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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(21): e2404763121, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38743626

RESUMO

Congenital stationary night blindness (CSNB) is an inherited retinal disease that causes a profound loss of rod sensitivity without severe retinal degeneration. One well-studied rhodopsin point mutant, G90D-Rho, is thought to cause CSNB because of its constitutive activity in darkness causing rod desensitization. However, the nature of this constitutive activity and its precise molecular source have not been resolved for almost 30 y. In this study, we made a knock-in (KI) mouse line with a very low expression of G90D-Rho (equal in amount to ~0.1% of normal rhodopsin, WT-Rho, in WT rods), with the remaining WT-Rho replaced by REY-Rho, a mutant with a very low efficiency of activating transducin due to a charge reversal of the highly conserved ERY motif to REY. We observed two kinds of constitutive noise: one being spontaneous isomerization (R*) of G90D-Rho at a molecular rate (R* s-1) 175-fold higher than WT-Rho and the other being G90D-Rho-generated dark continuous noise comprising low-amplitude unitary events occurring at a very high molecular rate equivalent in effect to ~40,000-fold of R* s-1 from WT-Rho. Neither noise type originated from G90D-Opsin because exogenous 11-cis-retinal had no effect. Extrapolating the above observations at low (0.1%) expression of G90D-Rho to normal disease exhibited by a KI mouse model with RhoG90D/WTand RhoG90D/G90D genotypes predicts the disease condition very well quantitatively. Overall, the continuous noise from G90D-Rho therefore predominates, constituting the major equivalent background light causing rod desensitization in CSNB.


Assuntos
Oftalmopatias Hereditárias , Doenças Genéticas Ligadas ao Cromossomo X , Miopia , Cegueira Noturna , Rodopsina , Animais , Cegueira Noturna/genética , Cegueira Noturna/metabolismo , Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/metabolismo , Camundongos , Rodopsina/genética , Rodopsina/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Miopia/genética , Miopia/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Escuridão , Transducina/genética , Transducina/metabolismo , Técnicas de Introdução de Genes , Modelos Animais de Doenças
2.
Proc Natl Acad Sci U S A ; 119(20): e2111051119, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35537054

RESUMO

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.


Assuntos
Endocitose , Transmissão Sináptica , Sinaptotagmina I , Cálcio/metabolismo , Endocitose/fisiologia , Exocitose/fisiologia , Neurônios/metabolismo , Sinaptotagmina I/metabolismo
3.
EMBO Rep ; 23(11): e54507, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36148511

RESUMO

A central principle of synaptic transmission is that action potential-induced presynaptic neurotransmitter release occurs exclusively via Ca2+ -dependent secretion (CDS). The discovery and mechanistic investigations of Ca2+ -independent but voltage-dependent secretion (CiVDS) have demonstrated that the action potential per se is sufficient to trigger neurotransmission in the somata of primary sensory and sympathetic neurons in mammals. One key question remains, however, whether CiVDS contributes to central synaptic transmission. Here, we report, in the central transmission from presynaptic (dorsal root ganglion) to postsynaptic (spinal dorsal horn) neurons in vitro, (i) excitatory postsynaptic currents (EPSCs) are mediated by glutamate transmission through both CiVDS (up to 87%) and CDS; (ii) CiVDS-mediated EPSCs are independent of extracellular and intracellular Ca2+ ; (iii) CiVDS is faster than CDS in vesicle recycling with much less short-term depression; (iv) the fusion machinery of CiVDS includes Cav2.2 (voltage sensor) and SNARE (fusion pore). Together, an essential component of activity-induced EPSCs is mediated by CiVDS in a central synapse.


Assuntos
Gânglios Espinais , Células do Corno Posterior , Animais , Células do Corno Posterior/fisiologia , Transmissão Sináptica/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Sinapses , Mamíferos
4.
Proc Natl Acad Sci U S A ; 117(37): 23033-23043, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32873651

RESUMO

Numerous rhodopsin mutations have been implicated in night blindness and retinal degeneration, often with unclear etiology. D190N-rhodopsin (D190N-Rho) is a well-known inherited human mutation causing retinitis pigmentosa. Both higher-than-normal spontaneous-isomerization activity and misfolding/mistargeting of the mutant protein have been proposed as causes of the disease, but neither explanation has been thoroughly examined. We replaced wild-type rhodopsin (WT-Rho) in RhoD190N/WT mouse rods with a largely "functionally silenced" rhodopsin mutant to isolate electrical responses triggered by D190N-Rho activity, and found that D190N-Rho at the single-molecule level indeed isomerizes more frequently than WT-Rho by over an order of magnitude. Importantly, however, this higher molecular dark activity does not translate into an overall higher cellular dark noise, owing to diminished D190N-Rho content in the rod outer segment. Separately, we found that much of the degeneration and shortened outer-segment length of RhoD190N/WT mouse rods was not averted by ablating rod transducin in phototransduction-also consistent with D190N-Rho's higher isomerization activity not being the primary cause of disease. Instead, the low pigment content, shortened outer-segment length, and a moderate unfolded protein response implicate protein misfolding as the major pathogenic problem. Finally, D190N-Rho also provided some insight into the mechanism of spontaneous pigment excitation.


Assuntos
Degeneração Retiniana/metabolismo , Rodopsina/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Células HEK293 , Humanos , Transdução de Sinal Luminoso/fisiologia , Camundongos , Mutação/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Retinose Pigmentar/metabolismo , Segmento Externo da Célula Bastonete/metabolismo
5.
Proc Natl Acad Sci U S A ; 116(40): 20201-20209, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31530723

RESUMO

Action potential-induced vesicular exocytosis is considered exclusively Ca2+ dependent in Katz's Ca2+ hypothesis on synaptic transmission. This long-standing concept gets an exception following the discovery of Ca2+-independent but voltage-dependent secretion (CiVDS) and its molecular mechanisms in dorsal root ganglion sensory neurons. However, whether CiVDS presents only in sensory cells remains elusive. Here, by combining multiple independent recordings, we report that [1] CiVDS robustly presents in the sympathetic nervous system, including sympathetic superior cervical ganglion neurons and slice adrenal chromaffin cells, [2] uses voltage sensors of Ca2+ channels (N-type and novel L-type), and [3] contributes to catecholamine release in both homeostatic and fight-or-flight like states; [4] CiVDS-mediated catecholamine release is faster than that of Ca2+-dependent secretion at the quantal level and [5] increases Ca2+ currents and contractility of cardiac myocytes. Together, CiVDS presents in the sympathetic nervous system with potential physiological functions, including cardiac muscle contractility.


Assuntos
Cálcio/metabolismo , Catecolaminas/metabolismo , Células Cromafins/metabolismo , Sistema Nervoso Simpático/metabolismo , Potenciais de Ação , Animais , Mamíferos , Modelos Biológicos , Células Musculares/metabolismo , Neurônios/metabolismo , Corno Dorsal da Medula Espinal/citologia , Corno Dorsal da Medula Espinal/metabolismo , Transmissão Sináptica
6.
Brain ; 142(10): 3099-3115, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31504219

RESUMO

The loss-of-function mutation in PARK7/DJ-1 is one of the most common causes of autosomal recessive Parkinson's disease, and patients carrying PARK7 mutations often exhibit both a progressive movement disorder and emotional impairment, such as anxiety. However, the causes of the emotional symptom accompanying PARK7-associated and other forms of Parkinson's disease remain largely unexplored. Using two-photon microscopic Ca2+ imaging in awake PARK7-/- and PARK7+/+ mice, we found that (i) PARK7-/- neurons in the frontal association cortex showed substantially higher circuit activity recorded as spontaneous somatic Ca2+ signals; (ii) both basal and evoked dopamine release remained intact, as determined by both electrochemical dopamine recordings and high performance liquid chromatography in vivo; (iii) D2 receptor expression was significantly decreased in postsynaptic frontal association cortical neurons, and the hyper-neuronal activity were rescued by D2 receptor intervention using either local pharmacology or viral D2 receptor over-expression; and (iv) PARK7-/- mice showed anxiety-like behaviours that were rescued by either local D2 receptor pharmacology or overexpression. Thus, for first time, we demonstrated a robust D2 receptor-dependent phenotype of individual neurons within the prefrontal cortex circuit in awake parkinsonian mice that linked with anxiety. Our work sheds light on early-onset phenotypes and the mechanisms underlying Parkinson's disease by imaging brain circuits in an awake mouse model.


Assuntos
Doença de Parkinson/genética , Doença de Parkinson/fisiopatologia , Proteína Desglicase DJ-1/genética , Animais , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Feminino , Humanos , Masculino , Camundongos , Doença de Parkinson/metabolismo , Transtornos Parkinsonianos/metabolismo , Córtex Pré-Frontal/metabolismo , Proteína Desglicase DJ-1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Substância Negra/metabolismo , Vigília
7.
EMBO Rep ; 17(1): 47-63, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26589353

RESUMO

Precise and efficient endocytosis is essential for vesicle recycling during a sustained neurotransmission. The regulation of endocytosis has been extensively studied, but inhibitors have rarely been found. Here, we show that synaptotagmin-11 (Syt11), a non-Ca(2+)-binding Syt implicated in schizophrenia and Parkinson's disease, inhibits clathrin-mediated endocytosis (CME) and bulk endocytosis in dorsal root ganglion neurons. The frequency of both types of endocytic event increases in Syt11 knockdown neurons, while the sizes of endocytosed vesicles and the kinetics of individual bulk endocytotic events remain unaffected. Specifically, clathrin-coated pits and bulk endocytosis-like structures increase on the plasma membrane in Syt11-knockdown neurons. Structural-functional analysis reveals distinct domain requirements for Syt11 function in CME and bulk endocytosis. Importantly, Syt11 also inhibits endocytosis in hippocampal neurons, implying a general role of Syt11 in neurons. Taken together, we propose that Syt11 functions to ensure precision in vesicle retrieval, mainly by limiting the sites of membrane invagination at the early stage of endocytosis.


Assuntos
Vesículas Revestidas por Clatrina/fisiologia , Clatrina/metabolismo , Endocitose , Neurônios/metabolismo , Sinaptotagminas/genética , Sinaptotagminas/metabolismo , Animais , Exocitose , Gânglios Espinais/citologia , Técnicas de Silenciamento de Genes , Neurônios/ultraestrutura , Ratos , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia
8.
Am J Hum Genet ; 93(5): 957-66, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24207120

RESUMO

Many ion channel genes have been associated with human genetic pain disorders. Here we report two large Chinese families with autosomal-dominant episodic pain. We performed a genome-wide linkage scan with microsatellite markers after excluding mutations in three known genes (SCN9A, SCN10A, and TRPA1) that cause similar pain syndrome to our findings, and we mapped the genetic locus to a 7.81 Mb region on chromosome 3p22.3-p21.32. By using whole-exome sequencing followed by conventional Sanger sequencing, we identified two missense mutations in the gene encoding voltage-gated sodium channel Nav1.9 (SCN11A): c.673C>T (p.Arg225Cys) and c.2423C>G (p.Ala808Gly) (one in each family). Each mutation showed a perfect cosegregation with the pain phenotype in the corresponding family, and neither of them was detected in 1,021 normal individuals. Both missense mutations were predicted to change a highly conserved amino acid residue of the human Nav1.9 channel. We expressed the two SCN11A mutants in mouse dorsal root ganglion (DRG) neurons and showed that both mutations enhanced the channel's electrical activities and induced hyperexcitablity of DRG neurons. Taken together, our results suggest that gain-of-function mutations in SCN11A can be causative of an autosomal-dominant episodic pain disorder.


Assuntos
Dor/genética , Animais , Povo Asiático/genética , Canais de Cálcio/genética , Feminino , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Ligação Genética , Marcadores Genéticos , Humanos , Masculino , Camundongos , Repetições de Microssatélites , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.9/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Neurônios/patologia , Dor/patologia , Linhagem , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/genética
9.
Nat Neurosci ; 27(2): 272-285, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38172439

RESUMO

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.


Assuntos
Neuralgia , Área Tegmentar Ventral , Camundongos , Animais , Giro do Cíngulo , Hiperalgesia , Retroalimentação , Neurônios Dopaminérgicos/fisiologia , Ácido gama-Aminobutírico
10.
Sci Adv ; 6(51)2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33328242

RESUMO

Almost a century ago, Stiles and Crawford reported that the human eye is more sensitive to light entering through the pupil center than through its periphery (Stiles-Crawford effect). This psychophysical phenomenon, later found to correlate with photoreceptor orientation toward the pupil, was dynamically phototropic, adjustable within days to an eccentrically displaced pupil. For decades, this phototropism has been speculated to involve coordinated movements of the rectilinear photoreceptor outer and inner segments. We report here that, unexpectedly, the murine photoreceptor outer segment has a seemingly light-independent orientation, but the inner segment's orientation undergoes light-dependent movement, giving rise to nonrectilinear outer and inner segments in adult mice born and reared in darkness. Light during an early critical period (~P0 to P8), however, largely sets the correct photoreceptor orientation permanently afterward. Unexpectedly, abolishing rod and cone phototransductions did not mimic darkness in early life, suggesting photosignaling extrinsic to rods and cones is involved.

11.
Neuron ; 102(1): 173-183.e4, 2019 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-30773347

RESUMO

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.


Assuntos
Trifosfato de Adenosina/metabolismo , Catecolaminas/metabolismo , Células Cromafins/metabolismo , Exocitose/fisiologia , Vesículas Secretórias/metabolismo , Transmissão Sináptica/fisiologia , Medula Suprarrenal/citologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Células HEK293 , Humanos , Fusão de Membrana , Camundongos , Camundongos Knockout , Neurotransmissores/metabolismo , Técnicas de Patch-Clamp , Sinaptotagminas/genética
12.
Nat Commun ; 9(1): 81, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311685

RESUMO

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.


Assuntos
Doença de Parkinson/patologia , Sinaptotagminas/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Comportamento Animal , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Endocitose/fisiologia , Feminino , Predisposição Genética para Doença , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Nanopartículas , Doença de Parkinson/metabolismo , Ratos , Ratos Wistar , Substância Negra/metabolismo , Substância Negra/patologia , Especificidade por Substrato , Sinaptotagminas/genética , Sinaptotagminas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
13.
Front Mol Neurosci ; 10: 47, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28348516

RESUMO

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.

14.
Sci Signal ; 10(484)2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28634208

RESUMO

Neuropeptides released from dorsal root ganglion (DRG) neurons play essential roles in the neurotransmission of sensory inputs, including those underlying nociception and pathological pain. Neuropeptides are released from intracellular vesicles through two modes: a partial release mode called "kiss-and-run" (KAR) and a full release mode called "full fusion-like" (FFL). Using total internal reflection fluorescence (TIRF) microscopy, we traced the release of pH-sensitive green fluorescent protein-tagged neuropeptide Y (pHluorin-NPY) from individual dense-core vesicles in the soma and axon of single DRG neurons after Ca2+ influx through either voltage-gated Ca2+ channels (VGCCs) or ligand-gated transient receptor potential vanilloid 1 (TRPV1) channels. We found that Ca2+ influx through VGCCs stimulated FFL and a greater single release of neuropeptides. In contrast, Ca2+ influx through TRPV1 channels stimulated KAR and a pulsed but prolonged release of neuropeptides that was partially mediated by Dynamin 1, which limits fusion pore expansion. Suppressing the Ca2+ gradient to an extent similar to that seen after TRPV1 activation abolished the VGCC preference for FFL. The findings suggest that by generating a steeper Ca2+ gradient, VGCCs promote a more robust fusion pore opening that facilitates FFL. Thus, KAR and FFL release modes are differentially regulated by the two principal types of Ca2+-permeable channels in DRG neurons.


Assuntos
Canais de Cálcio/metabolismo , Gânglios Espinais/citologia , Neuropeptídeos/metabolismo , Células Receptoras Sensoriais/citologia , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Dinamina I/metabolismo , Exocitose , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Membranas Sinápticas/metabolismo , Transmissão Sináptica
15.
Neuron ; 96(6): 1317-1326.e4, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-29198756

RESUMO

Action potential induces membrane depolarization and triggers intracellular free Ca2+ concentration (Ca2+)-dependent secretion (CDS) via Ca2+ influx through voltage-gated Ca2+ channels. We report a new type of somatic exocytosis triggered by the action potential per se-Ca2+-independent but voltage-dependent secretion (CiVDS)-in dorsal root ganglion neurons. Here we uncovered the molecular mechanism of CiVDS, comprising a voltage sensor, fusion machinery, and their linker. Specifically, the voltage-gated N-type Ca2+ channel (CaV2.2) is the voltage sensor triggering CiVDS, the SNARE complex functions as the vesicle fusion machinery, the "synprint" of CaV2.2 serves as a linker between the voltage sensor and the fusion machinery, and ATP is a cargo of CiVDS vesicles. Thus, CiVDS releases ATP from the soma while CDS releases glutamate from presynaptic terminals, establishing the CaV2.2-SNARE "voltage-gating fusion pore" as a novel pathway co-existing with the canonical "Ca2+-gating fusion pore" pathway for neurotransmitter release following action potentials in primary sensory neurons.


Assuntos
Canais de Cálcio Tipo N/genética , Canais de Cálcio Tipo N/metabolismo , Cálcio/metabolismo , Ativação do Canal Iônico/genética , Células Receptoras Sensoriais/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Cafeína/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Células Cultivadas , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/ultraestrutura , Exocitose/efeitos dos fármacos , Exocitose/genética , Gânglios Espinais/citologia , Gânglios Espinais/ultraestrutura , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Fusão de Membrana/efeitos dos fármacos , Fusão de Membrana/genética , Modelos Moleculares , Inibidores de Fosfodiesterase/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/fisiologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Células Receptoras Sensoriais/ultraestrutura , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Transdução Genética , ômega-Conotoxina GVIA/farmacologia
16.
J Cell Biol ; 215(3): 369-381, 2016 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-27799370

RESUMO

Transient receptor potential A1 (TRPA1) is a nonselective cation channel implicated in thermosensation and inflammatory pain. In this study, we show that TRPA1 (activated by allyl isothiocyanate, acrolein, and 4-hydroxynonenal) elevates the intracellular Ca2+ concentration ([Ca2+]i) in dorsal root ganglion (DRG) neurons in the presence and absence of extracellular Ca2+ Pharmacological and immunocytochemical analyses revealed the presence of TRPA1 channels both on the plasma membrane and in endolysosomes. Confocal line-scan imaging demonstrated Ca2+ signals elicited from individual endolysosomes ("lysosome Ca2+ sparks") by TRPA1 activation. In physiological solutions, the TRPA1-mediated endolysosomal Ca2+ release contributed to ∼40% of the overall [Ca2+]i rise and directly triggered vesicle exocytosis and calcitonin gene-related peptide release, which greatly enhanced the excitability of DRG neurons. Thus, in addition to working via Ca2+ influx, TRPA1 channels trigger vesicle release in sensory neurons by releasing Ca2+ from lysosome-like organelles.


Assuntos
Cálcio/metabolismo , Gânglios Espinais/metabolismo , Espaço Intracelular/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Acroleína , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Sinalização do Cálcio , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Exocitose , Hiperalgesia/metabolismo , Ativação do Canal Iônico , Isotiocianatos , Masculino , Camundongos Endogâmicos C57BL , Neuropeptídeos/metabolismo , Soluções , Canal de Cátion TRPA1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA