Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-37961167

RESUMO

Sleep disturbances are associated with poor long-term memory (LTM) formation, yet the underlying cell types and neural circuits involved have not been fully decoded. Dopamine neurons (DANs) are involved in memory processing at multiple stages. Here, we show that brief activation of protocerebral anterior medial DANs (PAM-DANs) or inhibition of a pair of dorsal posterior medial (DPM) neurons during the first few hours of memory consolidation impairs 24 h LTM. Interestingly, sleep deprivation elevates the neural activity of PAM-DANs and DPM neurons, and brief thermos-activation of PAM-DANs or inactivation of DPM neurons results in sleep loss and fragmentation. Pharmacological rescue of sleep after this manipulation restores LTM. A specific subset of PAM-DANs, PAM-α1 that synapse onto DPM neurons specify the microcircuit that links sleep and memory. PAM-DANs, including PAM-α1, form functional synapses with DPM neurons mainly via Dop1R1 receptor to inhibit DPM. Our data suggest that the post-training activity of PAM(-α1)-DPM microcircuit, especially during memory consolidation, plays an essential role in maintaining the sleep necessary for LTM consolidation, providing a new cellular and circuit basis for the complex relationship between sleep and memory.

2.
Sci Adv ; 9(22): eadg9836, 2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37267358

RESUMO

While neurotransmitter identity was once considered singular and immutable for mature neurons, it is now appreciated that one neuron can release multiple neuroactive substances (cotransmission) whose identities can even change over time. To explore the mechanisms that tune the suite of transmitters a neuron releases, we developed transcriptional and translational reporters for cholinergic, glutamatergic, and GABAergic signaling in Drosophila. We show that many glutamatergic and GABAergic cells also transcribe cholinergic genes, but fail to accumulate cholinergic effector proteins. Suppression of cholinergic signaling involves posttranscriptional regulation of cholinergic transcripts by the microRNA miR-190; chronic loss of miR-190 function allows expression of cholinergic machinery, reducing and fragmenting sleep. Using a "translation-trap" strategy, we show that neurons in these populations have episodes of transient translation of cholinergic proteins, demonstrating that suppression of cotransmission is actively modulated. Posttranscriptional restriction of fast transmitter cotransmission provides a mechanism allowing reversible tuning of neuronal output.


Assuntos
MicroRNAs , Neurônios , Neurônios/metabolismo , Transmissão Sináptica/genética , Sono/fisiologia , Colinérgicos , MicroRNAs/genética , MicroRNAs/metabolismo
3.
bioRxiv ; 2023 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-36909471

RESUMO

While neurotransmitter identity was once considered singular and immutable for mature neurons, it is now appreciated that one neuron can release multiple neuroactive substances (co-transmission) whose identities can even change over time. To explore the mechanisms that tune the suite of transmitters a neuron releases, we developed transcriptional and translational reporters for cholinergic, glutamatergic, and GABAergic signaling in Drosophila . We show that many glutamatergic and GABAergic cells also transcribe cholinergic genes, but fail to accumulate cholinergic effector proteins. Suppression of cholinergic signaling involves posttranscriptional regulation of cholinergic transcripts by the microRNA miR-190; chronic loss of miR-190 function allows expression of cholinergic machinery, reducing and fragmenting sleep. Using a "translation-trap" strategy we show that neurons in these populations have episodes of transient translation of cholinergic proteins, demonstrating that suppression of co-transmission is actively modulated. Posttranscriptional restriction of fast transmitter co-transmission provides a mechanism allowing reversible tuning of neuronal output. One-Sentence Summary: Cholinergic co-transmission in large populations of glutamatergic and GABAergic neurons in the Drosophila adult brain is controlled by miR-190.

4.
J Neurosci ; 43(5): 764-786, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36535771

RESUMO

The ellipsoid body (EB) is a major structure of the central complex of the Drosophila melanogaster brain. Twenty-two subtypes of EB ring neurons have been identified based on anatomic and morphologic characteristics by light-level microscopy and EM connectomics. A few studies have associated ring neurons with the regulation of sleep homeostasis and structure. However, cell type-specific and population interactions in the regulation of sleep remain unclear. Using an unbiased thermogenetic screen of EB drivers using female flies, we found the following: (1) multiple ring neurons are involved in the modulation of amount of sleep and structure in a synergistic manner; (2) analysis of data for ΔP(doze)/ΔP(wake) using a mixed Gaussian model detected 5 clusters of GAL4 drivers which had similar effects on sleep pressure and/or depth: lines driving arousal contained R4m neurons, whereas lines that increased sleep pressure had R3m cells; (3) a GLM analysis correlating ring cell subtype and activity-dependent changes in sleep parameters across all lines identified several cell types significantly associated with specific sleep effects: R3p was daytime sleep-promoting, and R4m was nighttime wake-promoting; and (4) R3d cells present in 5HT7-GAL4 and in GAL4 lines, which exclusively affect sleep structure, were found to contribute to fragmentation of sleep during both day and night. Thus, multiple subtypes of ring neurons distinctively control sleep amount and/or structure. The unique highly interconnected structure of the EB suggests a local-network model worth future investigation; understanding EB subtype interactions may provide insight how sleep circuits in general are structured.SIGNIFICANCE STATEMENT How multiple brain regions, with many cell types, can coherently regulate sleep remains unclear, but identification of cell type-specific roles can generate opportunities for understanding the principles of integration and cooperation. The ellipsoid body (EB) of the fly brain exhibits a high level of connectivity and functional heterogeneity yet is able to tune multiple behaviors in real-time, including sleep. Leveraging the powerful genetic tools available in Drosophila and recent progress in the characterization of the morphology and connectivity of EB ring neurons, we identify several EB subtypes specifically associated with distinct aspects of sleep. Our findings will aid in revealing the rules of coding and integration in the brain.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Feminino , Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Sono/fisiologia , Neurônios/fisiologia , Nível de Alerta/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
5.
iScience ; 25(9): 104874, 2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36034229

RESUMO

Sleep circuitry evolved to have both dedicated and context-dependent modulatory elements. Identifying modulatory subcircuits and understanding their molecular machinery is a major challenge for the sleep field. Previously, we identified 25 sleep-regulating microRNAs in Drosophila melanogaster, including the developmentally important microRNA bantam. Here we show that bantam acts in the adult to promote early nighttime sleep through a population of glutamatergic neurons that is intimately involved in applying contextual information to behaviors, the γ5ß'2a/ß'2mp/ß'2mp_bilateral Mushroom Body Output Neurons (MBONs). Calcium imaging revealed that bantam inhibits the activity of these cells during the early night, but not the day. Blocking synaptic transmission in these MBONs rescued the effect of bantam knockdown. This suggests bantam promotes early night sleep via inhibition of the γ5ß'2a/ß'2mp/ß'2mp_bilateral MBONs. RNAseq identifies Kelch and CCHamide-2 receptor as possible mediators, establishing a new role for bantam as an active regulator of sleep and neural activity in the adult fly.

6.
Curr Biol ; 32(12): 2730-2738.e5, 2022 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-35545085

RESUMO

How compartment-specific local proteomes are generated and maintained is inadequately understood, particularly in neurons, which display extreme asymmetries. Here we show that local enrichment of Ca2+/calmodulin-dependent protein kinase II (CaMKII) in axons of Drosophila mushroom body neurons is necessary for cellular plasticity and associative memory formation. Enrichment is achieved via enhanced axoplasmic translation of CaMKII mRNA, through a mechanism requiring the RNA-binding protein Mub and a 23-base Mub-recognition element in the CaMKII 3' UTR. Perturbation of either dramatically reduces axonal, but not somatic, CaMKII protein without altering the distribution or amount of mRNA in vivo, and both are necessary and sufficient to enhance axonal translation of reporter mRNA. Together, these data identify elevated levels of translation of an evenly distributed mRNA as a novel strategy for generating subcellular biochemical asymmetries. They further demonstrate the importance of distributional asymmetry in the computational and biological functions of neurons.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Neurônios , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Drosophila/genética , Corpos Pedunculados/metabolismo , Neurônios/metabolismo , RNA Mensageiro/metabolismo
7.
J Neurosci ; 42(21): 4297-4310, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35474278

RESUMO

In Drosophila, in vivo functional imaging studies revealed that associative memory formation is coupled to a cascade of neural plasticity events in distinct compartments of the mushroom body (MB). In-depth investigation of the circuit dynamics, however, will require an ex vivo model that faithfully mirrors these events to allow direct manipulations of circuit elements that are inaccessible in the intact fly. The current ex vivo models have been able to reproduce the fundamental plasticity of aversive short-term memory, a potentiation of the MB intrinsic neuron (Kenyon cells [KCs]) responses after artificial learning ex vivo However, this potentiation showed different localization and encoding properties from those reported in vivo and failed to generate the previously reported suppression plasticity in the MB output neurons (MBONs). Here, we develop an ex vivo model using the female Drosophila brain that recapitulates behaviorally evoked plasticity in the KCs and MBONs. We demonstrate that this plasticity accurately localizes to the MB α'3 compartment and is encoded by a coincidence between KC activation and dopaminergic input. The formed plasticity is input-specific, requiring pairing of the conditioned stimulus and unconditioned stimulus pathways; hence, we name it pairing-dependent plasticity. Pairing-dependent plasticity formation requires an intact CaMKII gene and is blocked by previous-night sleep deprivation but is rescued by rebound sleep. In conclusion, we show that our ex vivo preparation recapitulates behavioral and imaging results from intact animals and can provide new insights into mechanisms of memory formation at the level of molecules, circuits, and brain state.SIGNIFICANCE STATEMENT The mammalian ex vivo LTP model enabled in-depth investigation of the hippocampal memory circuit. We develop a parallel model to study the Drosophila mushroom body (MB) memory circuit. Pairing activation of the conditioned stimulus and unconditioned stimulus pathways in dissected brains induces a potentiation pairing-dependent plasticity (PDP) in the axons of α'ß' Kenyon cells and a suppression PDP in the dendrites of their postsynaptic MB output neurons, localized in the MB α'3 compartment. This PDP is input-specific and requires the 3' untranslated region of CaMKII Interestingly, ex vivo PDP carries information about the animal's experience before dissection; brains from sleep-deprived animals fail to form PDP, whereas those from animals who recovered 2 h of their lost sleep form PDP.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Drosophila melanogaster , Animais , Encéfalo , Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Feminino , Mamíferos , Corpos Pedunculados/fisiologia , Sono/fisiologia
8.
Front Behav Neurosci ; 15: 681593, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34220464

RESUMO

Maladaptive operant conditioning contributes to development of neuropsychiatric disorders. Candidate genes have been identified that contribute to this maladaptive plasticity, but the neural basis of operant conditioning in genetic model organisms remains poorly understood. The fruit fly Drosophila melanogaster is a versatile genetic model organism that readily forms operant associations with punishment stimuli. However, operant conditioning with a food reward has not been demonstrated in flies, limiting the types of neural circuits that can be studied. Here we present the first sucrose-reinforced operant conditioning paradigm for flies. In the paradigm, flies walk along a Y-shaped track with reward locations at the terminus of each hallway. When flies turn in the reinforced direction at the center of the track, they receive a sucrose reward at the end of the hallway. Only flies that rest early in training learn the reward contingency normally. Flies rewarded independently of their behavior do not form a learned association but have the same amount of rest as trained flies, showing that rest is not driven by learning. Optogenetically-induced sleep does not promote learning, indicating that sleep itself is not sufficient for learning the operant task. We validated the sensitivity of this assay to detect the effect of genetic manipulations by testing the classic learning mutant dunce. Dunce flies are learning-impaired in the Y-Track task, indicating a likely role for cAMP in the operant coincidence detector. This novel training paradigm will provide valuable insight into the molecular mechanisms of disease and the link between sleep and learning.

9.
Neurosci Bull ; 37(6): 831-852, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33779893

RESUMO

Learning to associate a positive or negative experience with an unrelated cue after the presentation of a reward or a punishment defines associative learning. The ability to form associative memories has been reported in animal species as complex as humans and as simple as insects and sea slugs. Associative memory has even been reported in tardigrades [1], species that diverged from other animal phyla 500 million years ago. Understanding the mechanisms of memory formation is a fundamental goal of neuroscience research. In this article, we work on resolving the current contradictions between different Drosophila associative memory circuit models and propose an updated version of the circuit model that predicts known memory behaviors that current models do not. Finally, we propose a model for how dopamine may function as a reward prediction error signal in Drosophila, a dopamine function that is well-established in mammals but not in insects [2, 3].


Assuntos
Dopamina , Drosophila , Animais , Condicionamento Clássico , Humanos , Recompensa
10.
Nature ; 589(7843): 582-585, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33268891

RESUMO

Sleep remains a major mystery of biology, with little understood about its basic function. One of the most commonly proposed functions of sleep is the consolidation of memory1-3. However, as conditions such as starvation require the organism to be awake and active4, the ability to switch to a memory consolidation mechanism that is not contingent on sleep may confer an evolutionary advantage. Here we identify an adaptive circuit-based mechanism that enables Drosophila to form sleep-dependent and sleep-independent memory. Flies fed after appetitive conditioning needed increased sleep for memory consolidation, but flies starved after training did not require sleep to form memories. Memory in fed flies is mediated by the anterior-posterior α'/ß' neurons of the mushroom body, while memory under starvation is mediated by medial α'/ß' neurons. Sleep-dependent and sleep-independent memory rely on distinct dopaminergic neurons and corresponding mushroom body output neurons. However, sleep and memory are coupled such that mushroom body neurons required for sleep-dependent memory also promote sleep. Flies lacking Neuropeptide F display sleep-dependent memory even when starved, suggesting that circuit selection is determined by hunger. This plasticity in memory circuits enables flies to retain essential information in changing environments.


Assuntos
Drosophila melanogaster/fisiologia , Comportamento Alimentar/fisiologia , Alimentos , Fome/fisiologia , Consolidação da Memória/fisiologia , Plasticidade Neuronal , Sono/fisiologia , Animais , Comportamento Apetitivo , Neurônios Dopaminérgicos/fisiologia , Drosophila melanogaster/citologia , Comportamento Alimentar/psicologia , Feminino , Masculino , Corpos Pedunculados/citologia , Corpos Pedunculados/fisiologia , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Inanição/fisiopatologia , Vigília/fisiologia
11.
J Neurosci ; 40(47): 9066-9077, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33106351

RESUMO

Dissociation between the output of the circadian clock and external environmental cues is a major cause of human cognitive dysfunction. While the effects of ablation of the molecular clock on memory have been studied in many systems, little has been done to test the role of specific clock circuit output signals. To address this gap, we examined the effects of mutations of Pigment-dispersing factor (Pdf) and its receptor, Pdfr, on associative memory in male and female Drosophila Loss of PDF signaling significantly decreases the ability to form associative memory. Appetitive short-term memory (STM), which in wild-type (WT) is time-of-day (TOD) independent, is decreased across the day by mutation of Pdf or Pdfr, but more substantially in the morning than in the evening. This defect is because of PDFR expression in adult neurons outside the core clock circuit and the mushroom body (MB) Kenyon cells (KCs). The acquisition of a TOD difference in mutants implies the existence of multiple oscillators that act to normalize memory formation across the day for appetitive processes. Interestingly, aversive STM requires PDF but not PDFR, suggesting that there are valence-specific pathways downstream of PDF that regulate memory formation. These data argue that the circadian clock uses circuit-specific and molecularly diverse output pathways to enhance the ability of animals to optimize responses to changing conditions.SIGNIFICANCE STATEMENT From humans to invertebrates, cognitive processes are influenced by organisms' internal circadian clocks, the pace of which is linked to the solar cycle. Disruption of this link is increasingly common (e.g., jetlag, social jetlag disorders) and causes cognitive impairments that are costly and long lasting. A detailed understanding of how the internal clock regulates cognition is critical for the development of therapeutic methods. Here, we show for the first time that olfactory associative memory in Drosophila requires signaling by Pigment-dispersing factor (PDF), a neuromodulatory signaling peptide produced only by circadian clock circuit neurons. We also find a novel role for the clock circuit in stabilizing appetitive sucrose/odor memory across the day.


Assuntos
Aprendizagem por Associação/fisiologia , Proteínas de Drosophila/fisiologia , Memória/fisiologia , Neuropeptídeos/fisiologia , Olfato/fisiologia , Animais , Apetite/fisiologia , Aprendizagem da Esquiva/fisiologia , Relógios Circadianos , Ritmo Circadiano , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiologia , Feminino , Masculino , Memória de Curto Prazo/fisiologia , Corpos Pedunculados/fisiologia , Mutação , Neurônios/fisiologia , Neuropeptídeos/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia
12.
Proc Natl Acad Sci U S A ; 117(18): 10024-10034, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32303656

RESUMO

Sleep pressure and sleep depth are key regulators of wake and sleep. Current methods of measuring these parameters in Drosophila melanogaster have low temporal resolution and/or require disrupting sleep. Here we report analysis tools for high-resolution, noninvasive measurement of sleep pressure and depth from movement data. Probability of initiating activity, P(Wake), measures sleep depth while probability of ceasing activity, P(Doze), measures sleep pressure. In vivo and computational analyses show that P(Wake) and P(Doze) are largely independent and control the amount of total sleep. We also develop a Hidden Markov Model that allows visualization of distinct sleep/wake substates. These hidden states have a predictable relationship with P(Doze) and P(Wake), suggesting that the methods capture the same behaviors. Importantly, we demonstrate that both the Doze/Wake probabilities and the sleep/wake substates are tied to specific biological processes. These metrics provide greater mechanistic insight into behavior than measuring the amount of sleep alone.


Assuntos
Ritmo Circadiano/fisiologia , Drosophila melanogaster/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Comportamento Animal/fisiologia , Humanos , Modelos Estatísticos , Movimento/fisiologia
13.
Nat Commun ; 11(1): 1092, 2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-32107390

RESUMO

Micro(mi)RNA-based post-transcriptional regulatory mechanisms have been broadly implicated in the assembly and modulation of synaptic connections required to shape neural circuits, however, relatively few specific miRNAs have been identified that control synapse formation. Using a conditional transgenic toolkit for competitive inhibition of miRNA function in Drosophila, we performed an unbiased screen for novel regulators of synapse morphogenesis at the larval neuromuscular junction (NMJ). From a set of ten new validated regulators of NMJ growth, we discovered that miR-34 mutants display synaptic phenotypes and cell type-specific functions suggesting distinct downstream mechanisms in the presynaptic and postsynaptic compartments. A search for conserved downstream targets for miR-34 identified the junctional receptor CNTNAP4/Neurexin-IV (Nrx-IV) and the membrane cytoskeletal effector Adducin/Hu-li tai shao (Hts) as proteins whose synaptic expression is restricted by miR-34. Manipulation of miR-34, Nrx-IV or Hts-M function in motor neurons or muscle supports a model where presynaptic miR-34 inhibits Nrx-IV to influence active zone formation, whereas, postsynaptic miR-34 inhibits Hts to regulate the initiation of bouton formation from presynaptic terminals.


Assuntos
Proteínas de Ligação a Calmodulina/genética , Moléculas de Adesão Celular Neuronais/genética , Proteínas de Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/metabolismo , Terminações Pré-Sinápticas/fisiologia , Animais , Animais Geneticamente Modificados , Proteínas de Ligação a Calmodulina/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Larva/crescimento & desenvolvimento , Morfogênese/genética , Mutação , Junção Neuromuscular/citologia , Junção Neuromuscular/crescimento & desenvolvimento
14.
G3 (Bethesda) ; 10(1): 43-55, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31694853

RESUMO

Locomotion is an ancient and fundamental output of the nervous system required for animals to perform many other complex behaviors. Although the formation of motor circuits is known to be under developmental control of transcriptional mechanisms that define the fates and connectivity of the many neurons, glia and muscle constituents of these circuits, relatively little is known about the role of post-transcriptional regulation of locomotor behavior. MicroRNAs have emerged as a potentially rich source of modulators for neural development and function. In order to define the microRNAs required for normal locomotion in Drosophila melanogaster, we utilized a set of transgenic Gal4-dependent competitive inhibitors (microRNA sponges, or miR-SPs) to functionally assess ca. 140 high-confidence Drosophila microRNAs using automated quantitative movement tracking systems followed by multiparametric analysis. Using ubiquitous expression of miR-SP constructs, we identified a large number of microRNAs that modulate aspects of normal baseline adult locomotion. Addition of temperature-dependent Gal80 to identify microRNAs that act during adulthood revealed that the majority of these microRNAs play developmental roles. Comparison of ubiquitous and neural-specific miR-SP expression suggests that most of these microRNAs function within the nervous system. Parallel analyses of spontaneous locomotion in adults and in larvae also reveal that very few of the microRNAs required in the adult overlap with those that control the behavior of larval motor circuits. These screens suggest that a rich regulatory landscape underlies the formation and function of motor circuits and that many of these mechanisms are stage and/or parameter-specific.


Assuntos
Locomoção/genética , MicroRNAs/genética , Animais , Drosophila melanogaster , Gânglios dos Invertebrados/metabolismo , MicroRNAs/metabolismo
15.
Curr Biol ; 29(21): 3635-3646.e5, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31668619

RESUMO

Both the structure and the amount of sleep are important for brain function. Entry into deep, restorative stages of sleep is time dependent; short sleep bouts selectively eliminate these states. Fragmentation-induced cognitive dysfunction is a feature of many common human sleep pathologies. Whether sleep structure is normally regulated independent of the amount of sleep is unknown. Here, we show that in Drosophila melanogaster, activation of a subset of serotonergic neurons fragments sleep without major changes in the total amount of sleep, dramatically reducing long episodes that may correspond to deep sleep states. Disruption of sleep structure results in learning deficits that can be rescued by pharmacologically or genetically consolidating sleep. We identify two reciprocally connected sets of ellipsoid body neurons that form the heart of a serotonin-modulated circuit that controls sleep architecture. Taken together, these findings define a circuit essential for controlling the structure of sleep independent of its amount.


Assuntos
Cognição , Drosophila melanogaster/fisiologia , Neurônios Serotoninérgicos/fisiologia , Serotonina/fisiologia , Sono/fisiologia , Animais , Feminino
16.
Cell Rep ; 23(13): 3776-3786, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29949763

RESUMO

To discover microRNAs that regulate sleep, we performed a genetic screen using a library of miRNA sponge-expressing flies. We identified 25 miRNAs that regulate baseline sleep; 17 were sleep-promoting and 8 promoted wake. We identified one miRNA that is required for recovery sleep after deprivation and 8 miRNAs that limit the extent of recovery sleep. 65% of the hits belong to human-conserved families. Interestingly, the majority (75%), but not all, of the baseline sleep-regulating miRNAs are required in neurons. Sponges that target miRNAs in the same family, including the miR-92a/92b/310 family and the miR-263a/263b family, have similar effects. Finally, mutation of one of the screen's strongest hits, let-7, using CRISPR/Cas-9, phenocopies sponge-mediated let-7 inhibition. Cell-type-specific and temporally restricted let-7 sponge expression experiments suggest that let-7 is required in the mushroom body both during development and in adulthood. This screen sets the stage for understanding the role of miRNAs in sleep.


Assuntos
Drosophila/metabolismo , MicroRNAs/metabolismo , Sono/genética , Animais , Ritmo Circadiano/genética , Drosophila/genética , MicroRNAs/antagonistas & inibidores , Neurônios/metabolismo
17.
J Neurophysiol ; 119(5): 1665-1680, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29364071

RESUMO

Drosophila ether-à-go-go ( eag) is the founding member of a large family of voltage-gated K+ channels, the KCNH family, which includes Kv10, 11, and 12. Concurrent binding of calcium/calmodulin (Ca2+/CaM) to NH2- and COOH-terminal sites inhibits mammalian EAG1 channels at submicromolar Ca2+ concentrations, likely by causing pore constriction. Although the Drosophila EAG channel was believed to be Ca2+-insensitive (Schönherr R, Löber K, Heinemann SH. EMBO J 19: 3263-3271, 2000.), both the NH2- and COOH-terminal sites are conserved. In this study we show that Drosophila EAG is inhibited by high Ca2+ concentrations that are only present at plasma membrane Ca2+ channel microdomains. To test the role of this regulation in vivo, we engineered mutations that block CaM-binding to the major COOH-terminal site of the endogenous eag locus, disrupting Ca2+-dependent inhibition. eag CaMBD mutants have reduced evoked release from larval motor neuron presynaptic terminals and show decreased Ca2+ influx in stimulated adult projection neuron presynaptic terminals, consistent with an increase in K+ conductance. These results are predicted by a conductance-based multicompartment model of the presynaptic terminal in which some fraction of EAG is localized to the Ca2+ channel microdomains that control neurotransmitter release. The reduction of release in the larval neuromuscular junction drives a compensatory increase in motor neuron somatic excitability. This misregulation of synaptic and somatic excitability has consequences for systems-level processes and leads to defects in associative memory formation in adults. NEW & NOTEWORTHY Regulation of excitability is critical to tuning the nervous system for complex behaviors. We demonstrate in this article that the EAG family of voltage-gated K+ channels exhibit conserved gating by Ca2+/CaM. Disruption of this inhibition in Drosophila results in decreased evoked neurotransmitter release due to truncated Ca2+ influx in presynaptic terminals. In adults, disrupted Ca2+ dynamics cripples memory formation. These data demonstrate that the biophysical details of channels have important implications for cell function and behavior.


Assuntos
Cálcio/metabolismo , Calmodulina/metabolismo , Proteínas de Drosophila/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Terminações Pré-Sinápticas/metabolismo , Animais , Drosophila , Feminino , Masculino
18.
J Neurosci ; 37(44): 10554-10566, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28954869

RESUMO

A null mutation of the Drosophila calcium/calmodulin-dependent protein kinase II gene (CaMKII) was generated using homologous recombination. Null animals survive to larval and pupal stages due to a large maternal contribution of CaMKII mRNA, which consists of a short 3'-untranslated region (UTR) form lacking regulatory elements that guide local translation. The selective loss of the long 3'UTR mRNA in CaMKII-null larvae allows us to test its role in plasticity. Development and evoked function of the larval neuromuscular junction are surprisingly normal, but the resting rate of miniature excitatory junctional potentials (mEJPs) is significantly lower in CaMKII mutants. Mutants also lack the ability to increase mEJP rate in response to spaced depolarization, a type of activity-dependent plasticity shown to require both transcription and translation. Consistent with this, overexpression of miR-289 in wild-type animals blocks plasticity of spontaneous release. In addition to the defects in regulation of mEJP rate, CaMKII protein is largely lost from synapses in the mutant. All phenotypes are non-sex-specific and rescued by a fosmid containing the entire wild-type CaMKII locus, but only viability and CaMKII localization are rescued by genomic fosmids lacking the long 3'UTR. This suggests that synaptic CaMKII accumulates by two distinct mechanisms: local synthesis requiring the long 3'UTR form of CaMKII mRNA and a process that requires zygotic transcription of CaMKII mRNA. The origin of synaptic CaMKII also dictates its functionality. Locally translated CaMKII has a privileged role in regulation of spontaneous release, which cannot be fulfilled by synaptic CaMKII from the other pool.SIGNIFICANCE STATEMENT As a regulator of synaptic development and plasticity, CaMKII has important roles in both normal and pathological function of the nervous system. CaMKII shows high conservation between Drosophila and humans, underscoring the usefulness of Drosophila in modeling its function. Drosophila CaMKII-null mutants remain viable throughout development, enabling morphological and electrophysiological characterization. Although the structure of the synapse is normal, maternally contributed CaMKII does not localize to synapses. Zygotic production of CaMKII mRNA with a long 3'-untranslated region is necessary for modulating spontaneous neurotransmission in an activity-dependent manner, but not for viability. These data argue that regulation of CaMKII localization and levels by local transcriptional processes is conserved. This is the first demonstration of distinct functions for Drosophila CaMKII mRNA variants.


Assuntos
Regiões 3' não Traduzidas/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Mutação/genética , Plasticidade Neuronal/fisiologia , Animais , Animais Geneticamente Modificados , Sequência de Bases , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/deficiência , Drosophila melanogaster , Feminino , Potenciais da Membrana/fisiologia , RNA Mensageiro/genética
20.
J Neurophysiol ; 117(6): 2125-2136, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28298298

RESUMO

Homeostatic control of intrinsic excitability is important for long-term regulation of neuronal activity. In conjunction with many other forms of plasticity, intrinsic homeostasis helps neurons maintain stable activity regimes in the face of external input variability and destabilizing genetic mutations. In this study, we report a mechanism by which Drosophila melanogaster larval motor neurons stabilize hyperactivity induced by the loss of the delayed rectifying K+ channel Shaker cognate B (Shab), by upregulating the Ca2+-dependent K+ channel encoded by the slowpoke (slo) gene. We also show that loss of SLO does not trigger a reciprocal compensatory upregulation of SHAB, implying that homeostatic signaling pathways utilize compensatory pathways unique to the channel that was mutated. SLO upregulation due to loss of SHAB involves nuclear Ca2+ signaling and dCREB, suggesting that the slo homeostatic response is transcriptionally mediated. Examination of the changes in gene expression induced by these mutations suggests that there is not a generic transcriptional response to increased excitability in motor neurons, but that homeostatic compensations are influenced by the identity of the lost conductance.NEW & NOTEWORTHY The idea that activity-dependent homeostatic plasticity is driven solely by firing has wide credence. In this report we show that homeostatic compensation after loss of an ion channel conductance is tailored to identity of the channel lost, not its properties.


Assuntos
Proteínas de Drosophila/metabolismo , Homeostase/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Neurônios Motores/fisiologia , Canais de Potássio Shab/metabolismo , Transmissão Sináptica/fisiologia , Animais , Animais Geneticamente Modificados , Cálcio/metabolismo , Cátions Bivalentes/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Técnicas de Silenciamento de Genes , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Larva , Plasticidade Neuronal/fisiologia , Técnicas de Patch-Clamp , Interferência de RNA , RNA Mensageiro/metabolismo , Canais de Potássio Shab/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...