Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 44(13)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38286625

RESUMO

Modern, high-density neuronal recordings reveal at ever higher precision how information is represented by neural populations. Still, we lack the tools to understand these processes bottom-up, emerging from the biophysical properties of neurons, synapses, and network structure. The concept of the dynamic gain function, a spectrally resolved approximation of a population's coding capability, has the potential to link cell-level properties to network-level performance. However, the concept is not only useful but also very complex because the dynamic gain's shape is co-determined by axonal and somato-dendritic parameters and the population's operating regime. Previously, this complexity precluded an understanding of any individual parameter's impact. Here, we decomposed the dynamic gain function into three components corresponding to separate signal transformations. This allowed attribution of network-level encoding features to specific cell-level parameters. Applying the method to data from real neurons and biophysically plausible models, we found: (1) The encoding bandwidth of real neurons, approximately 400 Hz, is constrained by the voltage dependence of axonal currents during early action potential initiation. (2) State-of-the-art models only achieve encoding bandwidths around 100 Hz and are limited mainly by subthreshold processes instead. (3) Large dendrites and low-threshold potassium currents modulate the bandwidth by shaping the subthreshold stimulus-to-voltage transformation. Our decomposition provides physiological interpretations when the dynamic gain curve changes, for instance during spectrinopathies and neurodegeneration. By pinpointing shortcomings of current models, it also guides inference of neuron models best suited for large-scale network simulations.


Assuntos
Dendritos , Neurônios , Dendritos/fisiologia , Neurônios/fisiologia , Canais Iônicos/fisiologia , Potenciais de Ação/fisiologia , Axônios , Modelos Neurológicos
2.
Learn Mem ; 31(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38862166

RESUMO

Drug addiction and the circuitry for learning and memory are intimately intertwined. Drugs of abuse create strong, inappropriate, and lasting memories that contribute to many of their destructive properties, such as continued use despite negative consequences and exceptionally high rates of relapse. Studies in Drosophila melanogaster are helping us understand how drugs of abuse, especially alcohol, create memories at the level of individual neurons and in the circuits where they function. Drosophila is a premier organism for identifying the mechanisms of learning and memory. Drosophila also respond to drugs of abuse in ways that remarkably parallel humans and rodent models. An emerging consensus is that, for alcohol, the mushroom bodies participate in the circuits that control acute drug sensitivity, not explicitly associative forms of plasticity such as tolerance, and classical associative memories of their rewarding and aversive properties. Moreover, it is becoming clear that drugs of abuse use the mushroom body circuitry differently from other behaviors, potentially providing a basis for their addictive properties.


Assuntos
Memória , Corpos Pedunculados , Animais , Memória/efeitos dos fármacos , Memória/fisiologia , Corpos Pedunculados/fisiologia , Corpos Pedunculados/efeitos dos fármacos , Aprendizagem/fisiologia , Aprendizagem/efeitos dos fármacos , Transtornos Relacionados ao Uso de Substâncias , Drosophila melanogaster/fisiologia , Humanos , Drosophila/fisiologia , Drogas Ilícitas/farmacologia
3.
J Neurosci ; 43(12): 2210-2220, 2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36750369

RESUMO

Ethanol tolerance is the first type of behavioral plasticity and neural plasticity that is induced by ethanol intake, and yet its molecular and circuit bases remain largely unexplored. Here, we characterize the following three distinct forms of ethanol tolerance in male Drosophila: rapid, chronic, and repeated. Rapid tolerance is composed of two short-lived memory-like states, one that is labile and one that is consolidated. Chronic tolerance, induced by continuous exposure, lasts for 2 d, induces ethanol preference, and hinders the development of rapid tolerance through the activity of histone deacetylases (HDACs). Unlike rapid tolerance, chronic tolerance is independent of the immediate early gene Hr38/Nr4a Chronic tolerance is suppressed by the sirtuin HDAC Sirt1, whereas rapid tolerance is enhanced by Sirt1 Moreover, rapid and chronic tolerance map to anatomically distinct regions of the mushroom body learning and memory centers. Chronic tolerance, like long-term memory, is dependent on new protein synthesis and it induces the kayak/c-fos immediate early gene, but it depends on CREB signaling outside the mushroom bodies, and it does not require the Radish GTPase. Thus, chronic ethanol exposure creates an ethanol-specific memory-like state that is molecularly and anatomically different from other forms of ethanol tolerance.SIGNIFICANCE STATEMENT The pattern and concentration of initial ethanol exposure causes operationally distinct types of ethanol tolerance to form. We identify separate molecular and neural circuit mechanisms for two forms of ethanol tolerance, rapid and chronic. We also discover that chronic tolerance forms an ethanol-specific long-term memory-like state that localizes to learning and memory circuits, but it is different from appetitive and aversive long-term memories. By contrast, rapid tolerance is composed of labile and consolidated short-term memory-like states. The multiple forms of ethanol memory-like states are genetically tractable for understanding how initial forms of ethanol-induced neural plasticity form a substrate for the longer-term brain changes associated with alcohol use disorder.


Assuntos
Alcoolismo , Proteínas de Drosophila , Animais , Masculino , Drosophila/metabolismo , Sirtuína 1/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Etanol/farmacologia , Alcoolismo/metabolismo , Corpos Pedunculados/metabolismo , Drosophila melanogaster/genética , Receptores Citoplasmáticos e Nucleares/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34903668

RESUMO

Fast oscillations in cortical circuits critically depend on GABAergic interneurons. Which interneuron types and populations can drive different cortical rhythms, however, remains unresolved and may depend on brain state. Here, we measured the sensitivity of different GABAergic interneurons in prefrontal cortex under conditions mimicking distinct brain states. While fast-spiking neurons always exhibited a wide bandwidth of around 400 Hz, the response properties of spike-frequency adapting interneurons switched with the background input's statistics. Slowly fluctuating background activity, as typical for sleep or quiet wakefulness, dramatically boosted the neurons' sensitivity to gamma and ripple frequencies. We developed a time-resolved dynamic gain analysis and revealed rapid sensitivity modulations that enable neurons to periodically boost gamma oscillations and ripples during specific phases of ongoing low-frequency oscillations. This mechanism predicts these prefrontal interneurons to be exquisitely sensitive to high-frequency ripples, especially during brain states characterized by slow rhythms, and to contribute substantially to theta-gamma cross-frequency coupling.


Assuntos
Ritmo Gama/fisiologia , Interneurônios/fisiologia , Córtex Pré-Frontal/citologia , Ritmo Teta/fisiologia , Animais , Feminino , Masculino , Camundongos , Rede Nervosa/fisiologia , Técnicas de Patch-Clamp
5.
Neuropathol Appl Neurobiol ; 49(5): e12935, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37705188

RESUMO

AIMS: Fibroblast growth factor (FGF) signalling is dysregulated in multiple sclerosis (MS) and other neurological and psychiatric conditions, but there is little or no consensus as to how individual FGF family members contribute to disease pathogenesis. Lesion development in MS is associated with increased expression of FGF1, FGF2 and FGF9, all of which modulate remyelination in a variety of experimental settings. However, FGF9 is also selectively upregulated in major depressive disorder (MDD), prompting us to speculate it may also have a direct effect on neuronal function and survival. METHODS: Transcriptional profiling of myelinating cultures treated with FGF1, FGF2 or FGF9 was performed, and the effects of FGF9 on cortical neurons investigated using a combination of transcriptional, electrophysiological and immunofluorescence microscopic techniques. The in vivo effects of FGF9 were explored by stereotactic injection of adeno-associated viral (AAV) vectors encoding either FGF9 or EGFP into the rat motor cortex. RESULTS: Transcriptional profiling of myelinating cultures after FGF9 treatment revealed a distinct neuronal response with a pronounced downregulation of gene networks associated with axonal transport and synaptic function. In cortical neuronal cultures, FGF9 also rapidly downregulated expression of genes associated with synaptic function. This was associated with a complete block in the development of photo-inducible spiking activity, as demonstrated using multi-electrode recordings of channel rhodopsin-transfected rat cortical neurons in vitro and, ultimately, neuronal cell death. Overexpression of FGF9 in vivo resulted in rapid loss of neurons and subsequent development of chronic grey matter lesions with neuroaxonal reduction and ensuing myelin loss. CONCLUSIONS: These observations identify overexpression of FGF9 as a mechanism by which neuroaxonal pathology could develop independently of immune-mediated demyelination in MS. We suggest targeting neuronal FGF9-dependent pathways may provide a novel strategy to slow if not halt neuroaxonal atrophy and loss in MS, MDD and potentially other neurodegenerative diseases.


Assuntos
Transtorno Depressivo Maior , Esclerose Múltipla , Animais , Ratos , Fator 1 de Crescimento de Fibroblastos , Fator 2 de Crescimento de Fibroblastos , Fator 9 de Crescimento de Fibroblastos
6.
PLoS Comput Biol ; 18(1): e1009775, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35041645

RESUMO

Populations of cortical neurons respond to common input within a millisecond. Morphological features and active ion channel properties were suggested to contribute to this astonishing processing speed. Here we report an exhaustive study of ultrafast population coding for varying axon initial segment (AIS) location, soma size, and axonal current properties. In particular, we studied their impact on two experimentally observed features 1) precise action potential timing, manifested in a wide-bandwidth dynamic gain, and 2) high-frequency boost under slowly fluctuating correlated input. While the density of axonal channels and their distance from the soma had a very small impact on bandwidth, it could be moderately improved by increasing soma size. When the voltage sensitivity of axonal currents was increased we observed ultrafast coding and high-frequency boost. We conclude that these computationally relevant features are strongly dependent on axonal ion channels' voltage sensitivity, but not their number or exact location. We point out that ion channel properties, unlike dendrite size, can undergo rapid physiological modification, suggesting that the temporal accuracy of neuronal population encoding could be dynamically regulated. Our results are in line with recent experimental findings in AIS pathologies and establish a framework to study structure-function relations in AIS molecular design.


Assuntos
Potenciais de Ação/fisiologia , Axônios/fisiologia , Modelos Neurológicos , Neurônios/fisiologia , Biologia Computacional , Canais Iônicos/metabolismo
7.
Addict Biol ; 28(8): e13304, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37500483

RESUMO

Alcohol tolerance is a simple form of behavioural and neural plasticity that occurs with the first drink. Neural plasticity in tolerance is likely a substrate for longer term adaptations that can lead to alcohol use disorder. Drosophila develop tolerance with characteristics similar to vertebrates, and it is a useful model for determining the molecular and circuit encoding mechanisms in detail. Rapid tolerance, measured after the first alcohol exposure is completely metabolized, is localized to specific brain regions that are not interconnected in an obvious way. We used a forward neuroanatomical screen to identify three new neural sites for rapid tolerance encoding. One of these was composed of two groups of neurons, the DN1a and DN1p glutamatergic neurons, that are part of the Drosophila circadian clock. We localized rapid tolerance to the two DN1a neurons that regulate arousal by light at night, temperature-dependent sleep timing, and night-time sleep. Two clock neurons that regulate evening activity, LNd6 and the 5th LNv, are postsynaptic to the DN1as, and they promote rapid tolerance via the metabotropic glutamate receptor. Thus, rapid tolerance to alcohol overlaps with sleep regulatory neural circuitry, suggesting a mechanistic link.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ritmo Circadiano , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Sono , Neurônios/metabolismo
8.
EMBO J ; 37(24)2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30396994

RESUMO

Optogenetic tools, providing non-invasive control over selected cells, have the potential to revolutionize sensory prostheses for humans. Optogenetic stimulation of spiral ganglion neurons (SGNs) in the ear provides a future alternative to electrical stimulation used in cochlear implants. However, most channelrhodopsins do not support the high temporal fidelity pertinent to auditory coding because they require milliseconds to close after light-off. Here, we biophysically characterized the fast channelrhodopsin Chronos and revealed a deactivation time constant of less than a millisecond at body temperature. In order to enhance neural expression, we improved its trafficking to the plasma membrane (Chronos-ES/TS). Following efficient transduction of SGNs using early postnatal injection of the adeno-associated virus AAV-PHPB into the mouse cochlea, fiber-based optical stimulation elicited optical auditory brainstem responses (oABR) with minimal latencies of 1 ms, thresholds of 5 µJ and 100 µs per pulse, and sizable amplitudes even at 1,000 Hz of stimulation. Recordings from single SGNs demonstrated good temporal precision of light-evoked spiking. In conclusion, efficient virus-mediated expression of targeting-optimized Chronos-ES/TS achieves ultrafast optogenetic control of neurons.


Assuntos
Channelrhodopsins/biossíntese , Dependovirus , Expressão Gênica , Neurônios/metabolismo , Optogenética , Gânglio Espiral da Cóclea/metabolismo , Transdução Genética , Animais , Tronco Encefálico/metabolismo , Channelrhodopsins/genética , Potenciais Evocados Auditivos , Células HEK293 , Humanos , Camundongos , Ratos , Ratos Wistar
9.
PLoS Comput Biol ; 16(1): e1007588, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31917809

RESUMO

Real-world agents, humans as well as animals, observe each other during interactions and choose their own actions taking the partners' ongoing behaviour into account. Yet, classical game theory assumes that players act either strictly sequentially or strictly simultaneously without knowing each other's current choices. To account for action visibility and provide a more realistic model of interactions under time constraints, we introduce a new game-theoretic setting called transparent games, where each player has a certain probability of observing the partner's choice before deciding on its own action. By means of evolutionary simulations, we demonstrate that even a small probability of seeing the partner's choice before one's own decision substantially changes the evolutionary successful strategies. Action visibility enhances cooperation in an iterated coordination game, but reduces cooperation in a more competitive iterated Prisoner's Dilemma. In both games, "Win-stay, lose-shift" and "Tit-for-tat" strategies are predominant for moderate transparency, while a "Leader-Follower" strategy emerges for high transparency. Our results have implications for studies of human and animal social behaviour, especially for the analysis of dyadic and group interactions.


Assuntos
Tomada de Decisões/fisiologia , Teoria dos Jogos , Relações Interpessoais , Modelos Biológicos , Animais , Comportamento Animal , Biologia Computacional , Comportamento Cooperativo , Humanos
10.
EMBO Rep ; 20(12): e47755, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31663248

RESUMO

The spatial and temporal dynamics of cell contractility plays a key role in tissue morphogenesis, wound healing, and cancer invasion. Here, we report a simple optochemical method to induce cell contractions in vivo during Drosophila morphogenesis at single-cell resolution. We employed the photolabile Ca2+ chelator o-nitrophenyl EGTA to induce bursts of intracellular free Ca2+ by laser photolysis in the epithelial tissue. Ca2+ bursts appear within seconds and are restricted to individual target cells. Cell contraction reliably followed within a minute, causing an approximately 50% drop in the cross-sectional area. Increased Ca2+ levels are reversible, and the target cells further participated in tissue morphogenesis. Depending on Rho kinase (ROCK) activity but not RhoGEF2, cell contractions are paralleled with non-muscle myosin II accumulation in the apico-medial cortex, indicating that Ca2+ bursts trigger non-muscle myosin II activation. Our approach can be, in principle, adapted to many experimental systems and species, as no specific genetic elements are required.


Assuntos
Drosophila melanogaster/citologia , Células Epiteliais/fisiologia , Animais , Animais Geneticamente Modificados , Fenômenos Biomecânicos , Quelantes de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Forma Celular/efeitos dos fármacos , Forma Celular/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Miosina Tipo II/fisiologia , Processos Fotoquímicos , Análise de Célula Única , Análise Espaço-Temporal
11.
Proc Natl Acad Sci U S A ; 115(30): E7184-E7192, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29991598

RESUMO

Neocortical pyramidal neurons express several distinct subtypes of voltage-gated Na+ channels. In mature cells, Nav1.6 is the dominant channel subtype in the axon initial segment (AIS) as well as in the nodes of Ranvier. Action potentials (APs) are initiated in the AIS, and it has been proposed that the high excitability of this region is related to the unique characteristics of the Nav1.6 channel. Knockout or loss-of-function mutation of the Scn8a gene is generally lethal early in life because of the importance of this subtype in noncortical regions of the nervous system. Using the Cre/loxP system, we selectively deleted Nav1.6 in excitatory neurons of the forebrain and characterized the excitability of Nav1.6-deficient layer 5 pyramidal neurons by patch-clamp and Na+ and Ca2+ imaging recordings. We now report that, in the absence of Nav1.6 expression, the AIS is occupied by Nav1.2 channels. However, APs are generated in the AIS, and differences in AP propagation to soma and dendrites are minimal. Moreover, the channels that are expressed in the AIS still show a clear hyperpolarizing shift in voltage dependence of activation, compared with somatic channels. The only major difference between Nav1.6-null and wild-type neurons was a strong reduction in persistent sodium current. We propose that the molecular environment of the AIS confers properties on whatever Na channel subtype is present and that some other benefit must be conferred by the selective axonal presence of the Nav1.6 channel.


Assuntos
Potenciais de Ação/fisiologia , Axônios/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Neocórtex/metabolismo , Células Piramidais/metabolismo , Animais , Deleção de Genes , Camundongos , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.2/genética , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Neocórtex/citologia , Células Piramidais/citologia
12.
J Neurosci ; 39(39): 7790-7800, 2019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31399533

RESUMO

Cortical regions that are damaged by insults, such as ischemia, hypoxia, and trauma, frequently generate spreading depolarization (SD). At the neuronal level, SDs entail complete breakdown of ionic gradients, persisting for seconds to minutes. It is unclear whether these transient events have a more lasting influence on neuronal function. Here, we describe electrophysiological changes in cortical neurons after recovery from hypoxia-induced SD. When examined with standard measures of neuronal excitability several hours after recovery from SD, layer 5 pyramidal neurons in brain slices from mice of either sex appear surprisingly normal. However, we here introduce an additional parameter, dynamic gain, which characterizes the bandwidth of action potential encoding by a neuron, and thereby reflects its potential efficiency in a multineuronal circuit. We find that the ability of neurons that recover from SD to track high-frequency inputs is markedly curtailed; exposure to hypoxia did not have this effect when SD was prevented pharmacologically. Staining for Ankyrin G revealed at least a fourfold decrease in the number of intact axon initial segments in post-SD slices. Since this effect, along with the effect on encoding, was blocked by an inhibitor of the Ca2+-dependent enzyme, calpain, we conclude that both effects were mediated by the SD-induced rise in intracellular Ca2+ Although effects of calpain activation were detected in the axon initial segment, changes in soma-dendritic compartments may also be involved. Whatever the precise molecular mechanism, our findings indicate that in the context of cortical circuit function, effectiveness of neurons that survive SD may be limited.SIGNIFICANCE STATEMENT Spreading depolarization, which commonly accompanies cortical injury, entails transient massive breakdown of neuronal ionic gradients. The function of cortical neurons that recover from hypoxia-induced spreading depolarization is not obviously abnormal when tested for usual measures of neuronal excitability. However, we now demonstrate that they have a reduced bandwidth, reflecting a significant impairment of their ability to precisely encode high-frequency components of their synaptic input in output spike trains. Thus, neurons that recover from spreading depolarizations are less able to function normally as elements in the multineuronal cortical circuitry. These changes are correlated with activation of the calcium-dependent enzyme, calpain.


Assuntos
Calpaína/metabolismo , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Hipóxia Encefálica/fisiopatologia , Modelos Neurológicos , Neurônios/fisiologia , Potenciais de Ação/fisiologia , Animais , Feminino , Hipóxia Encefálica/metabolismo , Masculino , Camundongos
13.
EMBO J ; 33(3): 247-64, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24442635

RESUMO

Cochlear inner hair cells (IHCs) develop from pre-sensory pacemaker to sound transducer. Here, we report that this involves changes in structure and function of the ribbon synapses between IHCs and spiral ganglion neurons (SGNs) around hearing onset in mice. As synapses matured they changed from holding several small presynaptic active zones (AZs) and apposed postsynaptic densities (PSDs) to one large AZ/PSD complex per SGN bouton. After the onset of hearing (i) IHCs had fewer and larger ribbons; (ii) CaV1.3 channels formed stripe-like clusters rather than the smaller and round clusters at immature AZs; (iii) extrasynaptic CaV1.3-channels were selectively reduced, (iv) the intrinsic Ca(2)(+) dependence of fast exocytosis probed by Ca(2)(+) uncaging remained unchanged but (v) the apparent Ca(2)(+) dependence of exocytosis linearized, when assessed by progressive dihydropyridine block of Ca(2)(+) influx. Biophysical modeling of exocytosis at mature and immature AZ topographies suggests that Ca(2)(+) influx through an individual channel dominates the [Ca(2)(+)] driving exocytosis at each mature release site. We conclude that IHC synapses undergo major developmental refinements, resulting in tighter spatial coupling between Ca(2)(+) influx and exocytosis.


Assuntos
Cálcio/metabolismo , Exocitose/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Modelos Neurológicos , Gânglio Espiral da Cóclea/fisiologia , Sinapses/fisiologia , Animais , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Eletrofisiologia , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas Internas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Mutação , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/ultraestrutura , Gânglio Espiral da Cóclea/citologia , Sinapses/ultraestrutura
14.
Proc Natl Acad Sci U S A ; 112(9): E1028-37, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25691754

RESUMO

EF-hand Ca(2+)-binding proteins are thought to shape the spatiotemporal properties of cellular Ca(2+) signaling and are prominently expressed in sensory hair cells in the ear. Here, we combined genetic disruption of parvalbumin-α, calbindin-D28k, and calretinin in mice with patch-clamp recording, in vivo physiology, and mathematical modeling to study their role in Ca(2+) signaling, exocytosis, and sound encoding at the synapses of inner hair cells (IHCs). IHCs lacking all three proteins showed excessive exocytosis during prolonged depolarizations, despite enhanced Ca(2+)-dependent inactivation of their Ca(2+) current. Exocytosis of readily releasable vesicles remained unchanged, in accordance with the estimated tight spatial coupling of Ca(2+) channels and release sites (effective "coupling distance" of 17 nm). Substitution experiments with synthetic Ca(2+) chelators indicated the presence of endogenous Ca(2+) buffers equivalent to 1 mM synthetic Ca(2+)-binding sites, approximately half of them with kinetics as fast as 1,2-Bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA). Synaptic sound encoding was largely unaltered, suggesting that excess exocytosis occurs extrasynaptically. We conclude that EF-hand Ca(2+) buffers regulate presynaptic IHC function for metabolically efficient sound coding.


Assuntos
Calbindina 1/metabolismo , Calbindina 2/metabolismo , Sinalização do Cálcio/fisiologia , Exocitose/fisiologia , Células Ciliadas Auditivas Internas/metabolismo , Parvalbuminas/metabolismo , Animais , Calbindina 1/genética , Calbindina 2/genética , Sinalização do Cálcio/efeitos dos fármacos , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Exocitose/efeitos dos fármacos , Células Ciliadas Auditivas Internas/citologia , Audição/efeitos dos fármacos , Audição/fisiologia , Camundongos , Camundongos Knockout , Parvalbuminas/genética , Sinapses/genética , Sinapses/metabolismo
15.
J Neurosci ; 36(19): 5241-51, 2016 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-27170122

RESUMO

UNLABELLED: Acute ethanol inebriation causes neuroadaptive changes in behavior that favor increased intake. Ethanol-induced alterations in gene expression, through epigenetic and other means, are likely to change cellular and neural circuit function. Ethanol markedly changes histone acetylation, and the sirtuin Sir2/SIRT1 that deacetylates histones and transcription factors is essential for the rewarding effects of long-term drug use. The molecular transformations leading from short-term to long-term ethanol responses mostly remain to be discovered. We find that Sir2 in the mushroom bodies of the fruit fly Drosophila promotes short-term ethanol-induced behavioral plasticity by allowing changes in the expression of presynaptic molecules. Acute inebriation strongly reduces Sir2 levels and increases histone H3 acetylation in the brain. Flies lacking Sir2 globally, in the adult nervous system, or specifically in the mushroom body α/ß-lobes show reduced ethanol sensitivity and tolerance. Sir2-dependent ethanol reward is also localized to the mushroom bodies, and Sir2 mutants prefer ethanol even without a priming ethanol pre-exposure. Transcriptomic analysis reveals that specific presynaptic molecules, including the synaptic vesicle pool regulator Synapsin, depend on Sir2 to be regulated by ethanol. Synapsin is required for ethanol sensitivity and tolerance. We propose that the regulation of Sir2/SIRT1 by acute inebriation forms part of a transcriptional program in mushroom body neurons to alter presynaptic properties and neural responses to favor the development of ethanol tolerance, preference, and reward. SIGNIFICANCE STATEMENT: We identify a mechanism by which acute ethanol inebriation leads to changes in nervous system function that may be an important basis for increasing ethanol intake and addiction liability. The findings are significant because they identify ethanol-driven transcriptional events that target presynaptic properties and direct behavioral plasticity. They also demonstrate that multiple forms of ethanol behavioral plasticity that are relevant to alcoholism are initiated by a shared mechanism. Finally, they link these events to the Drosophila brain region that associates context with innate approach and avoidance responses to code for reward and other higher-order behavior, similar in aspects to the role of the vertebrate mesolimbic system.


Assuntos
Intoxicação Alcoólica/metabolismo , Alcoolismo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Histona Desacetilases/metabolismo , Terminações Pré-Sinápticas/metabolismo , Recompensa , Sirtuínas/metabolismo , Intoxicação Alcoólica/genética , Alcoolismo/genética , Animais , Drosophila/genética , Drosophila/fisiologia , Proteínas de Drosophila/genética , Histona Desacetilases/genética , Histonas/metabolismo , Corpos Pedunculados/metabolismo , Terminações Pré-Sinápticas/fisiologia , Sirtuínas/genética , Sinapsinas/genética , Sinapsinas/metabolismo , Transcriptoma
16.
PLoS Comput Biol ; 11(12): e1004636, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26720924

RESUMO

The response of a neuronal population over a space of inputs depends on the intrinsic properties of its constituent neurons. Two main modes of single neuron dynamics-integration and resonance-have been distinguished. While resonator cell types exist in a variety of brain areas, few models incorporate this feature and fewer have investigated its effects. To understand better how a resonator's frequency preference emerges from its intrinsic dynamics and contributes to its local area's population firing rate dynamics, we analyze the dynamic gain of an analytically solvable two-degree of freedom neuron model. In the Fokker-Planck approach, the dynamic gain is intractable. The alternative Gauss-Rice approach lifts the resetting of the voltage after a spike. This allows us to derive a complete expression for the dynamic gain of a resonator neuron model in terms of a cascade of filters on the input. We find six distinct response types and use them to fully characterize the routes to resonance across all values of the relevant timescales. We find that resonance arises primarily due to slow adaptation with an intrinsic frequency acting to sharpen and adjust the location of the resonant peak. We determine the parameter regions for the existence of an intrinsic frequency and for subthreshold and spiking resonance, finding all possible intersections of the three. The expressions and analysis presented here provide an account of how intrinsic neuron dynamics shape dynamic population response properties and can facilitate the construction of an exact theory of correlations and stability of population activity in networks containing populations of resonator neurons.


Assuntos
Potenciais de Ação/fisiologia , Modelos Neurológicos , Neurônios/fisiologia , Biologia Computacional , Simulação por Computador , Potenciais da Membrana/fisiologia
17.
PLoS Comput Biol ; 11(11): e1004602, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26575467

RESUMO

The architecture of iso-orientation domains in the primary visual cortex (V1) of placental carnivores and primates apparently follows species invariant quantitative laws. Dynamical optimization models assuming that neurons coordinate their stimulus preferences throughout cortical circuits linking millions of cells specifically predict these invariants. This might indicate that V1's intrinsic connectome and its functional architecture adhere to a single optimization principle with high precision and robustness. To validate this hypothesis, it is critical to closely examine the quantitative predictions of alternative candidate theories. Random feedforward wiring within the retino-cortical pathway represents a conceptually appealing alternative to dynamical circuit optimization because random dimension-expanding projections are believed to generically exhibit computationally favorable properties for stimulus representations. Here, we ask whether the quantitative invariants of V1 architecture can be explained as a generic emergent property of random wiring. We generalize and examine the stochastic wiring model proposed by Ringach and coworkers, in which iso-orientation domains in the visual cortex arise through random feedforward connections between semi-regular mosaics of retinal ganglion cells (RGCs) and visual cortical neurons. We derive closed-form expressions for cortical receptive fields and domain layouts predicted by the model for perfectly hexagonal RGC mosaics. Including spatial disorder in the RGC positions considerably changes the domain layout properties as a function of disorder parameters such as position scatter and its correlations across the retina. However, independent of parameter choice, we find that the model predictions substantially deviate from the layout laws of iso-orientation domains observed experimentally. Considering random wiring with the currently most realistic model of RGC mosaic layouts, a pairwise interacting point process, the predicted layouts remain distinct from experimental observations and resemble Gaussian random fields. We conclude that V1 layout invariants are specific quantitative signatures of visual cortical optimization, which cannot be explained by generic random feedforward-wiring models.


Assuntos
Modelos Neurológicos , Células Ganglionares da Retina/fisiologia , Córtex Visual/citologia , Córtex Visual/fisiologia , Animais , Biologia Computacional , Mamíferos , Rede Nervosa/fisiologia
18.
Dev Biol ; 390(2): 208-20, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24681004

RESUMO

The majority of membrane and secreted proteins, including many developmentally important signalling proteins, receptors and adhesion molecules, are cotranslationally N-glycosylated in the endoplasmic reticulum. The structure of the N-glycan is invariant for all substrates and conserved in eukaryotes. Correspondingly, the enzymes are conserved, which successively assemble the glycan precursor from activated monosaccharides prior to transfer to nascent proteins. Despite the well-defined biochemistry, the physiological and developmental role of N-glycosylation and of the responsible enzymes has not been much investigated in metazoa. We identified a mutation in the Drosophila gene, xiantuan (xit, CG4542), which encodes one of the conserved enzymes involved in addition of the terminal glucose residues to the glycan precursor. xit is required for timely apical constriction of mesoderm precursor cells and ventral furrow formation in early embryogenesis. Furthermore, cell intercalation in the lateral epidermis during germband extension is impaired in xit mutants. xit affects glycosylation and intracellular distribution of E-Cadherin, albeit not the total amount of E-Cadherin protein. As depletion of E-Cadherin by RNAi induces a similar cell intercalation defect, E-Cadherin may be the major xit target that is functionally relevant for germband extension.


Assuntos
Caderinas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Retículo Endoplasmático/metabolismo , Gastrulação/fisiologia , Regulação da Expressão Gênica/fisiologia , Glucosiltransferases/metabolismo , Movimento/fisiologia , Junções Aderentes/fisiologia , Animais , Sequência de Bases , Western Blotting , Drosophila/enzimologia , Proteínas de Drosophila/genética , Fluorescência , Componentes do Gene , Regulação da Expressão Gênica/genética , Glucosiltransferases/genética , Microscopia Confocal , Dados de Sequência Molecular , Interferência de RNA , Análise de Sequência de DNA
19.
J Comput Neurosci ; 39(1): 63-75, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26001536

RESUMO

Somatic action potentials (AP) of cortical pyramidal neurons have characteristically high onset-rapidness. The onset of the AP waveform is an indirect measure for the ability of a neuron to respond to temporally fast-changing stimuli. Theoretical studies on the pyramidal neuron response usually involves a canonical Hodgkin-Huxley (HH) type ion channel gating model, which assumes statistically independent gating of each individual channel. However, cooperative activity of ion channels are observed for various cell types, meaning that the activity (e.g. opening) of one channel triggers the activity (e.g. opening) of a certain fraction of its neighbors and hence, these groups of channels behave as a unit. In this study, we describe a multi-compartmental conductance-based model with cooperatively gating voltage-gated Na channels in the axon initial segment. Our model successfully reproduced the somatic sharp AP onsets of cortical pyramidal neurons. The onset latencies from the initiation site to the soma and the conduction velocities were also in agreement with the previous experimental studies.


Assuntos
Potenciais de Ação/fisiologia , Axônios/fisiologia , Ativação do Canal Iônico/fisiologia , Modelos Neurológicos , Neurônios/fisiologia , Canais de Sódio/fisiologia , Animais , Estimulação Elétrica , Humanos , Neurônios/citologia
20.
J Neurosci ; 33(6): 2281-92, 2013 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-23392659

RESUMO

The abilities of neuronal populations to encode rapidly varying stimuli and respond quickly to abrupt input changes are crucial for basic neuronal computations, such as coincidence detection, grouping by synchrony, and spike-timing-dependent plasticity, as well as for the processing speed of neuronal networks. Theoretical analyses have linked these abilities to the fast-onset dynamics of action potentials (APs). Using a combination of whole-cell recordings from rat neocortical neurons and computer simulations, we provide the first experimental evidence for this conjecture and prove its validity for the case of distal AP initiation in the axon initial segment (AIS), typical for cortical neurons. Neocortical neurons with fast-onset APs in the soma can phase-lock their population firing to signal frequencies up to ∼300-400 Hz and respond within 1-2 ms to subtle changes of input current. The ability to encode high frequencies and response speed were dramatically reduced when AP onset was slowed by experimental manipulations or was intrinsically slow due to immature AP generation mechanisms. Multicompartment conductance-based models reproducing the initiation of spikes in the AIS could encode high frequencies only if AP onset was fast at the initiation site (e.g., attributable to cooperative gating of a fraction of sodium channels) but not when fast onset of somatic AP was produced solely by backpropagation. We conclude that fast-onset dynamics is a genuine property of cortical AP generators. It enables fast computations in cortical circuits that are rich in recurrent connections both within each region and across the hierarchy of areas.


Assuntos
Potenciais de Ação/fisiologia , Simulação por Computador , Modelos Neurológicos , Córtex Visual/fisiologia , Animais , Feminino , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA