Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nature ; 629(8010): 235-243, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38499039

RESUMEN

Biogenic monoamines-vital transmitters orchestrating neurological, endocrinal and immunological functions1-5-are stored in secretory vesicles by vesicular monoamine transporters (VMATs) for controlled quantal release6,7. Harnessing proton antiport, VMATs enrich monoamines around 10,000-fold and sequester neurotoxicants to protect neurons8-10. VMATs are targeted by an arsenal of therapeutic drugs and imaging agents to treat and monitor neurodegenerative disorders, hypertension and drug addiction1,8,11-16. However, the structural mechanisms underlying these actions remain unclear. Here we report eight cryo-electron microscopy structures of human VMAT1 in unbound form and in complex with four monoamines (dopamine, noradrenaline, serotonin and histamine), the Parkinsonism-inducing MPP+, the psychostimulant amphetamine and the antihypertensive drug reserpine. Reserpine binding captures a cytoplasmic-open conformation, whereas the other structures show a lumenal-open conformation stabilized by extensive gating interactions. The favoured transition to this lumenal-open state contributes to monoamine accumulation, while protonation facilitates the cytoplasmic-open transition and concurrently prevents monoamine binding to avoid unintended depletion. Monoamines and neurotoxicants share a binding pocket that possesses polar sites for specificity and a wrist-and-fist shape for versatility. Variations in this pocket explain substrate preferences across the SLC18 family. Overall, these structural insights and supporting functional studies elucidate the mechanism of vesicular monoamine transport and provide the basis to develop therapeutics for neurodegenerative diseases and substance abuse.


Asunto(s)
Monoaminas Biogénicas , Interacciones Farmacológicas , Proteínas de Transporte Vesicular de Monoaminas , Humanos , 1-Metil-4-fenilpiridinio/química , 1-Metil-4-fenilpiridinio/metabolismo , 1-Metil-4-fenilpiridinio/farmacología , Anfetamina/química , Anfetamina/farmacología , Anfetamina/metabolismo , Sitios de Unión , Monoaminas Biogénicas/química , Monoaminas Biogénicas/metabolismo , Microscopía por Crioelectrón , Dopamina/química , Dopamina/metabolismo , Modelos Moleculares , Norepinefrina/química , Norepinefrina/metabolismo , Unión Proteica , Protones , Reserpina/farmacología , Reserpina/química , Reserpina/metabolismo , Serotonina/química , Serotonina/metabolismo , Especificidad por Sustrato , Proteínas de Transporte Vesicular de Monoaminas/química , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/ultraestructura
2.
Nature ; 626(7998): 427-434, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38081299

RESUMEN

Vesicular monoamine transporter 2 (VMAT2) accumulates monoamines in presynaptic vesicles for storage and exocytotic release, and has a vital role in monoaminergic neurotransmission1-3. Dysfunction of monoaminergic systems causes many neurological and psychiatric disorders, including Parkinson's disease, hyperkinetic movement disorders and depression4-6. Suppressing VMAT2 with reserpine and tetrabenazine alleviates symptoms of hypertension and Huntington's disease7,8, respectively. Here we describe cryo-electron microscopy structures of human VMAT2 complexed with serotonin and three clinical drugs at 3.5-2.8 Å, demonstrating the structural basis for transport and inhibition. Reserpine and ketanserin occupy the substrate-binding pocket and lock VMAT2 in cytoplasm-facing and lumen-facing states, respectively, whereas tetrabenazine binds in a VMAT2-specific pocket and traps VMAT2 in an occluded state. The structures in three distinct states also reveal the structural basis of the VMAT2 transport cycle. Our study establishes a structural foundation for the mechanistic understanding of substrate recognition, transport, drug inhibition and pharmacology of VMAT2 while shedding light on the rational design of potential therapeutic agents.


Asunto(s)
Microscopía por Crioelectrón , Proteínas de Transporte Vesicular de Monoaminas , Humanos , Sitios de Unión , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Ketanserina/química , Ketanserina/metabolismo , Ketanserina/farmacología , Reserpina/química , Reserpina/metabolismo , Reserpina/farmacología , Serotonina/química , Serotonina/metabolismo , Especificidad por Sustrato , Tetrabenazina/química , Tetrabenazina/metabolismo , Tetrabenazina/farmacología , Proteínas de Transporte Vesicular de Monoaminas/antagonistas & inhibidores , Proteínas de Transporte Vesicular de Monoaminas/química , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/ultraestructura
3.
Nature ; 623(7989): 1086-1092, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37914936

RESUMEN

Monoamine neurotransmitters such as dopamine and serotonin control important brain pathways, including movement, sleep, reward and mood1. Dysfunction of monoaminergic circuits has been implicated in various neurodegenerative and neuropsychiatric disorders2. Vesicular monoamine transporters (VMATs) pack monoamines into vesicles for synaptic release and are essential to neurotransmission3-5. VMATs are also therapeutic drug targets for a number of different conditions6-9. Despite the importance of these transporters, the mechanisms of substrate transport and drug inhibition of VMATs have remained elusive. Here we report cryo-electron microscopy structures of the human vesicular monoamine transporter VMAT2 in complex with the antichorea drug tetrabenazine, the antihypertensive drug reserpine or the substrate serotonin. Remarkably, the two drugs use completely distinct inhibition mechanisms. Tetrabenazine binds VMAT2 in a lumen-facing conformation, locking the luminal gating lid in an occluded state to arrest the transport cycle. By contrast, reserpine binds in a cytoplasm-facing conformation, expanding the vestibule and blocking substrate access. Structural analyses of VMAT2 also reveal the conformational changes following transporter isomerization that drive substrate transport into the vesicle. These findings provide a structural framework for understanding the physiology and pharmacology of neurotransmitter packaging by synaptic vesicular transporters.


Asunto(s)
Neurotransmisores , Reserpina , Serotonina , Tetrabenazina , Proteínas de Transporte Vesicular de Monoaminas , Humanos , Inhibidores de Captación Adrenérgica/química , Inhibidores de Captación Adrenérgica/farmacología , Transporte Biológico/efectos de los fármacos , Microscopía por Crioelectrón , Neurotransmisores/química , Neurotransmisores/farmacología , Reserpina/química , Reserpina/farmacología , Serotonina/metabolismo , Transmisión Sináptica , Tetrabenazina/química , Tetrabenazina/farmacología , Proteínas de Transporte Vesicular de Monoaminas/antagonistas & inhibidores , Proteínas de Transporte Vesicular de Monoaminas/química , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/ultraestructura , Especificidad por Sustrato/efectos de los fármacos
4.
PLoS One ; 8(9): e75952, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098749

RESUMEN

UNLABELLED: (18)F-FP-(+)-DTBZ is a novel PET radiotracer targeting vesicular monoamine transporter type 2 (VMAT2). The goal was to explore the image features in normal human brains with (18)F-FP-(+)-DTBZ as a reference of molecular landmark for clinical diagnosis in Parkinson's disease (PD) and related disorders. MATERIALS AND METHODS: A total of 22 healthy subjects (59.3±6.0 years old) including 7 men and 15 women were recruited for MRI and (18)F-FP-(+)-DTBZ PET scans. A total number of 55 brain VOIs were selected for quantitation analysis. The regional specific uptake ratio (SUR) was calculated with occipital as reference from MRI-based spatially normalized (18)F-FP-(+)-DTBZ images. Regional percentage SUR to that of anterior putamen was calculated. Average SUR images were displayed in 2D and 3D space to illustrate the image patterns. The correlation between age and regional VMAT2 uptake was also examined. RESULTS: Visual assessment showed symmetric uptake of (18)F-FP-(+)-DTBZ and obviously highest in striatum, followed by nucleus accumbens, hypothalamus, substantia nigra, and raphe nuclei. Quantification analysis revealed striatal VMAT2 density of anterior putamen>posterior putamen>caudate nucleus. Other subcortical regions were with moderate VMAT2 distribution (6∼51% SUR of anterior putamen), while slightly lower VMAT2 was observed in cerebellum (10.60% SUR) and much lower in neocortex (<5% SUR). No significant correlation of SUR to age was found in subcortical regions. CONCLUSION: Using (18)F-FP-(+)-DTBZ PET, we showed the 2D and 3D imaging features of the VMAT2 distribution in vivo in healthy aging brains. The in vivo imaging characteristics of VMAT2 is consistent with the expression of VMAT2 in a recent autopsy study. Therefore, 3D visualization and higher image quality of (18)F-FP-(+)-DTBZ PET imaging might potentially be a powerful biomarker in detecting VMAT2 distribution of subcortical regions, and for Parkinson's disease and related neuropsychiatric disorders involving related monoaminergic systems.


Asunto(s)
Neuroimagen/métodos , Enfermedad de Parkinson/diagnóstico , Tomografía de Emisión de Positrones/métodos , Proteínas de Transporte Vesicular de Monoaminas/ultraestructura , Femenino , Radioisótopos de Flúor , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Persona de Mediana Edad , Estándares de Referencia , Estadísticas no Paramétricas
5.
Ultrastruct Pathol ; 37(2): 102-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23573890

RESUMEN

Our previous histochemical and ultrastructural studies have identified, in human catecholamine neurons, abundant spherical acidophilic protein bodies (pb), which originate from regular mitochondria, retaining their double membrane. In locus coeruleus (LC) neurons, pb have somatodendritic distribution and are unequivocal storage vesicles for noradrenaline, as demonstrated by immunolocalization of Dopamine-ß-Hydroxylase. In the present study, in order to reinforce the identity of pb as monoamine storage sites in human LC, and to assess their potential of somatodendritic release, we studied the subcellular immunolocalization of chromogranin A (CgA) and vesicular monoamine transporter 2 (VMAT2), given the fact that their localization defines the vesicles capacity of filling with monoamine and hence exocytotic release. The data provided in the present study, demonstrate the novel ultrastructural immunolocalization of both CgA and VMAT2 in protein bodies, supporting their involvement in somatodendritic storage and release of noradrenaline in human LC. Since the molecular mechanism of LC somatodendritic exocytosis remains largely elusive, the present study may shed light to a better understanding of this mechanism.


Asunto(s)
Cromogranina A/ultraestructura , Locus Coeruleus/ultraestructura , Neuronas/ultraestructura , Orgánulos/ultraestructura , Proteínas de Transporte Vesicular de Monoaminas/ultraestructura , Adulto , Anciano , Anciano de 80 o más Años , Cromogranina A/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patología , Femenino , Humanos , Locus Coeruleus/metabolismo , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Norepinefrina/metabolismo , Orgánulos/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA