insulin-producing cells. Kreutzberger, Kiessling et al. wanted to know whether pro-inflammatory molecules and fatty acids affect insulin granules differently at the molecular level. To do this, insulin-producing cells were grown in the lab and treated with either fatty acids or pro-inflammatory molecules. The insulin granules of these cells were then isolated. Next, the composition of the granules and how they fused to lab-made membranes that mimic the cell membrane was examined. The experiments revealed that healthy ß-cells have two types of granules, each with a different version of a protein called synaptotagmin. Cells treated with molecules mimicking type 1 diabetes lost granules with synaptotagmin-7, while granules with synaptotagmin-9 were lost in cells treated with fatty acids to imitate type 2 diabetes. Each type of granule responded differently to calcium levels in the cell and secreted different molecules, indicating that each elicits a different diabetic response in the body. These findings suggest that understanding how insulin granules are formed and regulated may help find treatments for type 1 and 2 diabetes, possibly leading to therapies that reverse the loss of different types of granules. Additionally, the molecules of these granules may also be used as markers to determine the stage of diabetes. More broadly, these results show how understanding how molecule release changes with disease in different cell types may help diagnose or stage a disease.


Asunto(s)
Calcio/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Exocitosis , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Colesterol/metabolismo , Citocinas/farmacología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Exocitosis/efectos de los fármacos , Humanos , Insulina/genética , Células Secretoras de Insulina/efectos de los fármacos , Células PC12 , Palmitatos/farmacología , Ratas , Proteínas SNARE/metabolismo , Vías Secretoras , Esfingomielinas/metabolismo , Sinaptotagminas/metabolismo
9.
Sci Rep ; 10(1): 7540, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32371955

RESUMEN

Large dense-core vesicles (LDCVs) contain a variety of neurotransmitters, proteins, and hormones such as biogenic amines and peptides, together with microRNAs (miRNAs). Isolation of LDCVs is essential for functional studies including vesicle fusion, vesicle acidification, monoamine transport, and the miRNAs stored in LDCVs. Although several methods were reported for purifying LDCVs, the final fractions are significantly contaminated by other organelles, compromising biochemical characterization. Here we isolated LDCVs (chromaffin granules) with high yield and purity from bovine adrenal medulla. The fractionation protocol combines differential and continuous sucrose gradient centrifugation, allowing for reducing major contaminants such as mitochondria. Purified LDCVs show robust acidification by the endogenous V-ATPase and undergo SNARE-mediated fusion with artificial membranes. Interestingly, LDCVs contain specific miRNAs such as miR-375 and miR-375 is stabilized by protein complex against RNase A. This protocol can be useful in research on the biological functions of LDCVs.


Asunto(s)
Médula Suprarrenal/fisiología , Técnicas Citológicas/métodos , Animales , Bovinos , Fraccionamiento Celular , Gránulos Cromafines/metabolismo , Fusión de Membrana , MicroARNs/metabolismo , Mitocondrias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurotransmisores/metabolismo
10.
Nat Commun ; 10(1): 3904, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467284

RESUMEN

Regulated exocytosis of synaptic vesicles is substantially faster than of endocrine dense core vesicles despite similar molecular machineries. The reasons for this difference are unknown and could be due to different regulatory proteins, different spatial arrangements, different vesicle sizes, or other factors. To address these questions, we take a reconstitution approach and compare regulated SNARE-mediated fusion of purified synaptic and dense core chromaffin and insulin vesicles using a single vesicle-supported membrane fusion assay. In all cases, Munc18 and complexin are required to restrict fusion in the absence of calcium. Calcium triggers fusion of all docked vesicles. Munc13 (C1C2MUN domain) is required for synaptic and enhanced insulin vesicle fusion, but not for chromaffin vesicles, correlating inversely with the presence of CAPS protein on purified vesicles. Striking disparities in calcium-triggered fusion rates are observed, increasing with curvature with time constants 0.23 s (synaptic vesicles), 3.3 s (chromaffin vesicles), and 9.1 s (insulin vesicles) and correlating with rate differences in cells.


Asunto(s)
Fusión de Membrana/fisiología , Proteínas SNARE/metabolismo , Vesículas Secretoras/metabolismo , Vesículas Sinápticas/metabolismo , Animales , Transporte Biológico , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Membrana Celular/metabolismo , Exocitosis , Humanos , Insulina , Proteínas Munc18/metabolismo , Proteínas del Tejido Nervioso , Células PC12 , Ratas
11.
Elife ; 82019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31090538

RESUMEN

Synaptophysins 1 and 2 and synaptogyrins 1 and 3 constitute a major family of synaptic vesicle membrane proteins. Unlike other widely expressed synaptic vesicle proteins such as vSNAREs and synaptotagmins, the primary function has not been resolved. Here, we report robust elevation in the probability of release of readily releasable vesicles with both high and low release probabilities at a variety of synapse types from knockout mice missing all four family members. Neither the number of readily releasable vesicles, nor the timing of recruitment to the readily releasable pool was affected. The results suggest that family members serve as negative regulators of neurotransmission, acting directly at the level of exocytosis to dampen connection strength selectively when presynaptic action potentials fire at low frequency. The widespread expression suggests that chemical synapses may play a frequency filtering role in biological computation that is more elemental than presently envisioned. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Asunto(s)
Neuronas/metabolismo , Vesículas Sinápticas/metabolismo , Sinaptogirinas/deficiencia , Sinaptofisina/deficiencia , Animales , Ratones Noqueados , Transmisión Sináptica
12.
Cereb Cortex ; 29(10): 4263-4276, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-30541023

RESUMEN

Brain functions are extremely sensitive to pH changes because of the pH-dependence of proteins involved in neuronal excitability and synaptic transmission. Here, we show that the Na+/H+ exchanger Nhe1, which uses the Na+ gradient to extrude H+, is expressed at both inhibitory and excitatory presynapses. We disrupted Nhe1 specifically in mice either in Emx1-positive glutamatergic neurons or in parvalbumin-positive cells, mainly GABAergic interneurons. While Nhe1 disruption in excitatory neurons had no effect on overall network excitability, mice with disruption of Nhe1 in parvalbumin-positive neurons displayed epileptic activity. From our electrophysiological analyses in the CA1 of the hippocampus, we conclude that the disruption in parvalbumin-positive neurons impairs the release of GABA-loaded vesicles, but increases the size of GABA quanta. The latter is most likely an indirect pH-dependent effect, as Nhe1 was not expressed in purified synaptic vesicles itself. Conclusively, our data provide first evidence that Nhe1 affects network excitability via modulation of inhibitory interneurons.


Asunto(s)
Región CA1 Hipocampal/fisiología , Potenciales de la Membrana , Terminales Presinápticos/fisiología , Intercambiador 1 de Sodio-Hidrógeno/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Epilepsia/fisiopatología , Femenino , Neuronas GABAérgicas/fisiología , Ácido Glutámico/metabolismo , Interneuronas/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Terminales Presinápticos/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Ácido gamma-Aminobutírico/metabolismo
13.
Cell Rep ; 23(2): 535-545, 2018 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-29642010

RESUMEN

Vesicular glutamate transporters (VGLUTs) fill synaptic vesicles with glutamate and are thus essential for glutamatergic neurotransmission. However, VGLUTs were originally discovered as members of a transporter subfamily specific for inorganic phosphate (Pi). It is still unclear how VGLUTs accommodate glutamate transport coupled to an electrochemical proton gradient ΔµH+ with inversely directed Pi transport coupled to the Na+ gradient and the membrane potential. Using both functional reconstitution and heterologous expression, we show that VGLUT transports glutamate and Pi using a single substrate binding site but different coupling to cation gradients. When facing the cytoplasm, both ions are transported into synaptic vesicles in a ΔµH+-dependent fashion, with glutamate preferred over Pi. When facing the extracellular space, Pi is transported in a Na+-coupled manner, with glutamate competing for binding but at lower affinity. We conclude that VGLUTs have dual functions in both vesicle transmitter loading and Pi homeostasis within glutamatergic neurons.


Asunto(s)
Fosfatos/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Animales , Sitios de Unión , Transporte Biológico/efectos de los fármacos , Membrana Celular/metabolismo , Exocitosis/efectos de los fármacos , Ácido Glutámico/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Liposomas/química , Liposomas/metabolismo , Nigericina/farmacología , Células PC12 , Cloruro de Potasio/farmacología , Ratas , Ratas Wistar , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Especificidad por Sustrato , Vesículas Sinápticas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/genética
14.
Science ; 351(6276): 981-4, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26912364

RESUMEN

Synaptic transmission is mediated by the release of neurotransmitters, which involves exo-endocytotic cycling of synaptic vesicles. To maintain synaptic function, synaptic vesicles are refilled with thousands of neurotransmitter molecules within seconds after endocytosis, using the energy provided by an electrochemical proton gradient. However, it is unclear how transmitter molecules carrying different net charges can be efficiently sequestered while maintaining charge neutrality and osmotic balance. We used single-vesicle imaging to monitor pH and electrical gradients and directly showed different uptake mechanisms for glutamate and γ-aminobutyric acid (GABA) operating in parallel. In contrast to glutamate, GABA was exchanged for protons, with no other ions participating in the transport cycle. Thus, only a few components are needed to guarantee reliable vesicle filling with different neurotransmitters.


Asunto(s)
Ácido Glutámico/metabolismo , Transmisión Sináptica , Vesículas Sinápticas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Transporte Biológico , Endocitosis , Concentración de Iones de Hidrógeno , Ratones , Ratones Transgénicos , Imagen Molecular , Protones , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
15.
Biol Psychiatry ; 77(6): 526-36, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25176177

RESUMEN

BACKGROUND: The neuromodulatory transmitters, biogenic amines, have profound effects on multiple neurons and are essential for normal behavior and mental health. Here we report that the orphan transporter SLC10A4, which in the brain is exclusively expressed in presynaptic vesicles of monoaminergic and cholinergic neurons, has a regulatory role in dopamine homeostasis. METHODS: We used a combination of molecular and behavioral analyses, pharmacology, and in vivo amperometry to assess the role of SLC10A4 in dopamine-regulated behaviors. RESULTS: We show that SLC10A4 is localized on the same synaptic vesicles as either vesicular acetylcholine transporter or vesicular monoamine transporter 2. We did not find evidence for direct transport of dopamine by SLC10A4; however, synaptic vesicle preparations lacking SLC10A4 showed decreased dopamine vesicular uptake efficiency. Furthermore, we observed an increased acidification in synaptic vesicles isolated from mice overexpressing SLC10A4. Loss of SLC10A4 in mice resulted in reduced striatal serotonin, noradrenaline, and dopamine concentrations and a significantly higher dopamine turnover ratio. Absence of SLC10A4 led to slower dopamine clearance rates in vivo, which resulted in accumulation of extracellular dopamine. Finally, whereas SLC10A4 null mutant mice were slightly hypoactive, they displayed hypersensitivity to administration of amphetamine and tranylcypromine. CONCLUSIONS: Our results demonstrate that SLC10A4 is a vesicular monoaminergic and cholinergic associated transporter that is important for dopamine homeostasis and neuromodulation in vivo. The discovery of SLC10A4 and its role in dopaminergic signaling reveals a novel mechanism for neuromodulation and represents an unexplored target for the treatment of neurological and mental disorders.


Asunto(s)
Dopamina/metabolismo , Homeostasis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Anfetamina/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Ratones Transgénicos , Inhibidores de la Monoaminooxidasa/farmacología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/genética , Norepinefrina/metabolismo , ARN Mensajero/metabolismo , Serotonina/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Simportadores , Vesículas Sinápticas/metabolismo , Tranilcipromina/farmacología , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Proteínas de Transporte Vesicular/genética
16.
Neuron ; 84(6): 1287-301, 2014 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-25433636

RESUMEN

Vesicular glutamate transporters (VGLUTs) accumulate the neurotransmitter glutamate in synaptic vesicles. Transport depends on a V-ATPase-dependent electrochemical proton gradient (ΔµH+) and requires chloride ions, but how chloride acts and how ionic and charge balance is maintained during transport is controversial. Using a reconstitution approach, we used an exogenous proton pump to drive VGLUT-mediated transport either in liposomes containing purified VGLUT1 or in synaptic vesicles fused with proton-pump-containing liposomes. Our data show that chloride stimulation can be induced at both sides of the membrane. Moreover, chloride competes with glutamate at high concentrations. In addition, VGLUT1 possesses a cation binding site capable of binding H+ or K+ ions, allowing for proton antiport or K+ / H+ exchange. We conclude that VGLUTs contain two anion binding sites and one cation binding site, allowing the transporter to adjust to the changing ionic conditions during vesicle filling without being dependent on other transporters or channels.


Asunto(s)
Cloruros/metabolismo , Ácido Glutámico/metabolismo , Neurotransmisores/metabolismo , Potasio/metabolismo , Vesículas Sinápticas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/química , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Animales , Aniones , Sitios de Unión , Cationes Monovalentes/metabolismo , Liposomas/metabolismo , Ratones , Ratas
17.
J Biol Chem ; 289(23): 16326-35, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24778182

RESUMEN

Neuronal exocytosis is mediated by soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins. Before fusion, SNARE proteins form complexes bridging the membrane followed by assembly toward the C-terminal membrane anchors, thus initiating membrane fusion. After fusion, the SNARE complex is disassembled by the AAA-ATPase N-ethylmaleimide-sensitive factor that requires the cofactor α-SNAP to first bind to the assembled SNARE complex. Using chromaffin granules and liposomes we now show that α-SNAP on its own interferes with the zippering of membrane-anchored SNARE complexes midway through the zippering reaction, arresting SNAREs in a partially assembled trans-complex and preventing fusion. Intriguingly, the interference does not result in an inhibitory effect on synaptic vesicles, suggesting that membrane properties also influence the final outcome of α-SNAP interference with SNARE zippering. We suggest that binding of α-SNAP to the SNARE complex affects the ability of the SNARE complex to harness energy or transmit force to the membrane.


Asunto(s)
Fusión de Membrana , Proteínas SNARE/fisiología , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/fisiología , Animales , Bovinos , Endocitosis , Polarización de Fluorescencia , Transferencia Resonante de Energía de Fluorescencia , Proteolípidos
18.
Mol Cancer Res ; 7(3): 285-99, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19276185

RESUMEN

EpCAM has been described as a therapeutically relevant tumor marker. We noted an interaction between EpCAM and the tight junction protein claudin-7 and here explored the nature of this interaction and its effect on EpCAM-mediated functions. The interaction between EpCAM and claudin-7 was defined in HEK293 cells transfected with rat claudin-7 and EpCAM cDNA. Deletions of the epidermal growth factor-like and the thyroglobin repeat domains of EpCAM or the cytoplasmic domain of EpCAM or claudin-7 did not prevent the EpCAM-claudin-7 association. A chimeric EpCAM molecule with an exchange of the cytoplasmic and transmembrane domains and an EpCAM molecule with point mutations in an AxxxG motif in the transmembrane region do not associate with claudin-7. HEK cells and the rat pancreatic tumor line BSp73AS, transfected with (mutated) EpCAM and claudin-7 cDNA, revealed that the association of both molecules severely alters the functional activity of EpCAM. Claudin-7-associated EpCAM is recruited into tetraspanin-enriched membrane microdomains (TEM). The TEM-located claudin-7-EpCAM complex supports proliferation accompanied by sustained extracellular signal-regulated kinase-1/2 phosphorylation, up-regulation of antiapoptotic proteins, and drug resistance, but not EpCAM-mediated cell-cell adhesion. Enhanced motility may be supported by colocalization of claudin-7 with actin bundles, which is only seen in EpCAM-claudin-7-expressing cells. The EpCAM-claudin-7 complex strongly promotes tumorigenicity, accelerates tumor growth, and supports ascites production and thymic metastasis formation. High expression of the tumor marker EpCAM is frequently associated with poor prognosis, which could well rely on the EpCAM-claudin-7 association that prohibits EpCAM-mediated cell-cell adhesion but promotes migration, proliferation, apoptosis resistance, and tumorigenicity.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas de la Membrana/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Claudinas , Progresión de la Enfermedad , Molécula de Adhesión Celular Epitelial , Humanos , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Estructura Terciaria de Proteína , Ratas , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...