Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 44(25)2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38641407

RESUMO

Vertebrate vision begins with light absorption by rod and cone photoreceptors, which transmit signals from their synaptic terminals to second-order neurons: bipolar and horizontal cells. In mouse rods, there is a single presynaptic ribbon-type active zone at which the release of glutamate occurs tonically in the dark. This tonic glutamatergic signaling requires continuous exo- and endocytosis of synaptic vesicles. At conventional synapses, endocytosis commonly requires dynamins: GTPases encoded by three genes (Dnm1-3), which perform membrane scission. Disrupting endocytosis by dynamin deletions impairs transmission at conventional synapses, but the impact of disrupting endocytosis and the role(s) of specific dynamin isoforms at rod ribbon synapses are understood incompletely. Here, we used cell-specific knock-outs (KOs) of the neuron-specific Dnm1 and Dnm3 to investigate the functional roles of dynamin isoforms in rod photoreceptors in mice of either sex. Analysis of synaptic protein expression, synapse ultrastructure, and retinal function via electroretinograms (ERGs) showed that dynamins 1 and 3 act redundantly and are essential for supporting the structural and functional integrity of rod ribbon synapses. Single Dnm3 KO showed no phenotype, and single Dnm1 KO only modestly reduced synaptic vesicle density without affecting vesicle size and overall synapse integrity, whereas double Dnm1/Dnm3 KO impaired vesicle endocytosis profoundly, causing enlarged vesicles, reduced vesicle density, reduced ERG responses, synaptic terminal degeneration, and disassembly and degeneration of postsynaptic processes. Concurrently, cone function remained intact. These results show the fundamental redundancy of dynamins 1 and 3 in regulating the structure and function of rod ribbon synapses.


Assuntos
Dinamina III , Dinamina I , Eletrorretinografia , Camundongos Knockout , Células Fotorreceptoras Retinianas Bastonetes , Sinapses , Animais , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/ultraestrutura , Camundongos , Sinapses/fisiologia , Sinapses/metabolismo , Sinapses/ultraestrutura , Masculino , Feminino , Dinamina I/metabolismo , Dinamina I/genética , Dinamina III/genética , Dinamina III/metabolismo , Camundongos Endogâmicos C57BL
2.
PLoS Biol ; 21(11): e3002386, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37983249

RESUMO

Defensive responses to visually threatening stimuli represent an essential fear-related survival instinct, widely detected across species. The neural circuitry mediating visually triggered defensive responses has been delineated in the midbrain. However, the molecular mechanisms regulating the development and function of these circuits remain unresolved. Here, we show that midbrain-specific deletion of the transcription factor Brn3b causes a loss of neurons projecting to the lateral posterior nucleus of the thalamus. Brn3b deletion also down-regulates the expression of the neuropeptide tachykinin 2 (Tac2). Furthermore, Brn3b mutant mice display impaired defensive freezing responses to visual threat precipitated by social isolation. This behavioral phenotype could be ameliorated by overexpressing Tac2, suggesting that Tac2 acts downstream of Brn3b in regulating defensive responses to threat. Together, our experiments identify specific genetic components critical for the functional organization of midbrain fear-related visual circuits. Similar mechanisms may contribute to the development and function of additional long-range brain circuits underlying fear-associated behavior.


Assuntos
Medo , Mesencéfalo , Animais , Camundongos , Medo/fisiologia , Mesencéfalo/fisiologia , Neurônios/fisiologia , Tálamo
3.
bioRxiv ; 2023 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-38234775

RESUMO

Visual information processing is sculpted by a diverse group of inhibitory interneurons in the retina called amacrine cells. Yet, for most of the >60 amacrine cell types, molecular identities and specialized functional attributes remain elusive. Here, we developed an intersectional genetic strategy to target a group of wide-field amacrine cells (WACs) in mouse retina that co-express the transcription factor Bhlhe22 and the Kappa Opioid Receptor (KOR; B/K WACs). B/K WACs feature straight, unbranched dendrites spanning over 0.5 mm (∼15° visual angle) and produce non-spiking responses to either light increments or decrements. Two-photon dendritic population imaging reveals Ca 2+ signals tuned to the physical orientations of B/K WAC dendrites, signifying a robust structure-function alignment. B/K WACs establish divergent connections with multiple retinal neurons, including unexpected connections with non-orientation-tuned ganglion cells and bipolar cells. Our work sets the stage for future comprehensive investigations of the most enigmatic group of retinal neurons: WACs.

4.
Front Cell Neurosci ; 15: 660773, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34381333

RESUMO

A presynaptic neuron can increase its computational capacity by transmitting functionally distinct signals to each of its postsynaptic cell types. To determine whether such computational specialization occurs over fine spatial scales within a neurite arbor, we investigated computation at output synapses of the starburst amacrine cell (SAC), a critical component of the classical direction-selective (DS) circuit in the retina. The SAC is a non-spiking interneuron that co-releases GABA and acetylcholine and forms closely spaced (<5 µm) inhibitory synapses onto two postsynaptic cell types: DS ganglion cells (DSGCs) and neighboring SACs. During dynamic optogenetic stimulation of SACs in mouse retina, whole-cell recordings of inhibitory postsynaptic currents revealed that GABAergic synapses onto DSGCs exhibit stronger low-pass filtering than those onto neighboring SACs. Computational analyses suggest that this filtering difference can be explained primarily by presynaptic properties, rather than those of the postsynaptic cells per se. Consistent with functionally diverse SAC presynapses, blockade of N-type voltage-gated calcium channels abolished GABAergic currents in SACs but only moderately reduced GABAergic and cholinergic currents in DSGCs. These results jointly demonstrate how specialization of synaptic outputs could enhance parallel processing in a compact interneuron over fine spatial scales. Moreover, the distinct transmission kinetics of GABAergic SAC synapses are poised to support the functional diversity of inhibition within DS circuitry.

5.
Cell ; 184(16): 4299-4314.e12, 2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34297923

RESUMO

Retinal ganglion cells (RGCs) are the sole output neurons that transmit visual information from the retina to the brain. Diverse insults and pathological states cause degeneration of RGC somas and axons leading to irreversible vision loss. A fundamental question is whether manipulation of a key regulator of RGC survival can protect RGCs from diverse insults and pathological states, and ultimately preserve vision. Here, we report that CaMKII-CREB signaling is compromised after excitotoxic injury to RGC somas or optic nerve injury to RGC axons, and reactivation of this pathway robustly protects RGCs from both injuries. CaMKII activity also promotes RGC survival in the normal retina. Further, reactivation of CaMKII protects RGCs in two glaucoma models where RGCs degenerate from elevated intraocular pressure or genetic deficiency. Last, CaMKII reactivation protects long-distance RGC axon projections in vivo and preserves visual function, from the retina to the visual cortex, and visually guided behavior.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Citoproteção , Células Ganglionares da Retina/patologia , Visão Ocular , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Encéfalo/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dependovirus/metabolismo , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Glaucoma/genética , Glaucoma/patologia , Camundongos Endogâmicos C57BL , Neurotoxinas/toxicidade , Traumatismos do Nervo Óptico/patologia , Transdução de Sinais
6.
J Neurosci ; 41(7): 1489-1504, 2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33397711

RESUMO

Intrinsically photosensitive retinal ganglion cells (ipRGCs) exhibit melanopsin-dependent light responses that persist in the absence of rod and cone photoreceptor-mediated input. In addition to signaling anterogradely to the brain, ipRGCs signal retrogradely to intraretinal circuitry via gap junction-mediated electrical synapses with amacrine cells (ACs). However, the targets and functions of these intraretinal signals remain largely unknown. Here, in mice of both sexes, we identify circuitry that enables M5 ipRGCs to locally inhibit retinal neurons via electrical synapses with a nonspiking GABAergic AC. During pharmacological blockade of rod- and cone-mediated input, whole-cell recordings of corticotropin-releasing hormone-expressing (CRH+) ACs reveal persistent visual responses that require both melanopsin expression and gap junctions. In the developing retina, ipRGC-mediated input to CRH+ ACs is weak or absent before eye opening, indicating a primary role for this input in the mature retina (i.e., in parallel with rod- and cone-mediated input). Among several ipRGC types, only M5 ipRGCs exhibit consistent anatomical and physiological coupling to CRH+ ACs. Optogenetic stimulation of local CRH+ ACs directly drives IPSCs in M4 and M5, but not M1-M3, ipRGCs. CRH+ ACs also inhibit M2 ipRGC-coupled spiking ACs, demonstrating direct interaction between discrete networks of ipRGC-coupled interneurons. Together, these results demonstrate a functional role for electrical synapses in translating ipRGC activity into feedforward and feedback inhibition of local retinal circuits.SIGNIFICANCE STATEMENT Melanopsin directly generates light responses in intrinsically photosensitive retinal ganglion cells (ipRGCs). Through gap junction-mediated electrical synapses with retinal interneurons, these uniquely photoreceptive RGCs may also influence the activity and output of neuronal circuits within the retina. Here, we identified and studied an electrical synaptic circuit that, in principle, could couple ipRGC activity to the chemical output of an identified retinal interneuron. Specifically, we found that M5 ipRGCs form electrical synapses with corticotropin-releasing hormone-expressing amacrine cells, which locally release GABA to inhibit specific RGC types. Thus, ipRGCs are poised to influence the output of diverse retinal circuits via electrical synapses with interneurons.


Assuntos
Inibição Neural/fisiologia , Células Fotorreceptoras de Vertebrados/fisiologia , Retina/fisiologia , Células Ganglionares da Retina/fisiologia , Células Amácrinas/fisiologia , Animais , Hormônio Liberador da Corticotropina/fisiologia , Fenômenos Eletrofisiológicos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Junções Comunicantes/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Optogenética , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Opsinas de Bastonetes/metabolismo , Sinapses/fisiologia , Ácido gama-Aminobutírico/fisiologia
7.
Front Cell Neurosci ; 14: 604163, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33324168

RESUMO

Direction selectivity represents an elementary sensory computation that can be related to underlying synaptic mechanisms. In mammalian retina, direction-selective ganglion cells (DSGCs) respond strongly to visual motion in a "preferred" direction and weakly to motion in the opposite, "null" direction. The DS mechanism depends on starburst amacrine cells (SACs), which provide null direction-tuned GABAergic inhibition and untuned cholinergic excitation to DSGCs. GABAergic inhibition depends on conventional synaptic transmission, whereas cholinergic excitation apparently depends on paracrine (i.e., non-synaptic) transmission. Despite its paracrine mode of transmission, cholinergic excitation is more transient than GABAergic inhibition, yielding a temporal difference that contributes essentially to the DS computation. To isolate synaptic mechanisms that generate the distinct temporal properties of cholinergic and GABAergic transmission from SACs to DSGCs, we optogenetically stimulated SACs while recording postsynaptic currents (PSCs) from DSGCs in mouse retina. Direct recordings from channelrhodopsin-2-expressing (ChR2+) SACs during quasi-white noise (WN) (0-30 Hz) photostimulation demonstrated precise, graded optogenetic control of SAC membrane current and potential. Linear systems analysis of ChR2-evoked PSCs recorded in DSGCs revealed cholinergic transmission to be faster than GABAergic transmission. A deconvolution-based analysis showed that distinct postsynaptic receptor kinetics fully account for the temporal difference between cholinergic and GABAergic transmission. Furthermore, GABAA receptor blockade prolonged cholinergic transmission, identifying a new functional role for GABAergic inhibition of SACs. Thus, fast cholinergic transmission from SACs to DSGCs arises from at least two distinct mechanisms, yielding temporal properties consistent with conventional synapses despite its paracrine nature.

8.
Elife ; 92020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32412412

RESUMO

Night vision in mammals depends fundamentally on rod photoreceptors and the well-studied rod bipolar (RB) cell pathway. The central neuron in this pathway, the AII amacrine cell (AC), exhibits a spatially tuned receptive field, composed of an excitatory center and an inhibitory surround, that propagates to ganglion cells, the retina's projection neurons. The circuitry underlying the surround of the AII, however, remains unresolved. Here, we combined structural, functional and optogenetic analyses of the mouse retina to discover that surround inhibition of the AII depends primarily on a single interneuron type, the NOS-1 AC: a multistratified, axon-bearing GABAergic cell, with dendrites in both ON and OFF synaptic layers, but with a pure ON (depolarizing) response to light. Our study demonstrates generally that novel neural circuits can be identified from targeted connectomic analyses and specifically that the NOS-1 AC mediates long-range inhibition during night vision and is a major element of the RB pathway.


Assuntos
Células Amácrinas/fisiologia , Neurônios GABAérgicos/fisiologia , Inibição Neural , Vias Neurais/fisiologia , Visão Noturna , Transmissão Sináptica , Células Amácrinas/metabolismo , Animais , Neurônios GABAérgicos/metabolismo , Genes Reporter , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Vias Neurais/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Optogenética
9.
J Comp Neurol ; 527(1): 117-132, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28856684

RESUMO

The mammalian retina encodes visual information in dim light using rod photoreceptors and a specialized circuit: rods→rod bipolar cells→AII amacrine cell. The AII amacrine cell uses sign-conserving electrical synapses to modulate ON cone bipolar cell terminals and sign-inverting chemical (glycinergic) synapses to modulate OFF cone cell bipolar terminals; these ON and OFF cone bipolar terminals then drive the output neurons, retinal ganglion cells (RGCs), following light increments and decrements, respectively. The AII amacrine cell also makes direct glycinergic synapses with certain RGCs, but it is not well established how many types receive this direct AII input. Here, we investigated functional AII amacrine→RGC synaptic connections in the retina of the guinea pig (Cavia porcellus) by recording inhibitory currents from RGCs in the presence of ionotropic glutamate receptor (iGluR) antagonists. This condition isolates a specific pathway through the AII amacrine cell that does not require iGluRs: cone→ON cone bipolar cell→AII amacrine cell→RGC. These recordings show that AII amacrine cells make direct synapses with OFF Alpha, OFF Delta and a smaller OFF transient RGC type that co-stratifies with OFF Alpha cells. However, AII amacrine cells avoid making synapses with numerous RGC types that co-stratify with the connected RGCs. Selective AII connections ensure that a privileged minority of RGC types receives direct input from the night-vision pathway, independent from OFF bipolar cell activity. Furthermore, these results illustrate the specificity of retinal connections, which cannot be predicted solely by co-stratification of dendrites and axons within the inner plexiform layer.


Assuntos
Células Amácrinas/ultraestrutura , Vias Neurais/ultraestrutura , Visão Noturna/fisiologia , Células Ganglionares da Retina/ultraestrutura , Sinapses/ultraestrutura , Células Amácrinas/fisiologia , Animais , Cobaias , Vias Neurais/fisiologia , Células Ganglionares da Retina/fisiologia , Sinapses/fisiologia
10.
Proc Natl Acad Sci U S A ; 115(51): E12083-E12090, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30509993

RESUMO

Synaptic inhibition controls a neuron's output via functionally distinct inputs at two subcellular compartments, the cell body and the dendrites. It is unclear whether the assembly of these distinct inhibitory inputs can be regulated independently by neurotransmission. In the mammalian retina, γ-aminobutyric acid (GABA) release from starburst amacrine cells (SACs) onto the dendrites of on-off direction-selective ganglion cells (ooDSGCs) is essential for directionally selective responses. We found that ooDSGCs also receive GABAergic input on their somata from other amacrine cells (ACs), including ACs containing the vasoactive intestinal peptide (VIP). When net GABAergic transmission is reduced, somatic, but not dendritic, GABAA receptor clusters on the ooDSGC increased in number and size. Correlative fluorescence imaging and serial electron microscopy revealed that these enlarged somatic receptor clusters are localized to synapses. By contrast, selectively blocking vesicular GABA release from either SACs or VIP ACs did not alter dendritic or somatic receptor distributions on the ooDSGCs, showing that neither SAC nor VIP AC GABA release alone is required for the development of inhibitory synapses in ooDSGCs. Furthermore, a reduction in net GABAergic transmission, but not a selective reduction from SACs, increased excitatory drive onto ooDSGCs. This increased excitation may drive a homeostatic increase in ooDSGC somatic GABAA receptors. Differential regulation of GABAA receptors on the ooDSGC's soma and dendrites could facilitate homeostatic control of the ooDSGC's output while enabling the assembly of the GABAergic connectivity underlying direction selectivity to be indifferent to altered transmission.


Assuntos
Células Ganglionares da Retina/fisiologia , Transmissão Sináptica/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Camundongos , Camundongos Transgênicos , Receptores de GABA-A/metabolismo , Receptores de GABA-A/fisiologia , Células Ganglionares da Retina/metabolismo , Sinapses/metabolismo , Sinapses/fisiologia , Transmissão Sináptica/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Ácido gama-Aminobutírico/metabolismo
11.
Nature ; 560(7719): 484-488, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30111842

RESUMO

In zebrafish, Müller glia (MG) are a source of retinal stem cells that can replenish damaged retinal neurons and restore vision1. In mammals, however, MG do not spontaneously re-enter the cell cycle to generate a population of stem or progenitor cells that differentiate into retinal neurons. Nevertheless, the regenerative machinery may exist in the mammalian retina, as retinal injury can stimulate MG proliferation followed by limited neurogenesis2-7. Therefore, there is still a fundamental question regarding whether MG-derived regeneration can be exploited to restore vision in mammalian retinas. Gene transfer of ß-catenin stimulates MG proliferation in the absence of injury in mouse retinas8. Here we report that following gene transfer of ß-catenin, cell-cycle-reactivated MG can be reprogrammed to generate rod photoreceptors by subsequent gene transfer of transcription factors essential for rod cell fate specification and determination. MG-derived rods restored visual responses in Gnat1rd17Gnat2cpfl3 double mutant mice, a model of congenital blindness9,10, throughout the visual pathway from the retina to the primary visual cortex. Together, our results provide evidence of vision restoration after de novo MG-derived genesis of rod photoreceptors in mammalian retinas.


Assuntos
Reprogramação Celular/genética , Neurogênese , Células Fotorreceptoras Retinianas Bastonetes/citologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células-Tronco/citologia , Animais , Cegueira/congênito , Cegueira/genética , Cegueira/terapia , Ciclo Celular , Proliferação de Células/genética , Modelos Animais de Doenças , Feminino , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/genética , Masculino , Camundongos , Neuroglia/citologia , Neuroglia/metabolismo , Medicina Regenerativa , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transducina/genética , Córtex Visual/citologia , Vias Visuais , beta Catenina/genética , beta Catenina/metabolismo
12.
Neuron ; 99(4): 630-632, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30138585

RESUMO

Melanopsin is a photopigment expressed by certain types of retinal ganglion cells that mediate non-image-forming visual functions, such as circadian photoentrainment. In this issue of Neuron, Sonoda et al. (2018) reveal how melanopsin also regulates the sensitivity of conventional image-forming vision.


Assuntos
Transdução de Sinal Luminoso , Opsinas de Bastonetes , Células Ganglionares da Retina , Visão Ocular
13.
J Neurosci ; 38(15): 3753-3766, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29572434

RESUMO

Inhibitory interneurons sculpt the outputs of excitatory circuits to expand the dynamic range of information processing. In mammalian retina, >30 types of amacrine cells provide lateral inhibition to vertical, excitatory bipolar cell circuits, but functional roles for only a few amacrine cells are well established. Here, we elucidate the function of corticotropin-releasing hormone (CRH)-expressing amacrine cells labeled in Cre-transgenic mice of either sex. CRH cells costratify with the ON alpha ganglion cell, a neuron highly sensitive to positive contrast. Electrophysiological and optogenetic analyses demonstrate that two CRH types (CRH-1 and CRH-3) make GABAergic synapses with ON alpha cells. CRH-1 cells signal via graded membrane potential changes, whereas CRH-3 cells fire action potentials. Both types show sustained ON-type responses to positive contrast over a range of stimulus conditions. Optogenetic control of transmission at CRH-1 synapses demonstrates that these synapses are tuned to low temporal frequencies, maintaining GABA release during fast hyperpolarizations during brief periods of negative contrast. CRH amacrine cell output is suppressed by prolonged negative contrast, when ON alpha ganglion cells continue to receive inhibitory input from converging OFF-pathway amacrine cells; the converging ON- and OFF-pathway inhibition balances tonic excitatory drive to ON alpha cells. Previously, it was demonstrated that CRH-1 cells inhibit firing by suppressed-by-contrast (SbC) ganglion cells during positive contrast. Therefore, divergent outputs of CRH-1 cells inhibit two ganglion cell types with opposite responses to positive contrast. The opposing responses of ON alpha and SbC ganglion cells are explained by differing excitation/inhibition balance in the two circuits.SIGNIFICANCE STATEMENT A goal of neuroscience research is to explain the function of neural circuits at the level of specific cell types. Here, we studied the function of specific types of inhibitory interneurons, corticotropin-releasing hormone (CRH) amacrine cells, in the mouse retina. Genetic tools were used to identify and manipulate CRH cells, which make GABAergic synapses with a well studied ganglion cell type, the ON alpha cell. CRH cells converge with other types of amacrine cells to tonically inhibit ON alpha cells and balance their high level of excitation. CRH cells diverge to different types of ganglion cell, the unique properties of which depend on their balance of excitation and inhibition.


Assuntos
Células Amácrinas/fisiologia , Hormônio Liberador da Corticotropina/metabolismo , Vias Visuais/citologia , Potenciais de Ação , Células Amácrinas/metabolismo , Animais , Feminino , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/fisiologia , Potenciais Sinápticos , Vias Visuais/fisiologia
14.
Nature ; 546(7659): 476-477, 2017 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-28607483
15.
Elife ; 52016 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-27841746

RESUMO

Visual processing depends on specific computations implemented by complex neural circuits. Here, we present a circuit-inspired model of retinal ganglion cell computation, targeted to explain their temporal dynamics and adaptation to contrast. To localize the sources of such processing, we used recordings at the levels of synaptic input and spiking output in the in vitro mouse retina. We found that an ON-Alpha ganglion cell's excitatory synaptic inputs were described by a divisive interaction between excitation and delayed suppression, which explained nonlinear processing that was already present in ganglion cell inputs. Ganglion cell output was further shaped by spike generation mechanisms. The full model accurately predicted spike responses with unprecedented millisecond precision, and accurately described contrast adaptation of the spike train. These results demonstrate how circuit and cell-intrinsic mechanisms interact for ganglion cell function and, more generally, illustrate the power of circuit-inspired modeling of sensory processing.


Assuntos
Potenciais de Ação , Modelos Neurológicos , Células Ganglionares da Retina/fisiologia , Percepção Visual , Adaptação Ocular , Animais , Camundongos
16.
Curr Biol ; 26(20): R1062-R1072, 2016 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-27780048

RESUMO

Sensory systems use receptors to extract information from the environment and neural circuits to perform subsequent computations. These computations may be described as algorithms composed of sequential mathematical operations. Comparing these operations across taxa reveals how different neural circuits have evolved to solve the same problem, even when using different mechanisms to implement the underlying math. In this review, we compare how insect and mammalian neural circuits have solved the problem of motion estimation, focusing on the fruit fly Drosophila and the mouse retina. Although the two systems implement computations with grossly different anatomy and molecular mechanisms, the underlying circuits transform light into motion signals with strikingly similar processing steps. These similarities run from photoreceptor gain control and spatiotemporal tuning to ON and OFF pathway structures, motion detection, and computed motion signals. The parallels between the two systems suggest that a limited set of algorithms for estimating motion satisfies both the needs of sighted creatures and the constraints imposed on them by metabolism, anatomy, and the structure and regularities of the visual world.


Assuntos
Drosophila/fisiologia , Camundongos/fisiologia , Percepção de Movimento , Animais , Insetos/fisiologia , Mamíferos/fisiologia
17.
Cell Rep ; 15(10): 2239-2250, 2016 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-27239031

RESUMO

Complexin (Cplx) proteins modulate the core SNARE complex to regulate exocytosis. To understand the contributions of Cplx to signaling in a well-characterized neural circuit, we investigated how Cplx3, a retina-specific paralog, shapes transmission at rod bipolar (RB)→AII amacrine cell synapses in the mouse retina. Knockout of Cplx3 strongly attenuated fast, phasic Ca(2+)-dependent transmission, dependent on local [Ca(2+)] nanodomains, but enhanced slower Ca(2+)-dependent transmission, dependent on global intraterminal [Ca(2+)] ([Ca(2+)]I). Surprisingly, coordinated multivesicular release persisted at Cplx3(-/-) synapses, although its onset was slowed. Light-dependent signaling at Cplx3(-/-) RB→AII synapses was sluggish, owing largely to increased asynchronous release at light offset. Consequently, propagation of RB output to retinal ganglion cells was suppressed dramatically. Our study links Cplx3 expression with synapse and circuit function in a specific retinal pathway and reveals a role for asynchronous release in circuit gain control.


Assuntos
Exocitose , Proteínas do Olho/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Retina/citologia , Retina/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Cálcio/farmacologia , Exocitose/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Corpos Multivesiculares/efeitos dos fármacos , Corpos Multivesiculares/metabolismo , Proteínas do Tecido Nervoso/deficiência , Retina/efeitos dos fármacos , Células Bipolares da Retina/efeitos dos fármacos , Células Bipolares da Retina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
18.
Neuron ; 90(2): 207-9, 2016 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-27100191

RESUMO

In this issue of Neuron, Kuo et al. (2016) show that coordinated interaction between electrical and chemical synapses in a defined retinal circuit enhances sensitivity to moving objects. Their work demonstrates how electrical and chemical synapses combine to improve information processing in a specific area of the CNS.


Assuntos
Sinapses Elétricas , Junções Comunicantes , Humanos , Neurônios , Retina , Sinapses
19.
J Comp Neurol ; 524(11): 2300-21, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-26713509

RESUMO

The superior colliculus (SC) is a midbrain center involved in controlling head and eye movements in response to inputs from multiple sensory modalities. Visual inputs arise from both the retina and visual cortex and converge onto the superficial layer of the SC (sSC). Neurons in the sSC send information to deeper layers of the SC and to thalamic nuclei that modulate visually guided behaviors. Presently, our understanding of sSC neurons is impeded by a lack of molecular markers that define specific cell types. To better understand the identity and organization of sSC neurons, we took a systematic approach to investigate gene expression within four molecular families: transcription factors, cell adhesion molecules, neuropeptides, and calcium binding proteins. Our analysis revealed 12 molecules with distinct expression patterns in mouse sSC: cadherin 7, contactin 3, netrin G2, cadherin 6, protocadherin 20, retinoid-related orphan receptor ß, brain-specific homeobox/POU domain protein 3b, Ets variant gene 1, substance P, somatostatin, vasoactive intestinal polypeptide, and parvalbumin. Double labeling experiments, by either in situ hybridization or immunostaining, demonstrated that the 12 molecular markers collectively define 10 different sSC neuronal types. The characteristic positions of these cell types divide the sSC into four distinct layers. The 12 markers identified here will serve as valuable tools to examine molecular mechanisms that regulate development of sSC neuronal types. These markers could also be used to examine the connections between specific cell types that form retinocollicular, corticocollicular, or colliculothalamic pathways. J. Comp. Neurol. 524:2300-2321, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Neurônios/classificação , Colículos Superiores/citologia , Animais , Biomarcadores/análise , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Transcriptoma
20.
J Neurosci ; 35(30): 10685-700, 2015 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-26224854

RESUMO

Visual processing in the retina depends on coordinated signaling by interneurons. Photoreceptor signals are relayed to ∼20 ganglion cell types through a dozen excitatory bipolar interneurons, each responsive to light increments (ON) or decrements (OFF). ON and OFF bipolar cell pathways become tuned through specific connections with inhibitory interneurons: horizontal and amacrine cells. A major obstacle for understanding retinal circuitry is the unknown function of most of the ∼30-40 amacrine cell types, each of which synapses onto a subset of bipolar cell terminals, ganglion cell dendrites, and other amacrine cells. Here, we used a transgenic mouse line in which vasoactive intestinal polypeptide-expressing (VIP+) GABAergic interneurons express Cre recombinase. Targeted whole-cell recordings of fluorescently labeled VIP+ cells revealed three predominant types: wide-field bistratified and narrow-field monostratified cells with somas in the inner nuclear layer (INL) and medium-field monostratified cells with somas in the ganglion cell layer (GCL). Bistratified INL cells integrated excitation and inhibition driven by both ON and OFF pathways with little spatial tuning. Narrow-field INL cells integrated excitation driven by the ON pathway and inhibition driven by both pathways, with pronounced hyperpolarizations at light offset. Monostratified GCL cells integrated excitation and inhibition driven by the ON pathway and showed center-surround spatial tuning. Optogenetic experiments showed that, collectively, VIP+ cells made strong connections with OFF δ, ON-OFF direction-selective, and W3 ganglion cells but weak, inconsistent connections with ON and OFF α cells. Revealing VIP+ cell morphologies, receptive fields and synaptic connections advances our understanding of their role in visual processing. SIGNIFICANCE STATEMENT: The retina is a model system for understanding nervous system function. At the first stage, rod and cone photoreceptors encode light and communicate with a complex network of interneurons. These interneurons drive the responses of ganglion cells, which form the optic nerve and transmit visual information to the brain. Presently, we lack information about many of the retina's inhibitory amacrine interneurons. In this study, we used genetically modified mice to study the light responses and intercellular connections of specific amacrine cell types. The results show diversity in the shape and function of the studied amacrine cells and elucidate their connections with specific types of ganglion cell. The findings advance our understanding of the cellular basis for retinal function.


Assuntos
Interneurônios/citologia , Interneurônios/fisiologia , Retina/citologia , Retina/fisiologia , Vias Visuais/citologia , Vias Visuais/fisiologia , Animais , Células Cultivadas , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Optogenética , Técnicas de Patch-Clamp , Peptídeo Intestinal Vasoativo/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...