Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 42(33): 6325-6343, 2022 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-35803735

RESUMO

It is a commonly accepted view that light stimulation of mammalian photoreceptors causes a graded change in membrane potential instead of developing a spike. The presynaptic Ca2+ channels serve as a crucial link for the coding of membrane potential variations into neurotransmitter release. Cav1.4 L-type Ca2+ channels are expressed in photoreceptor terminals, but the complete pool of Ca2+ channels in cone photoreceptors appears to be more diverse. Here, we discovered, employing whole-cell patch-clamp recording from cone photoreceptor terminals in both sexes of mice, that their Ca2+ currents are composed of low- (T-type Ca2+ channels) and high- (L-type Ca2+ channels) voltage-activated components. Furthermore, Ca2+ channels exerted self-generated spike behavior in dark membrane potentials, and spikes were generated in response to light/dark transition. The application of fast and slow Ca2+ chelators revealed that T-type Ca2+ channels are located close to the release machinery. Furthermore, capacitance measurements indicated that they are involved in evoked vesicle release. Additionally, RT-PCR experiments showed the presence of Cav3.2 T-type Ca2+ channels in cone photoreceptors but not in rod photoreceptors. Altogether, we found several crucial functions of T-type Ca2+ channels, which increase the functional repertoire of cone photoreceptors. Namely, they extend cone photoreceptor light-responsive membrane potential range, amplify dark responses, generate spikes, increase intracellular Ca2+ levels, and boost synaptic transmission.SIGNIFICANCE STATEMENT Photoreceptors provide the first synapse for coding light information. The key elements in synaptic transmission are the voltage-sensitive Ca2+ channels. Here, we provide evidence that mouse cone photoreceptors express low-voltage-activated Cav3.2 T-type Ca2+ channels in addition to high-voltage-activated L-type Ca2+ channels. The presence of T-type Ca2+ channels in cone photoreceptors appears to extend their light-responsive membrane potential range, amplify dark response, generate spikes, increase intracellular Ca2+ levels, and boost synaptic transmission. By these functions, Cav3.2 T-type Ca2+ channels increase the functional repertoire of cone photoreceptors.


Assuntos
Células Fotorreceptoras Retinianas Cones , Transmissão Sináptica , Animais , Cálcio , Feminino , Masculino , Mamíferos , Camundongos , Técnicas de Patch-Clamp , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia
2.
FASEB J ; 35(5): e21520, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33811381

RESUMO

Bassoon (BSN) is a presynaptic cytomatrix protein ubiquitously present at chemical synapses of the central nervous system, where it regulates synaptic vesicle replenishment and organizes voltage-gated Ca2+ channels. In sensory photoreceptor synapses, BSN additionally plays a decisive role in anchoring the synaptic ribbon, a presynaptic organelle and functional extension of the active zone, to the presynaptic membrane. In this study, we functionally and structurally analyzed two mutant mouse lines with a genetic disruption of Bsn-Bsngt and Bsnko -using electrophysiology and high-resolution microscopy. In both Bsn mutant mouse lines, full-length BSN was abolished, and photoreceptor synaptic function was similarly impaired, yet synapse structure was more severely affected in Bsngt/gt than in Bsnko/ko photoreceptors. The synaptic defects in Bsngt/gt retina coincide with remodeling of the outer retina-rod bipolar and horizontal cell sprouting, formation of ectopic ribbon synaptic sites-and death of cone photoreceptors, processes that did not occur in Bsnko/ko retina. An analysis of Bsngt/ko hybrid mice revealed that the divergent retinal phenotypes of Bsngt/gt and Bsnko/ko mice can be attributed to the expression of the Bsngt allele, which triggers cone photoreceptor death and neurite sprouting in the outer retina. These findings shed new light on the existing Bsn mutant mouse models and might help to understand mechanisms that drive photoreceptor death.


Assuntos
Modelos Animais de Doenças , Mutação , Proteínas do Tecido Nervoso/fisiologia , Retina/patologia , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Sinapses/patologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Sinapses/metabolismo , Transmissão Sináptica
3.
Exp Eye Res ; 195: 108028, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32277973

RESUMO

Expression patterns of voltage-gated ion channels determine the spatio-temporal dynamics of ion currents that supply excitable neurons in developing tissue with proper electrophysiological properties. The purpose of the study was to identify fast cationic inward currents in mouse retinal horizontal cells (HCs) and describe their biophysical properties at different developmental stages. We also aimed to reveal their physiological role in shaping light responses (LRs) in adult HCs. HCs were recorded in horizontal slices of wild-type mouse retina at postnatal stages ranging from p8 through p60. Voltage-dependent inward currents were isolated with appropriate voltage protocols and blockers specific for sodium and T-type calcium channels. LRs were evoked with full-field flashes (130 µW/cm2). Transient and steady inward currents were identified at all developmental stages. Transient currents were mediated by T-type calcium and TTX-sensitive sodium channels, whereas steady currents were blocked by cadmium, indicating the presence of high voltage-activated calcium channels. Activation and steady-state inactivation kinetics of T-type calcium channels revealed a contribution to the resting membrane potential during postnatal development. Additionally, both sodium and T-type calcium channels had an impact on HC LRs at light offset in adult animals. Our results showed that the voltage-dependent inward currents of postnatally developing mouse HCs consist of T-type calcium, TTX-sensitive sodium, and high voltage-activated calcium channels, and that transient ionic currents contributed to light-evoked responses of adult HCs, suggesting a role in HC information processing.


Assuntos
Canais de Cálcio/metabolismo , Potenciais da Membrana/fisiologia , Células Horizontais da Retina/metabolismo , Canais de Sódio/metabolismo , Animais , Canais de Cálcio/efeitos dos fármacos , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Técnicas de Patch-Clamp , Células Horizontais da Retina/citologia , Células Horizontais da Retina/efeitos dos fármacos , Canais de Sódio/efeitos dos fármacos , Tetrodotoxina/farmacologia
4.
J Neurosci ; 37(17): 4618-4634, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28363980

RESUMO

G-protein ßγ subunits (Gßγ) interact with presynaptic proteins and regulate neurotransmitter release downstream of Ca2+ influx. To accomplish their roles in sensory signaling, photoreceptor synapses use specialized presynaptic proteins that support neurotransmission at active zone structures known as ribbons. While several G-protein coupled receptors (GPCRs) influence synaptic transmission at ribbon synapses of cones and other retinal neurons, it is unknown whether Gßγ contributes to these effects. We tested whether activation of one particular GPCR, a metabotropic glutamate receptor (mGluR), can reduce cone synaptic transmission via Gßγ in tiger salamander retinas. In recordings from horizontal cells, we found that an mGluR agonist (L-AP4) reduced cone-driven light responses and mEPSC frequency. In paired recordings of cones and horizontal cells, L-AP4 slightly reduced cone ICa (∼10%) and caused a larger reduction in cone-driven EPSCs (∼30%). Proximity ligation assay revealed direct interactions between SNAP-25 and Gßγ subunits in retinal synaptic layers. Pretreatment with the SNAP-25 cleaving protease BoNT/A inhibited L-AP4 effects on synaptic transmission, as did introduction of a peptide derived from the SNAP-25 C terminus. Introducing Gßγ subunits directly into cones reduced EPSC amplitude. This effect was inhibited by BoNT/A, supporting a role for Gßγ/SNAP-25 interactions. However, the mGluR-dependent reduction in ICa was not mimicked by Gßγ, indicating that this effect was independent of Gßγ. The finding that synaptic transmission at cone ribbon synapses is regulated by Gßγ/SNAP-25 interactions indicates that these mechanisms are shared by conventional and ribbon-type synapses. Gßγ liberated from other photoreceptor GPCRs is also likely to regulate synaptic transmission.SIGNIFICANCE STATEMENT Dynamic regulation of synaptic transmission by presynaptic G-protein coupled receptors shapes information flow through neural circuits. At the first synapse in the visual system, presynaptic metabotropic glutamate receptors (mGluRs) regulate cone photoreceptor synaptic transmission, although the mechanisms and functional impact of this are unclear. We show that mGluRs regulate light response encoding across the cone synapse, accomplished in part by triggering G-protein ßγ subunits (Gßγ) interactions with SNAP-25, a core component of the synaptic vesicle fusion machinery. In addition to revealing a role in visual processing, this provides the first demonstration that Gßγ/SNAP-25 interactions regulate synaptic function at a ribbon-type synapse, contributing to an emerging picture of the ubiquity of Gßγ/SNARE interactions in regulating synaptic transmission throughout the nervous system.


Assuntos
Ambystoma/fisiologia , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Células Fotorreceptoras Retinianas Cones/fisiologia , Proteínas SNARE/metabolismo , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Células Horizontais da Retina/metabolismo , Células Horizontais da Retina/fisiologia
5.
J Neurosci ; 37(33): 7848-7863, 2017 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-28701482

RESUMO

RAB3A-interacting molecule (RIM) proteins are important regulators of transmitter release from active zones. At conventional chemical synapses, RIMs contribute substantially to vesicle priming and docking and their loss reduces the readily releasable pool of synaptic vesicles by up to 75%. The priming function of RIMs is mediated via the formation of a tripartite complex with Munc13 and RAB3A, which brings synaptic vesicles in close proximity to Ca2+ channels and the fusion site and activates Munc13. We reported previously that, at mouse photoreceptor ribbon synapses, vesicle priming is Munc13 independent. In this study, we examined RIM expression, distribution, and function at male and female mouse photoreceptor ribbon synapses. We provide evidence that RIM1α and RIM1ß are highly likely absent from mouse photoreceptors and that RIM2α is the major large RIM isoform present at photoreceptor ribbon synapses. We show that mouse photoreceptors predominantly express RIM2 variants that lack the interaction domain for Munc13. Loss of full-length RIM2α in a RIM2α mutant mouse only marginally perturbs photoreceptor synaptic transmission. Our findings therefore strongly argue for a priming mechanism at the photoreceptor ribbon synapse that is independent of the formation of a RIM-Munc13-RAB3A complex and thus provide further evidence for a fundamental difference between photoreceptor ribbon synapses and conventional chemical synapses in synaptic vesicle exocytosis.SIGNIFICANCE STATEMENT RAB3A-interacting molecules 1 and 2 (RIM1/2) are essential regulators of exocytosis. At conventional chemical synapses, their function involves Ca2+ channel clustering and synaptic vesicle priming and docking through interactions with Munc13 and RAB3A, respectively. Examining wild-type and RIM2 mutant mice, we show here that the sensory photoreceptor ribbon synapses most likely lack RIM1 and predominantly express RIM2 variants that lack the interaction domain for Munc13. Our findings demonstrate that the photoreceptor-specific RIM variants are not essential for synaptic vesicle priming at photoreceptor ribbon synapses, which represents a fundamental difference between photoreceptor ribbon synapses and conventional chemical synapses with respect to synaptic vesicle priming mechanisms.


Assuntos
Proteínas de Ligação ao GTP/biossíntese , Células Fotorreceptoras de Vertebrados/metabolismo , Sinapses/metabolismo , Animais , Células Cultivadas , Feminino , Proteínas de Ligação ao GTP/análise , Proteínas de Ligação ao GTP/genética , Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células NIH 3T3 , Células Fotorreceptoras de Vertebrados/química , Sinapses/química , Sinapses/genética
6.
J Neurosci ; 36(25): 6651-67, 2016 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-27335398

RESUMO

UNLABELLED: Complexins (Cplxs) are SNARE complex regulators controlling the speed and Ca(2+) sensitivity of SNARE-mediated synaptic vesicle fusion. We have shown previously that photoreceptor ribbon synapses in mouse retina are equipped with Cplx3 and Cplx4 and that lack of both Cplxs perturbs photoreceptor ribbon synaptic function; however, Cplx3/4 function in photoreceptor synaptic transmission remained elusive. To investigate Cplx3/4 function in photoreceptor ribbon synapses, voltage-clamp recordings from postsynaptic horizontal cells were performed in horizontal slice preparations of Cplx3/4 wild-type (WT) and Cplx3/4 double knock-out (DKO) mice. We measured tonic activity in light and dark, current responses to changes in luminous intensity, and electrically evoked postsynaptic responses. Cplx3/4 decreased the frequency of tonic events and shifted their amplitude distribution to smaller values. Light responses were sustained in the presence of Cplx3/4, but transient in their absence. Finally, Cplx3/4 increased synaptic vesicle release evoked by electrical stimulation. Using electron microscopy, we quantified the number of synaptic vesicles at presynaptic ribbons after light or dark adaptation. In Cplx3/4 WT photoreceptors, the number of synaptic vesicles associated with the ribbon base close to the release site was significantly lower in light than in dark. This is in contrast to Cplx3/4 DKO photoreceptors, in which the number of ribbon-associated synaptic vesicles remained unchanged regardless of the adaptational state. Our results indicate a suppressing and a facilitating action of Cplx3/4 on Ca(2+)-dependent tonic and evoked neurotransmitter release, respectively, and a regulatory role in the adaptation-dependent availability of synaptic vesicles for release at photoreceptor ribbon synapses. SIGNIFICANCE STATEMENT: Synaptic vesicle fusion at active zones of chemical synapses is executed by SNARE complexes. Complexins (Cplxs) are SNARE complex regulators and photoreceptor ribbon synapses are equipped with Cplx3 and Cplx4. The absence of both Cplxs perturbs ribbon synaptic function. Because we lack information on Cplx function in photoreceptor synaptic transmission, we investigated Cplx function using voltage-clamp recordings from postsynaptic horizontal cells of Cplx3/4 wild-type and Cplx3/4 double knock-out mice and quantified synaptic vesicle number at the ribbon after light and dark adaptation using electron microscopy. The findings reveal a suppressing action of Cplx3/4 on tonic neurotransmitter release, a facilitating action on evoked release, and a regulatory role of Cplx3/4 in the adaptation-dependent availability of synaptic vesicles at mouse photoreceptor ribbon synapses.


Assuntos
Proteínas do Olho/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células Fotorreceptoras de Vertebrados/fisiologia , Retina/citologia , Sinapses/fisiologia , Transmissão Sináptica/genética , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Animais , Cálcio/metabolismo , Proteínas do Olho/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Células Fotorreceptoras de Vertebrados/ultraestrutura , Proteínas SNARE/metabolismo , Sinapses/ultraestrutura , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura , Fatores de Tempo , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo
7.
PLoS Comput Biol ; 11(5): e1004253, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25951120

RESUMO

The spatial arrangement of Ca2+ channels and vesicles remains unknown for most CNS synapses, despite of the crucial importance of this geometrical parameter for the Ca2+ control of transmitter release. At a large model synapse, the calyx of Held, transmitter release is controlled by several Ca2+ channels in a "domain overlap" mode, at least in young animals. To study the geometrical constraints of Ca2+ channel placement in domain overlap control of release, we used stochastic MCell modelling, at active zones for which the position of docked vesicles was derived from electron microscopy (EM). We found that random placement of Ca2+ channels was unable to produce high slope values between release and presynaptic Ca2+ entry, a hallmark of domain overlap, and yielded excessively large release probabilities. The simple assumption that Ca2+ channels can be located anywhere at active zones, except below a critical distance of ~ 30 nm away from docked vesicles ("exclusion zone"), rescued high slope values and low release probabilities. Alternatively, high slope values can also be obtained by placing all Ca2+ channels into a single supercluster, which however results in significantly higher heterogeneity of release probabilities. We also show experimentally that high slope values, and the sensitivity to the slow Ca2+ chelator EGTA-AM, are maintained with developmental maturation of the calyx synapse. Taken together, domain overlap control of release represents a highly organized active zone architecture in which Ca2+ channels must obey a certain distance to docked vesicles. Furthermore, domain overlap can be employed by near-mature, fast-releasing synapses.


Assuntos
Canais de Cálcio/metabolismo , Modelos Neurológicos , Sinapses/metabolismo , Animais , Quelantes de Cálcio , Biologia Computacional , Simulação por Computador , Ácido Egtázico/análogos & derivados , Técnicas In Vitro , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Processos Estocásticos , Vesículas Sinápticas/metabolismo , Corpo Trapezoide/metabolismo
8.
J Neurosci ; 34(38): 12622-35, 2014 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-25232102

RESUMO

Ca(2+)-dependent transmitter release occurs in a fast and in a slow phase, but the differential roles of Ca(2+) buffers and Ca(2+) sensors in shaping release kinetics are still controversial. Replacing extracellular Ca(2+) by Sr(2+) causes decreased fast release but enhanced slow release at many synapses. Here, we established presynaptic Sr(2+) uncaging and made quantitative Sr(2+)- and Ca(2+)-imaging experiments at the mouse calyx of Held synapse, to reveal the interplay between Ca(2+) sensors and Ca(2+) buffers in the control of fast and slow release. We show that Sr(2+) activates the fast, Synaptotagmin-2 (Syt2) sensor for vesicle fusion with sixfold lower affinity but unchanged high cooperativity. Surprisingly, Sr(2+) also activates the slow sensor that remains in Syt2 knock-out synapses with a lower efficiency, and Sr(2+) was less efficient than Ca(2+) in the limit of low concentrations in wild-type synapses. Quantitative imaging experiments show that the buffering capacity of the nerve terminal is markedly lower for Sr(2+) than for Ca(2+) (~5-fold). This, together with an enhanced Sr(2+) permeation through presynaptic Ca(2+) channels (~2-fold), admits a drastically higher spatially averaged Sr(2+) transient compared with Ca(2+). Together, despite the lower affinity of Sr(2+) at the fast and slow sensors, the massively higher amplitudes of spatially averaged Sr(2+) transients explain the enhanced late release. This also allows us to conclude that Ca(2+) buffering normally controls late release and prevents the activation of the fast release sensor by residual Ca(2+).


Assuntos
Cálcio/metabolismo , Terminações Nervosas/fisiologia , Estrôncio/metabolismo , Transmissão Sináptica/fisiologia , Animais , Quelantes/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Knockout , Terminações Nervosas/efeitos dos fármacos , Terminações Nervosas/metabolismo , Imagem Óptica , Transmissão Sináptica/efeitos dos fármacos , Sinaptotagmina II/genética , Sinaptotagmina II/metabolismo
9.
J Neurophysiol ; 113(1): 255-63, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25343783

RESUMO

The localization and density of voltage-gated Ca(2+) channels at active zones are essential for the amount and kinetics of transmitter release at synapses. RIM proteins are scaffolding proteins at the active zone that bind to several other presynaptic proteins, including voltage-gated Ca(2+) channel α-subunits. The long isoforms of RIM proteins, which contain NH2-terminal Rab3- and Munc13-interacting domains, as well as a central PDZ domain and two COOH-terminal C2 domains, are encoded by two genes, Rim1 and Rim2. Here, we used the ideal accessibility of the large calyx of Held synapse for direct presynaptic electrophysiology to investigate whether the two Rim genes have redundant, or separate, functions in determining the presynaptic Ca(2+) channel density, and the size of a readily releasable vesicle pool (RRP). Quantitative PCR showed that cochlear nucleus neurons, which include calyx of Held generating neurons, express both RIM1 and RIM2. Conditional genetic inactivation of RIM2 at the calyx of Held led to a subtle reduction in presynaptic Ca(2+) current density, whereas deletion of RIM1 was ineffective. The release efficiency of brief presynaptic Ca(2+) "tail" currents and the RRP were unaffected in conditional single RIM1 and RIM2 knockout (KO) mice, whereas both parameters were strongly reduced in RIM1/2 double KO mice. Thus, despite a somewhat more decisive role for RIM2 in determining presynaptic Ca(2+) channel density, RIM1 and RIM2 can overall replace each other's presynaptic functions at a large relay synapse in the hindbrain, the calyx of Held.


Assuntos
Canais de Cálcio/metabolismo , Núcleo Coclear/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Sinapses/fisiologia , Vesículas Sinápticas/fisiologia , Proteínas rab3 de Ligação ao GTP/metabolismo , Animais , Cálcio/metabolismo , Núcleo Coclear/efeitos dos fármacos , Proteínas de Ligação ao GTP/genética , Ácido Glutâmico/metabolismo , Camundongos Knockout , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/fisiologia , Sinapses/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos , Técnicas de Cultura de Tecidos , Proteínas rab3 de Ligação ao GTP/genética
10.
Eur J Neurosci ; 42(9): 2615-32, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26173960

RESUMO

As all visual information is represented in the spatio-temporal dynamics of transmitter release from photoreceptors and the combined postsynaptic responses of second-order neurons, appropriate synaptic transfer functions are fundamental for a meaningful perception of the visual world. The functional contribution of horizontal cells to gain control and organization of bipolar and ganglion cell receptive fields can only be evaluated with an in-depth understanding of signal processing in horizontal cells. Therefore, a horizontal slice preparation of the mouse retina was established to record from horizontal cell bodies with their dendritic fields intact and receiving functional synaptic input from cone photoreceptors. Horizontal cell bodies showed spontaneous excitatory currents (spEPSCs) of monophasic and more complex multi-peak waveforms. spEPSCs were induced by quantal release of glutamate from presynaptic cones with a unitary amplitude of 3 pA. Non-stationary noise analysis revealed that spEPSCs with a monoexponential decay were mediated by 7-8 glutamate receptors with a single-channel amplitude of 1.55 pA. Responses to photopic full-field illumination were characterized by reduction of a tonic inward current or hyperpolarization, inhibition of spEPSCs, followed by a fast and transient inward current at light offset. The response to periodic dark/light transitions of different frequencies was dependent on the adaptational status of the cell with a limiting frequency of 10 Hz. Both on and off components of the light response were mediated by AMPA and kainate receptors. Detailed analysis of horizontal cell synaptic physiology is a prerequisite for understanding signal coding and processing at the photoreceptor ribbon synapse.


Assuntos
Potenciais Pós-Sinápticos Excitadores , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Horizontais da Retina/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Benzodiazepinas/farmacologia , Dendritos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glutamatos/farmacologia , Ácido Glutâmico/fisiologia , Camundongos , Camundongos Transgênicos , Vias Neurais/citologia , Vias Neurais/fisiologia , Estimulação Luminosa , Receptores de AMPA/agonistas , Receptores de AMPA/fisiologia , Receptores de Ácido Caínico/agonistas , Receptores de Ácido Caínico/fisiologia , Células Fotorreceptoras Retinianas Cones/citologia , Células Horizontais da Retina/citologia
11.
PLoS Biol ; 9(5): e1001057, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21559323

RESUMO

Cone photoreceptors and horizontal cells (HCs) have a reciprocal synapse that underlies lateral inhibition and establishes the antagonistic center-surround organization of the visual system. Cones transmit to HCs through an excitatory synapse and HCs feed back to cones through an inhibitory synapse. Here we report that HCs also transmit to cone terminals a positive feedback signal that elevates intracellular Ca(2+) and accelerates neurotransmitter release. Positive and negative feedback are both initiated by AMPA receptors on HCs, but positive feedback appears to be mediated by a change in HC Ca(2+), whereas negative feedback is mediated by a change in HC membrane potential. Local uncaging of AMPA receptor agonists suggests that positive feedback is spatially constrained to active HC-cone synapses, whereas the negative feedback signal spreads through HCs to affect release from surrounding cones. By locally offsetting the effects of negative feedback, positive feedback may amplify photoreceptor synaptic release without sacrificing HC-mediated contrast enhancement.


Assuntos
Retroalimentação Fisiológica , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Horizontais da Retina/fisiologia , Sinapses/fisiologia , Ambystoma , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Técnicas In Vitro , Lagartos , Potenciais da Membrana/efeitos dos fármacos , Quinoxalinas/farmacologia , Coelhos , Receptores de AMPA/antagonistas & inibidores , Receptores de AMPA/metabolismo , Receptores de Glutamato/metabolismo , Retina/citologia , Retina/efeitos dos fármacos , Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Horizontais da Retina/efeitos dos fármacos , Células Horizontais da Retina/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Peixe-Zebra , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
12.
Front Mol Neurosci ; 17: 1308466, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38481472

RESUMO

Adaptation of photoreceptor sensitivity to varying light intensities is a fundamental requirement for retinal function and vision. Adaptive mechanisms in signal transduction are well described, but little is known about the mechanisms that adapt the photoreceptor synapse to changing light intensities. The SNARE complex regulators Complexin 3 and Complexin 4 have been proposed to be involved in synaptic light adaptation by limiting synaptic vesicle recruitment and fusion. How this Complexin effect is exerted is unknown. Focusing on rod photoreceptors, we established Complexin 4 as the predominant Complexin in the light-dependent regulation of neurotransmitter release. The number of readily releasable synaptic vesicles is significantly smaller in light than in dark at wildtype compared to Complexin 4 deficient rod photoreceptor ribbon synapses. Electrophysiology indicates that Complexin 4 reduces or clamps Ca2+-dependent sustained synaptic vesicle release, thereby enhancing light signaling at the synapse. Complexin 4 deficiency increased synaptic vesicle release and desensitized light signaling. In a quantitative proteomic screen, we identified Transducin as an interactor of the Complexin 4-SNARE complex. Our results provide evidence for a presynaptic interplay of both Complexin 4 and Transducin with the SNARE complex, an interplay that may facilitate the adaptation of synaptic transmission to light at rod photoreceptor ribbon synapses.

13.
Proc Natl Acad Sci U S A ; 107(19): 8599-604, 2010 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-20445106

RESUMO

Structural features of neurons create challenges for effective production and distribution of essential metabolic energy. We investigated how metabolic energy is distributed between cellular compartments in photoreceptors. In avascular retinas, aerobic production of energy occurs only in mitochondria that are located centrally within the photoreceptor. Our findings indicate that metabolic energy flows from these central mitochondria as phosphocreatine toward the photoreceptor's synaptic terminal in darkness. In light, it flows in the opposite direction as ATP toward the outer segment. Consistent with this model, inhibition of creatine kinase in avascular retinas blocks synaptic transmission without influencing outer segment activity. Our findings also reveal how vascularization of neuronal tissue can influence the strategies neurons use for energy management. In vascularized retinas, mitochondria in the synaptic terminals of photoreceptors make neurotransmission less dependent on creatine kinase. Thus, vasculature of the tissue and the intracellular distribution of mitochondria can play key roles in setting the strategy for energy distribution in neurons.


Assuntos
Escuridão , Metabolismo Energético/fisiologia , Retina/fisiologia , Animais , Creatina Quinase/antagonistas & inibidores , Creatina Quinase/metabolismo , Dinitrofluorbenzeno/farmacologia , Eletrorretinografia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/efeitos da radiação , Glutamatos/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/efeitos da radiação , Modelos Biológicos , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/enzimologia , Terminações Pré-Sinápticas/efeitos da radiação , Inibidores de Proteínas Quinases/farmacologia , Retina/efeitos dos fármacos , Retina/enzimologia , Retina/efeitos da radiação , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/enzimologia , Células Fotorreceptoras Retinianas Cones/efeitos da radiação , Segmento Externo das Células Fotorreceptoras da Retina/efeitos dos fármacos , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/efeitos da radiação , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/enzimologia , Vasos Retinianos/efeitos da radiação , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/efeitos da radiação , Urodelos/fisiologia
14.
Invest Ophthalmol Vis Sci ; 63(3): 21, 2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35319739

RESUMO

Purpose: Cone photoreceptors of the retina use a sophisticated ribbon-containing synapse to convert light-dependent changes in membrane potential into release of synaptic vesicles (SVs). We aimed to study the functional and structural maturation of mouse cone photoreceptor ribbon synapses during postnatal development and to investigate the role of the synaptic ribbon in SV release. Methods: We performed patch-clamp recordings from cone photoreceptors and their postsynaptic partners, the horizontal cells during postnatal retinal development to reveal the functional parameters of the synapses. To investigate the occurring structural changes, we applied immunocytochemistry and electron microscopy. Results: We found that immature cone photoreceptor terminals were smaller, they had fewer active zones (AZs) and AZ-anchored synaptic ribbons, and they produced a smaller Ca2+ current than mature photoreceptors. The number of postsynaptic horizontal cell contacts to synaptic terminals increased with age. However, tonic and spontaneous SV release at synaptic terminals stayed similar during postnatal development. Multiquantal SV release was present in all age groups, but mature synapses produced larger multiquantal events than immature ones. Remarkably, at single AZs, tonic SV release was attenuated during maturation and showed an inverse relationship with the appearance of anchored synaptic ribbons. Conclusions: Our developmental study suggests that the presence of synaptic ribbons at the AZs attenuates tonic SV release and amplifies multiquantal SV release. However, spontaneous SV release may not depend on the presence of synaptic ribbons or voltage-sensitive Ca2+ channels at the AZs.


Assuntos
Células Fotorreceptoras Retinianas Cones , Sinapses , Animais , Citoesqueleto , Camundongos , Retina , Sinapses/fisiologia , Vesículas Sinápticas
15.
Front Cell Neurosci ; 16: 1022419, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36406751

RESUMO

Mammalian cone photoreceptors enable through their sophisticated synapse the high-fidelity transfer of visual information to second-order neurons in the retina. The synapse contains a proteinaceous organelle, called the synaptic ribbon, which tethers synaptic vesicles (SVs) at the active zone (AZ) close to voltage-gated Ca2+ channels. However, the exact contribution of the synaptic ribbon to neurotransmission is not fully understood, yet. In mice, precursors to synaptic ribbons appear within photoreceptor terminals shortly after birth as free-floating spherical structures, which progressively elongate and then attach to the AZ during the following days. Here, we took advantage of the process of synaptic ribbon maturation to study their contribution to SV release. We performed whole-cell patch-clamp recordings from cone photoreceptors at three postnatal (P) development stages (P8-9, P12-13, >P30) and measured evoked SV release, SV replenishment rate, recovery from synaptic depression, domain organization of voltage-sensitive Ca2+ channels, and Ca2+-sensitivity of exocytosis. Additionally, we performed electron microscopy to determine the density of SVs at ribbon-free and ribbon-occupied AZs. Our results suggest that ribbon attachment does not organize the voltage-sensitive Ca2+ channels into nanodomains or control SV release probability. However, ribbon attachment increases SV density at the AZ, increases the pool size of readily releasable SVs available for evoked SV release, facilitates SV replenishment without changing the SV pool refilling time, and increases the Ca2+- sensitivity of glutamate release.

16.
J Neurosci ; 30(47): 15866-77, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21106825

RESUMO

Cones release glutamate-filled vesicles continuously in darkness, and changing illumination modulates this release. Because sustained release in darkness is governed by vesicle replenishment rates, we analyzed how cone membrane potential regulates replenishment. Synaptic release from cones was measured by recording postsynaptic currents in Ambystoma tigrinum horizontal or OFF bipolar cells evoked by depolarization of simultaneously voltage-clamped cones. We measured replenishment after attaining a steady state between vesicle release and replenishment using trains of test pulses. Increasing Ca(2+) currents (I(Ca)) by changing the test step from -30 to -10 mV increased replenishment. Lengthening -30 mV test pulses to match the Ca(2+) influx during 25 ms test pulses to -10 mV produced similar replenishment rates. Reducing Ca(2+) driving force by using test steps to +30 mV slowed replenishment. Using UV flashes to reverse inhibition of I(Ca) by nifedipine accelerated replenishment. Increasing [Ca(2+)](i) by flash photolysis of caged Ca(2+) also accelerated replenishment. Replenishment, but not the initial burst of release, was enhanced by using an intracellular Ca(2+) buffer of 0.5 mm EGTA rather than 5 mm EGTA, and diminished by 1 mm BAPTA. This suggests that although release and replenishment exhibited similar Ca(2+) dependencies, release sites are <200 nm from Ca(2+) channels but replenishment sites are >200 nm away. Membrane potential thus regulates replenishment by controlling Ca(2+) influx, principally by effects on replenishment mechanisms but also by altering releasable pool size. This in turn provides a mechanism for converting changes in light intensity into changes in sustained release at the cone ribbon synapse.


Assuntos
Cálcio/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Sinapses/fisiologia , Vesículas Sinápticas/metabolismo , Ambystoma , Animais , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Vesículas Sinápticas/fisiologia
17.
J Neurophysiol ; 106(6): 2922-35, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21880934

RESUMO

Light hyperpolarizes cone photoreceptors, causing synaptic voltage-gated Ca(2+) channels to open infrequently. To understand neurotransmission under these conditions, we determined the number of L-type Ca(2+) channel openings necessary for vesicle fusion at the cone ribbon synapse. Ca(2+) currents (I(Ca)) were activated in voltage-clamped cones, and excitatory postsynaptic currents (EPSCs) were recorded from horizontal cells in the salamander retina slice preparation. Ca(2+) channel number and single-channel current amplitude were calculated by mean-variance analysis of I(Ca). Two different comparisons-one comparing average numbers of release events to average I(Ca) amplitude and the other involving deconvolution of both EPSCs and simultaneously recorded cone I(Ca)-suggested that fewer than three Ca(2+) channel openings accompanied fusion of each vesicle at the peak of release during the first few milliseconds of stimulation. Opening fewer Ca(2+) channels did not enhance fusion efficiency, suggesting that few unnecessary channel openings occurred during strong depolarization. We simulated release at the cone synapse, using empirically determined synaptic dimensions, vesicle pool size, Ca(2+) dependence of release, Ca(2+) channel number, and Ca(2+) channel properties. The model replicated observations when a barrier was added to slow Ca(2+) diffusion. Consistent with the presence of a diffusion barrier, dialyzing cones with diffusible Ca(2+) buffers did not affect release efficiency. The tight clustering of Ca(2+) channels, along with a high-Ca(2+) affinity release mechanism and diffusion barrier, promotes a linear coupling between Ca(2+) influx and vesicle fusion. This may improve detection of small light decrements when cones are hyperpolarized by bright light.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Cálcio/metabolismo , Ativação do Canal Iônico/fisiologia , Luz , Retina/citologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Sinapses/fisiologia , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Bário/farmacologia , Fenômenos Biofísicos/efeitos dos fármacos , Cálcio/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Quelantes/farmacologia , Simulação por Computador , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Técnicas In Vitro , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Modelos Biológicos , Nifedipino/farmacologia , Técnicas de Patch-Clamp , Probabilidade , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Células Horizontais da Retina/efeitos dos fármacos , Células Horizontais da Retina/fisiologia , Sinapses/efeitos dos fármacos , Urodelos
18.
Stem Cells ; 28(4): 695-703, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20166150

RESUMO

The direct reprogramming of somatic cells to a pluripotent state holds significant implications for treating intractable degenerative diseases by ex vivo cell therapy. In addition, the reprogrammed cells can serve as a model for diseases and the discovery of drugs and genes. Here, we demonstrate that mouse fibroblast induced pluripotent stem cells (iPSCs) represent a renewable and robust source of retinal progenitors, capable of generating a wide range of retinal cell types that includes retinal ganglion cells (RGCs), cone, and rod photoreceptors. They respond to simulated microenvironment of early and late retinal histogenesis by differentiating into stage-specific retinal cell types through the recruitment of normal mechanisms. The depth of the retinal potential of iPSCs suggests that they may be used to formulate stem cell approaches to understand and treat a wide range of retinal degenerative diseases from glaucoma to age-related macular degeneration (AMD).


Assuntos
Diferenciação Celular , Glaucoma/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Degeneração Macular/terapia , Células Fotorreceptoras/citologia , Células Ganglionares da Retina/citologia , Animais , Linhagem Celular , Linhagem da Célula , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Células Fotorreceptoras/metabolismo , Células Ganglionares da Retina/metabolismo
19.
Vis Neurosci ; 28(2): 145-54, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21463541

RESUMO

Retinal prosthetic devices are being developed to bypass degenerated retinal photoreceptors by directly activating retinal neurons with electrical stimulation. However, the retinal circuitry that is activated by epiretinal stimulation is not well characterized. Whole-cell patch clamp recordings were obtained from ganglion cells in normal and rd mice using flat-mount and retinal slice preparations. A stimulating electrode was positioned along the ganglion cell side of the preparation at different distances from the stimulated tissue. Pulses of cathodic current evoked action potentials in ganglion cells and less frequently evoked sustained inward currents that appeared synaptic in origin. Sustained currents reversed around E(Cl) and were inhibited by blockade of α-amino-3-hydroxyl-5-methyl-4-isoxazole-proprionate (AMPA)-type glutamate receptors with 2,3-dihydroxy-6-nitro-sulfamoyl-benzo(f)-quinoxaline-2,3-dione (NBQX), γ aminobutyric acid a/c (GABA(a/c)) receptors with picrotoxinin, or glycine receptors with strychnine. This suggests that epiretinal stimulation activates glutamate release from bipolar cell terminals, which in turn evokes release of GABA and glycine from amacrine cells. Synaptic current thresholds were lower in ON ganglion cells than OFF cells, but the modest difference did not attain statistical significance. Synaptic currents were rarely observed in rd mice lacking photoreceptors compared to normal retina. In addition, confocal calcium imaging experiments in normal mice retina slices revealed that epiretinal stimulation evoked calcium increases in the outer plexiform layer. These results imply a contribution from photoreceptor inputs to the synaptic currents observed in ganglion cells. The paucity of synaptic responses in rd mice retina slices suggests that it is better to target retinal ganglion cells directly rather than to attempt to engage the inner retinal circuitry.


Assuntos
Potenciais Evocados/genética , Potenciais Evocados/fisiologia , Retina/patologia , Degeneração Retiniana/patologia , Células Ganglionares da Retina/fisiologia , Animais , Biofísica , Cálcio/metabolismo , Modelos Animais de Doenças , Estimulação Elétrica/métodos , Potenciais Evocados/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Antagonistas de Receptores de GABA-A/farmacologia , Glicinérgicos/farmacologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Confocal , Técnicas de Patch-Clamp/métodos , Picrotoxina/análogos & derivados , Picrotoxina/farmacologia , Quinoxalinas/farmacologia , Degeneração Retiniana/classificação , Degeneração Retiniana/genética , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/efeitos da radiação , Sesterterpenos , Estricnina/farmacologia , Vias Visuais/efeitos dos fármacos , Vias Visuais/fisiologia
20.
Sci Rep ; 11(1): 2732, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33526839

RESUMO

CaV1.4 L-type calcium channels are predominantly expressed in photoreceptor terminals playing a crucial role for synaptic transmission and, consequently, for vision. Human mutations in the encoding gene are associated with congenital stationary night blindness type-2. Besides rod-driven scotopic vision also cone-driven photopic responses are severely affected in patients. The present study therefore examined functional and morphological changes in cones and cone-related pathways in mice carrying the CaV1.4 gain-of function mutation I756T (CaV1.4-IT) using multielectrode array, patch-clamp and immunohistochemical analyses. CaV1.4-IT ganglion cell responses to photopic stimuli were seen only in a small fraction of cells indicative of a major impairment in the cone pathway. Though cone photoreceptors underwent morphological rearrangements, they retained their ability to release glutamate. Our functional data suggested a postsynaptic cone bipolar cell defect, supported by the fact that the majority of cone bipolar cells showed sprouting, while horizontal cells maintained contacts with cones and cone-to-horizontal cell input was preserved. Furthermore a reduction of basal Ca2+ influx by a calcium channel blocker was not sufficient to rescue synaptic transmission deficits caused by the CaV1.4-IT mutation. Long term treatments with low-dose Ca2+ channel blockers might however be beneficial reducing Ca2+ toxicity without major effects on ganglion cells responses.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Vias Visuais/fisiologia , Animais , Canais de Cálcio Tipo L/genética , Forma Celular/fisiologia , Camundongos , Camundongos Transgênicos , Retina/citologia , Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/citologia , Sinapses/metabolismo , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA