Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nature ; 584(7820): E17, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32724206

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Nature ; 582(7813): 550-556, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32581380

RESUMEN

Parkinson's disease is characterized by loss of dopamine neurons in the substantia nigra1. Similar to other major neurodegenerative disorders, there are no disease-modifying treatments for Parkinson's disease. While most treatment strategies aim to prevent neuronal loss or protect vulnerable neuronal circuits, a potential alternative is to replace lost neurons to reconstruct disrupted circuits2. Here we report an efficient one-step conversion of isolated mouse and human astrocytes to functional neurons by depleting the RNA-binding protein PTB (also known as PTBP1). Applying this approach to the mouse brain, we demonstrate progressive conversion of astrocytes to new neurons that innervate into and repopulate endogenous neural circuits. Astrocytes from different brain regions are converted to different neuronal subtypes. Using a chemically induced model of Parkinson's disease in mouse, we show conversion of midbrain astrocytes to dopaminergic neurons, which provide axons to reconstruct the nigrostriatal circuit. Notably, re-innervation of striatum is accompanied by restoration of dopamine levels and rescue of motor deficits. A similar reversal of disease phenotype is also accomplished by converting astrocytes to neurons using antisense oligonucleotides to transiently suppress PTB. These findings identify a potentially powerful and clinically feasible approach to treating neurodegeneration by replacing lost neurons.


Asunto(s)
Astrocitos/citología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/citología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , Sustancia Negra/citología , Sustancia Negra/fisiología , Animales , Axones/fisiología , Dopamina/biosíntesis , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Femenino , Ribonucleoproteínas Nucleares Heterogéneas/deficiencia , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Técnicas In Vitro , Masculino , Ratones , Neostriado/citología , Neostriado/fisiología , Vías Nerviosas , Neurogénesis , Enfermedad de Parkinson/metabolismo , Fenotipo , Proteína de Unión al Tracto de Polipirimidina/deficiencia , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Sustancia Negra/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(20): e2111051119, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35537054

RESUMEN

Exocytosis and endocytosis are tightly coupled. In addition to initiating exocytosis, Ca2+ plays critical roles in exocytosis­endocytosis coupling in neurons and nonneuronal cells. Both positive and negative roles of Ca2+ in endocytosis have been reported; however, Ca2+ inhibition in endocytosis remains debatable with unknown mechanisms. Here, we show that synaptotagmin-1 (Syt1), the primary Ca2+ sensor initiating exocytosis, plays bidirectional and opposite roles in exocytosis­endocytosis coupling by promoting slow, small-sized clathrin-mediated endocytosis but inhibiting fast, large-sized bulk endocytosis. Ca2+-binding ability is required for Syt1 to regulate both types of endocytic pathways, the disruption of which leads to inefficient vesicle recycling under mild stimulation and excessive membrane retrieval following intense stimulation. Ca2+-dependent membrane tubulation may explain the opposite endocytic roles of Syt1 and provides a general membrane-remodeling working model for endocytosis determination. Thus, Syt1 is a primary bidirectional Ca2+ sensor facilitating clathrin-mediated endocytosis but clamping bulk endocytosis, probably by manipulating membrane curvature to ensure both efficient and precise coupling of endocytosis to exocytosis.


Asunto(s)
Endocitosis , Transmisión Sináptica , Sinaptotagmina I , Calcio/metabolismo , Endocitosis/fisiología , Exocitosis/fisiología , Neuronas/metabolismo , Sinaptotagmina I/metabolismo
4.
Basic Res Cardiol ; 115(5): 56, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32767120

RESUMEN

Sympathetic neural remodeling, which involves the inflammatory response, plays an important role in ventricular arrhythmias (VAs) after myocardial infarction (MI). Adrenergic receptors on macrophages potentially modulate the inflammatory response. We hypothesized that the increased level of catecholamines activates macrophages and regulates sympathetic neural remodeling after MI. We treated MI mice with either clodronate or metoprolol for 5 days following coronary artery ligation. Mice without treatment after MI and sham-operation mice served as the positive control and negative control, respectively. The norepinephrine levels in plasma and the peri-infarct myocardium increased by almost two-fold in the MI mice compared with the sham-operation mice. Both in vivo and ex vivo electrophysiology examinations showed that the vulnerability to VAs induced by MI was alleviated by macrophage depletion with clodronate and ß1-adrenergic blockade with metoprolol, which was in line with circulating and peri-infarct norepinephrine levels, sympathetic reinnervation, and the expression of nerve growth factor (NGF) 7 days after surgery. To further verify the interaction between catecholamines and macrophages, we preconditioned lipopolysaccharide-stimulated RAW 264.7 cells using epinephrine or epinephrine with selective adrenergic antagonists. The expression and release of inflammatory factors including NGF were enhanced by epinephrine. This effect was inhibited by metoprolol but not by other subtype antagonists. Our data suggested that the increased level of catecholamines, traditionally known as positive inotropes secreted from sympathetic nerve endings, might regulate cardiac sympathetic neural remodeling through ß1-adrenergic receptors on macrophages, subsequently inducing VAs after MI.


Asunto(s)
Arritmias Cardíacas/etiología , Macrófagos/fisiología , Infarto del Miocardio/complicaciones , Plasticidad Neuronal , Norepinefrina/sangre , Animales , Arritmias Cardíacas/sangre , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/sangre , Miocardio/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/metabolismo
5.
Cytotherapy ; 20(5): 670-686, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29576501

RESUMEN

BACKGROUND: This study explored the neural differentiation and therapeutic effects of stem cells from human exfoliated deciduous teeth (SHED) in a rat model of Parkinson's disease (PD). METHODS: The SHED were isolated from fresh dental pulp and were induced to differentiate to neurons and dopamine neurons by inhibiting similar mothers against dpp (SMAD) signaling with Noggin and increase conversion of dopamine neurons from SHED with CHIR99021, Sonic Hedgehog (SHH) and FGF8 in vitro. The neural-primed SHED were transplanted to the striatum of 6-hydroxydopamine (6-OHDA)-induced PD rats to evaluate their neural differentiation and functions in vivo. RESULTS: These SHED were efficiently differentiated to neurons (62.7%) and dopamine neurons (42.3%) through a newly developed method. After transplantation, the neural-induced SHED significantly improved recovery of the motor deficits of the PD rats. The grafted SHED were differentiated into neurons (61%), including dopamine neurons (22.3%), and integrated into the host rat brain by forming synaptic connections. Patch clamp analysis showed that neurons derived from grafted SHED have the same membrane potential profile as dopamine neurons, indicating these cells are dopamine neuron-like cells. The potential molecular mechanism of SHED transplantation in alleviating motor deficits of the rats is likely to be mediated by neuronal replacement and immune-modulation as we detected the transplanted dopamine neurons and released immune cytokines from SHED. CONCLUSION: Using neural-primed SHED to treat PD showed significant restorations of motor deficits in 6-OHDA-induced rats. These observations provide further evidence that SHED can be used for cell-based therapy of PD.


Asunto(s)
Cuerpo Estriado/trasplante , Actividad Motora , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/terapia , Trasplante de Células Madre , Células Madre/citología , Exfoliación Dental/patología , Diente Primario/citología , Animales , Conducta Animal , Diferenciación Celular , Supervivencia Celular , Niño , Preescolar , Citocinas/metabolismo , Neuronas Dopaminérgicas/citología , Humanos , Masculino , Oxidopamina , Ratas Wistar
6.
Proc Natl Acad Sci U S A ; 111(44): 15804-9, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25331880

RESUMEN

Embryonic stem cell-based therapies exhibit great potential for the treatment of Parkinson's disease (PD) because they can significantly rescue PD-like behaviors. However, whether the transplanted cells themselves release dopamine in vivo remains elusive. We and others have recently induced human embryonic stem cells into primitive neural stem cells (pNSCs) that are self-renewable for massive/transplantable production and can efficiently differentiate into dopamine-like neurons (pNSC-DAn) in culture. Here, we showed that after the striatal transplantation of pNSC-DAn, (i) pNSC-DAn retained tyrosine hydroxylase expression and reduced PD-like asymmetric rotation; (ii) depolarization-evoked dopamine release and reuptake were significantly rescued in the striatum both in vitro (brain slices) and in vivo, as determined jointly by microdialysis-based HPLC and electrochemical carbon fiber electrodes; and (iii) the rescued dopamine was released directly from the grafted pNSC-DAn (and not from injured original cells). Thus, pNSC-DAn grafts release and reuptake dopamine in the striatum in vivo and alleviate PD symptoms in rats, providing proof-of-concept for human clinical translation.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Células-Madre Neurales/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Trasplante de Células Madre , Animales , Diferenciación Celular , Cuerpo Estriado/patología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Xenoinjertos , Humanos , Masculino , Células-Madre Neurales/trasplante , Enfermedad de Parkinson/patología , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa/metabolismo
7.
Diabetologia ; 58(2): 324-33, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25381556

RESUMEN

AIMS/HYPOTHESIS: Insulin is a key metabolic regulator in health and diabetes. In pancreatic beta cells, insulin release is regulated by the major second messengers Ca(2+) and cAMP: exocytosis is triggered by Ca(2+) and mediated by the cAMP/protein kinase A (PKA) signalling pathway. However, the causal link between these two processes in primary beta cells remains undefined. METHODS: Time-resolved confocal imaging of fluorescence resonance energy transfer signals was performed to visualise PKA activity, and combined membrane capacitance recordings were used to monitor insulin secretion from patch-clamped rat beta cells. RESULTS: Membrane depolarisation-induced Ca(2+) influx caused an increase in cytosolic PKA activity via activating a Ca(2+)-sensitive adenylyl cyclase 8 (ADCY8) subpool. Glucose stimulation triggered coupled Ca(2+) oscillations and PKA activation. ADCY8 knockdown significantly reduced the level of depolarisation-evoked PKA activation and impaired replenishment of the readily releasable vesicle pool. Pharmacological inhibition of PKA by two inhibitors reduced depolarisation-induced PKA activation to a similar extent and reduced the capacity for sustained vesicle exocytosis and insulin release. CONCLUSIONS/INTERPRETATION: Our findings suggest that depolarisation-induced Ca(2+) influx plays dual roles in regulating exocytosis in rat pancreatic beta cells by triggering vesicle fusion and replenishing the vesicle pool to support sustained insulin release. Therefore, Ca(2+) influx may be important for glucose-stimulated insulin secretion.


Asunto(s)
Adenilil Ciclasas/metabolismo , Calcio/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Transducción de Señal
8.
Analyst ; 140(11): 3840-5, 2015 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-25651802

RESUMEN

Schizophrenia is a severely devastating mental disorder, the pathological process of which is proposed to be associated with the dysfunction of dopaminergic transmission. Our previous results have demonstrated slower kinetics of transmitter release (glutamate release in hippocampus and norepinephrine release in adrenal slice) in a schizophrenia model, dysbindin null-sandy mice. However, whether dopaminergic transmission in the nigrostriatal pathway contributes to the pathology of dysbindin-/- mice remains unknown. Here, we have provided a step-by-step protocol to be applied in the in vivo amperometric recording of dopamine (DA) release from the mouse striatum evoked by an action potential (AP) pattern. With this protocol, AP pattern-dependent DA release was recorded from dysbindin-/- mice striatum in vivo. On combining amperometric recording in slices and electrophysiology, we found that in dysbindin-/- mice, (1) presynaptically, AP-pattern dependent dopamine overflow and uptake were intact in vivo; (2) the recycling of the dopamine vesicle pool remained unchanged. (3) Postsynaptically, the excitability of medium spiny neuron (MSN) was also normal, as revealed by patch-clamp recordings in striatal slices. Taken together, in contrast to reduced norepinephrine release in adrenal chromaffin cells, the dopaminergic transmission remains unchanged in the nigrostriatal pathway in dysbindin-/- mice, providing a new insight into the functions of the schizophrenia susceptibility gene dysbindin.


Asunto(s)
Dopamina/metabolismo , Electroquímica/métodos , Neostriado/metabolismo , Esquizofrenia/metabolismo , Animales , Transporte Biológico , Modelos Animales de Enfermedad , Disbindina , Proteínas Asociadas a la Distrofina/deficiencia , Estimulación Eléctrica , Fenómenos Electrofisiológicos , Ratones , Ratones Endogámicos C57BL , Neostriado/patología , Neostriado/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Esquizofrenia/patología , Esquizofrenia/fisiopatología
9.
J Physiol ; 592(16): 3559-76, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24973407

RESUMEN

Striatal dopamine (DA) is critically involved in major brain functions such as motor control and deficits such as Parkinson's disease. DA is released following stimulation by two pathways: the nigrostriatal pathway and the cholinergic interneuron (ChI) pathway. The timing of synaptic transmission is critical in striatal circuits, because millisecond latency changes can reverse synaptic plasticity from long-term potentiation to long-term depression in a DA-dependent manner. Here, we determined the temporal components of ChI-driven DA release in striatal slices from optogenetic ChAT-ChR2-EYFP mice. After a light stimulus at room temperature, ChIs fired an action potential with a delay of 2.8 ms. The subsequent DA release mediated by nicotinic acetylcholine (ACh) receptors had a total latency of 17.8 ms, comprising 7.0 ms for cholinergic transmission and 10.8 ms for the downstream terminal DA release. Similar latencies of DA release were also found in striatal slices from wild-type mice. The latency of ChI-driven DA release was regulated by inhibiting the presynaptic vesicular ACh release. Moreover, we describe the time course of recovery of DA release via the two pathways and that of vesicle replenishment in DA terminals. Our work provides an example of unravelling the temporal building blocks during fundamental synaptic terminal-terminal transmission in motor regulation.


Asunto(s)
Potenciales de Acción , Neuronas Colinérgicas/metabolismo , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/metabolismo , Tiempo de Reacción , Transmisión Sináptica , Acetilcolina/metabolismo , Animales , Neuronas Colinérgicas/fisiología , Cuerpo Estriado/citología , Cuerpo Estriado/fisiología , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Femenino , Interneuronas/metabolismo , Interneuronas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Nicotínicos/metabolismo , Vesículas Sinápticas/metabolismo
10.
Toxicol Lett ; 397: 129-140, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38759938

RESUMEN

Zinc Oxide nanoparticles (ZnO NPs) have dualistic properties due to their advantage and toxicity. However, the impact and mechanisms of ZnO NPs on the prefrontal lobe have limited research. This study investigates the behavioral changes following exposure to ZnO NPs (34 mg/kg, 30 days), integrating multiple behaviors and bioinformatics analysis to identify critical factors and regulatory mechanisms. The essential differentially expressed genes (DEGs) were identified, including ORC1, DSP, AADAT, SLITRK6, and STEAP1. Analysis of the DEGs based on fold change reveals that ZnO NPs primarily regulate cell survival, proliferation, and apoptosis in neural cells, damaging the prefrontal lobe. Moreover, disruption of cell communication, mineral absorption, and immune pathways occurs. Gene set enrichment analysis (GSEA) further shows enrichment of behavior, neuromuscular process, signal transduction in function, synapses-related, cAMP signaling, and immune pathways. Furthermore, alternative splicing (AS) genes highlight synaptic structure/function, synaptic signal transduction, immune responses, cell proliferation, and communication.


Asunto(s)
Conducta Animal , Corteza Prefrontal , Óxido de Zinc , Animales , Óxido de Zinc/toxicidad , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratones , Conducta Animal/efectos de los fármacos , Masculino , Nanopartículas del Metal/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos
11.
Neurosci Bull ; 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38976218

RESUMEN

Endocytosis is a fundamental biological process that couples exocytosis to maintain the homeostasis of the plasma membrane and sustained neurotransmission. Super-resolution microscopy enables optical imaging of exocytosis and endocytosis in live cells and makes an essential contribution to understanding molecular mechanisms of endocytosis in neuronal somata and other types of cells. However, visualization of exo-endocytic events at the single vesicular level in a synapse with optical imaging remains a great challenge to reveal mechanisms governing the synaptic exo-endocytotic coupling. In this protocol, we describe the technical details of stimulated emission depletion (STED) imaging of synaptic endocytosis at the single-vesicle level, from sample preparation and microscopy calibration to data acquisition and analysis.

12.
Nat Neurosci ; 27(2): 272-285, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38172439

RESUMEN

The central mechanisms underlying pain chronicity remain elusive. Here, we identify a reciprocal neuronal circuit in mice between the anterior cingulate cortex (ACC) and the ventral tegmental area (VTA) that mediates mutual exacerbation between hyperalgesia and allodynia and their emotional consequences and, thereby, the chronicity of neuropathic pain. ACC glutamatergic neurons (ACCGlu) projecting to the VTA indirectly inhibit dopaminergic neurons (VTADA) by activating local GABAergic interneurons (VTAGABA), and this effect is reinforced after nerve injury. VTADA neurons in turn project to the ACC and synapse to the initial ACCGlu neurons to convey feedback information from emotional changes. Thus, an ACCGlu-VTAGABA-VTADA-ACCGlu positive-feedback loop mediates the progression to and maintenance of persistent pain and comorbid anxiodepressive-like behavior. Disruption of this feedback loop relieves hyperalgesia and anxiodepressive-like behavior in a mouse model of neuropathic pain, both acutely and in the long term.


Asunto(s)
Neuralgia , Área Tegmental Ventral , Ratones , Animales , Giro del Cíngulo , Hiperalgesia , Retroalimentación , Neuronas Dopaminérgicas/fisiología , Ácido gamma-Aminobutírico
13.
Sci Data ; 10(1): 577, 2023 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-37666871

RESUMEN

The development of the cardiac conduction system (CCS) is essential for correct heart function. However, critical details on the cell types populating the CCS in the mammalian heart during the development remain to be resolved. Using single-cell RNA sequencing, we generated a large dataset of transcriptomes of ~0.5 million individual cells isolated from murine hearts at six successive developmental corresponding to the early, middle and late stages of heart development. The dataset provides a powerful library for studying the development of the heart's CCS and other cardiac components. Our initial analysis identified distinct cell types between 20 to 26 cell types across different stages, of which ten are involved in forming the CCS. Our dataset allows researchers to reuse the datasets for data mining and a wide range of analyses. Collectively, our data add valuable transcriptomic resources for further study of cardiac development, such as gene expression, transcriptional regulation and functional gene activity in developing hearts, particularly the CCS.


Asunto(s)
Corazón , Análisis de Expresión Génica de una Sola Célula , Animales , Ratones , Minería de Datos , Perfilación de la Expresión Génica , Biblioteca de Genes , Mamíferos , Análisis de Secuencia de ARN
14.
J Neurochem ; 119(2): 342-53, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21854394

RESUMEN

Action potential (AP) patterns and dopamine (DA) release are known to correlate with rewarding behaviors, but how codes of AP bursts translate into DA release in vivo remains elusive. Here, a given AP pattern was defined by four codes, termed total AP number, frequency, number of AP bursts, and interburst time [N, f, b, i].. The 'burst effect' was calculated by the ratio (γ) of DA overflow by multiple bursts to that of a single burst when total AP number was fixed. By stimulating the medial forebrain bundle using AP codes at either physiological (20 Hz) or supraphysiological (80 Hz) frequencies, we found that DA was released from two kinetically distinct vesicle pools, the fast-releasable pool (FRP) and prolonged-releasable pool (PRP), in striatal dopaminergic terminals in vivo. We examined the effects of vesicle pools on AP-pattern dependent DA overflow and found, with given 'burst codes' [b=8, i=0.5 s], a large total AP number [N = 768, f = 80 Hz] produced a facilitating burst-effect (γ[b8/b1] = 126 ± 3%), while a small total AP number [N=96, 80 Hz] triggered a depressing-burst-effect (γ[b8/b1] = 29 ± 4%). Furthermore, we found that the PRP (but not the FRP) predominantly contributed to the facilitating-burst-effect and the FRP played an important role in the depressing-burst effect. Thus, our results suggest that striatal DA release captures pre-synaptic AP pattern information through different releasable pools.


Asunto(s)
Potenciales de Acción/fisiología , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Vesículas Sinápticas/fisiología , Algoritmos , Animales , Simulación por Computador , Estimulación Eléctrica , Electroquímica , Canales Iónicos/efectos de los fármacos , Canales Iónicos/metabolismo , Cinética , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Vesículas Sinápticas/metabolismo
15.
Neuroscientist ; 26(1): 9-15, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31046622

RESUMEN

Synaptotagmins (Syts) are well-established primary Ca2+ sensors to initiate presynaptic neurotransmitter release. They also play critical roles in the docking, priming, and fusion steps of exocytosis, as well as the tightly coupled exo-endocytosis, in presynapses. A recent study by Awasthi and others (2019) shows that Syt3 Ca2+-dependently modulates the postsynaptic receptor endocytosis and thereby promotes the long-term depression (LTD) and the decay of long-term potentiation (LTP). This work highlights the importance of Syt3 in modulating long-term synaptic plasticity and, importantly, extends the function of Syt proteins from presynaptic neurotransmitter release to a new promising postsynaptic receptor internalization.


Asunto(s)
Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Sinaptotagminas/metabolismo , Animales , Exocitosis/fisiología , Humanos , Plasticidad Neuronal
16.
Front Neurosci ; 13: 28, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30745863

RESUMEN

α-synuclein (α-Syn) is a presynaptic enriched protein involved in the pathogenesis of Parkinson's disease. However, the physiological roles of α-Syn remain poorly understood. Recent studies have indicated a critical role of α-Syn in the sensing and generation of membrane curvature during vesicular exocytosis and endocytosis. It has been known to modulate the assembly of SNARE complex during exocytosis including vesicle docking, priming and fusion steps. Growing evidence suggests that α-Syn also plays critical roles in the endocytosis of synaptic vesicles. It also modulates the availability of releasable vesicles by promoting synaptic vesicles clustering. Here, we provide an overview of recent progresses in understanding the function of α-Syn in the regulation of exocytosis, endocytosis, and vesicle recycling under physiological and pathological conditions.

17.
Neuron ; 102(1): 173-183.e4, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30773347

RESUMEN

Co-release of multiple neurotransmitters from secretory vesicles is common in neurons and neuroendocrine cells. However, whether and how the transmitters co-released from a single vesicle are differentially regulated remains unknown. In matrix-containing dense-core vesicles (DCVs) in chromaffin cells, there are two modes of catecholamine (CA) release from a single DCV: quantal and sub-quantal. By combining two microelectrodes to simultaneously record co-release of the native CA and ATP from a DCV, we report that (1) CA and ATP were co-released during a DCV fusion; (2) during kiss-and-run (KAR) fusion, the co-released CA was sub-quantal, whereas the co-released ATP was quantal; and (3) knockdown and knockout of the DCV matrix led to quantal co-release of both CA and ATP even in KAR mode. These findings strongly imply that, in contrast to sub-quantal CA release in chromaffin cells, fast synaptic transmission without transmitter-matrix binding is mediated exclusively via quantal release in neurons.


Asunto(s)
Adenosina Trifosfato/metabolismo , Catecolaminas/metabolismo , Células Cromafines/metabolismo , Exocitosis/fisiología , Vesículas Secretoras/metabolismo , Transmisión Sináptica/fisiología , Médula Suprarrenal/citología , Animales , Calcio/metabolismo , Señalización del Calcio , Células HEK293 , Humanos , Fusión de Membrana , Ratones , Ratones Noqueados , Neurotransmisores/metabolismo , Técnicas de Placa-Clamp , Sinaptotagminas/genética
18.
Nat Commun ; 9(1): 81, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29311685

RESUMEN

Loss-of-function mutations in Parkin are the most common causes of autosomal recessive Parkinson's disease (PD). Many putative substrates of parkin have been reported; their pathogenic roles, however, remain obscure due to poor characterization, particularly in vivo. Here, we show that synaptotagmin-11, encoded by a PD-risk gene SYT11, is a physiological substrate of parkin and plays critical roles in mediating parkin-linked neurotoxicity. Unilateral overexpression of full-length, but not C2B-truncated, synaptotagmin-11 in the substantia nigra pars compacta (SNpc) impairs ipsilateral striatal dopamine release, causes late-onset degeneration of dopaminergic neurons, and induces progressive contralateral motor abnormalities. Mechanistically, synaptotagmin-11 impairs vesicle pool replenishment and thus dopamine release by inhibiting endocytosis. Furthermore, parkin deficiency induces synaptotagmin-11 accumulation and PD-like neurotoxicity in mouse models, which is reversed by SYT11 knockdown in the SNpc or knockout of SYT11 restricted to dopaminergic neurons. Thus, PD-like neurotoxicity induced by parkin dysfunction requires synaptotagmin-11 accumulation in SNpc dopaminergic neurons.


Asunto(s)
Enfermedad de Parkinson/patología , Sinaptotagminas/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Conducta Animal , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Endocitosis/fisiología , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Nanopartículas , Enfermedad de Parkinson/metabolismo , Ratas , Ratas Wistar , Sustancia Negra/metabolismo , Sustancia Negra/patología , Especificidad por Sustrato , Sinaptotagminas/genética , Sinaptotagminas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
20.
Front Mol Neurosci ; 10: 47, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28348516

RESUMEN

Neuronal communication and brain function mainly depend on the fundamental biological events of neurotransmission, including the exocytosis of presynaptic vesicles (SVs) for neurotransmitter release and the subsequent endocytosis for SV retrieval. Neurotransmitters are released through the Ca2+- and SNARE-dependent fusion of SVs with the presynaptic plasma membrane. Following exocytosis, endocytosis occurs immediately to retrieve SV membrane and fusion machinery for local recycling and thus maintain the homeostasis of synaptic structure and sustained neurotransmission. Apart from the general endocytic machinery, recent studies have also revealed the involvement of SNARE proteins (synaptobrevin, SNAP25 and syntaxin), synaptophysin, Ca2+/calmodulin, and members of the synaptotagmin protein family (Syt1, Syt4, Syt7 and Syt11) in the balance and tight coupling of exo-endocytosis in neurons. Here, we provide an overview of recent progress in understanding how these neuron-specific adaptors coordinate to ensure precise and efficient endocytosis during neurotransmission.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA