Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(15): e2321338121, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38568969

RESUMO

To address the contribution of transcriptional regulation to Drosophila clock gene expression and to behavior, we generated a series of CRISPR-mediated deletions within two regions of the circadian gene timeless (tim), an intronic E-box region and an upstream E-box region that are both recognized by the key transcription factor Clock (Clk) and its heterodimeric partner Cycle. The upstream deletions but not an intronic deletion dramatically impact tim expression in fly heads; the biggest upstream deletion reduces peak RNA levels and tim RNA cycling amplitude to about 15% of normal, and there are similar effects on tim protein (TIM). The cycling amplitude of other clock genes is also strongly reduced, in these cases due to increases in trough levels. These data underscore the important contribution of the upstream E-box enhancer region to tim expression and of TIM to clock gene transcriptional repression in fly heads. Surprisingly, tim expression in clock neurons is only modestly affected by the biggest upstream deletion and is similarly affected by a deletion of the intronic E-box region. This distinction between clock neurons and glia is paralleled by a dramatically enhanced accessibility of the intronic enhancer region within clock neurons. This distinctive feature of tim chromatin was revealed by ATAC-seq (assay for transposase-accessible chromatin with sequencing) assays of purified neurons and glia as well as of fly heads. The enhanced cell type-specific accessibility of the intronic enhancer region explains the resilience of clock neuron tim expression and circadian behavior to deletion of the otherwise more prominent upstream tim E-box region.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Cromatina/metabolismo , Ritmo Circadiano/genética , Proteínas CLOCK/genética , DNA/metabolismo , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica , RNA/metabolismo
2.
Acta Neuropathol Commun ; 11(1): 164, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37845749

RESUMO

Identifying genetic modifiers of familial amyotrophic lateral sclerosis (ALS) may reveal targets for therapeutic modulation with potential application to sporadic ALS. GGGGCC (G4C2) repeat expansions in the C9orf72 gene underlie the most common form of familial ALS, and generate toxic arginine-containing dipeptide repeats (DPRs), which interfere with membraneless organelles, such as the nucleolus. Here we considered senataxin (SETX), the genetic cause of ALS4, as a modifier of C9orf72 ALS, because SETX is a nuclear helicase that may regulate RNA-protein interactions involved in ALS dysfunction. After documenting that decreased SETX expression enhances arginine-containing DPR toxicity and C9orf72 repeat expansion toxicity in HEK293 cells and primary neurons, we generated SETX fly lines and evaluated the effect of SETX in flies expressing either (G4C2)58 repeats or glycine-arginine-50 [GR(50)] DPRs. We observed dramatic suppression of disease phenotypes in (G4C2)58 and GR(50) Drosophila models, and detected a striking relocalization of GR(50) out of the nucleolus in flies co-expressing SETX. Next-generation GR(1000) fly models, that show age-related motor deficits in climbing and movement assays, were similarly rescued with SETX co-expression. We noted that the physical interaction between SETX and arginine-containing DPRs is partially RNA-dependent. Finally, we directly assessed the nucleolus in cells expressing GR-DPRs, confirmed reduced mobility of proteins trafficking to the nucleolus upon GR-DPR expression, and found that SETX dosage modulated nucleolus liquidity in GR-DPR-expressing cells and motor neurons. These findings reveal a hitherto unknown connection between SETX function and cellular processes contributing to neuron demise in the most common form of familial ALS.


Assuntos
Esclerose Amiotrófica Lateral , Demência Frontotemporal , Humanos , Animais , Esclerose Amiotrófica Lateral/metabolismo , Dipeptídeos/genética , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Arginina/genética , Arginina/metabolismo , Células HEK293 , Neurônios Motores/metabolismo , Drosophila/metabolismo , RNA/metabolismo , Demência Frontotemporal/genética , Expansão das Repetições de DNA/genética , DNA Helicases/genética , RNA Helicases/genética , Enzimas Multifuncionais/genética
3.
Curr Biol ; 33(17): 3660-3668.e4, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37552985

RESUMO

The needs fulfilled by sleep are unknown, though the effects of insufficient sleep are manifold. To better understand how the need to sleep is sensed and discharged, much effort has gone into identifying the neural circuits involved in regulating arousal, especially those that promote sleep. In prevailing models, the dorsal fan-shaped body (dFB) plays a central role in this process in the fly brain. In the present study we manipulated various properties of the dFB including its electrical activity, synaptic output, and endogenous gene expression. In each of these experimental contexts we were unable to identify any effect on sleep that could be unambiguously mapped to the dFB. Furthermore, we found evidence that sleep phenotypes previously attributed to the dFB were caused by genetic manipulations that inadvertently targeted the ventral nerve cord. We also examined expression of two genes whose purported effects have been attributed to functions within a specific subpopulation of dFB neurons. In both cases we found little to no expression in the expected cells. Collectively, our results cast doubt on the prevailing hypothesis that the dFB plays a central role in promoting sleep.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Sono/fisiologia , Privação do Sono
4.
J Neurosci ; 2022 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-35680412

RESUMO

The need to sleep is sensed and discharged in a poorly understood process that is homeostatically controlled over time. In flies, different contributions to this process have been attributed to peripheral ppk and central brain neurons, with the former serving as hypothetical inputs to the sleep homeostat and the latter reportedly serving as the homeostat itself. Here we re-evaluate these distinctions in light of new findings using female flies. First, activating neurons targeted by published ppk and brain drivers elicits similar phenotypes - namely sleep deprivation followed by rebound sleep. Second, inhibiting activity or synaptic output with one type of driver suppresses sleep homeostasis induced using the other type of driver. Third, drivers previously used to implicate central neurons in sleep homeostasis unexpectedly also label ppk neurons. Fourth, activating only this subset of co-labeled neurons is sufficient to elicit sleep homeostasis. Thus, many published contributions of central neurons to sleep homeostasis can be explained by previously unrecognized expression of brain drivers in peripheral ppk neurons, most likely those in the legs that promote walking. Lastly, we show that activation of certain non-ppk neurons can also induce sleep homeostasis. Notably, axons of these as well as ppk neurons terminate in the same ventral brain region, suggesting that a previously undefined neural circuit element of a sleep homeostat may lie nearby.SIGNIFICANCE STATEMENT:The biological need(s) that sleep fulfills are unknown, but they are reflected by an animal's ability to compensate for prior sleep loss in a process called sleep homeostasis. Researchers have searched for the neural circuitry that comprises the sleep homeostat so that the information it conveys can shed light on the nature of sleep need. Here we demonstrate that neurons originating outside of the brain are responsible for phenotypes previously attributed to the proposed central brain sleep homeostat in flies. Our results support a revised neural circuit model for sensing and discharging sleep need in which peripheral inputs connect to a sleep homeostat through previously unrecognized neural circuit elements in the ventral brain.

5.
J Neurosci ; 41(41): 8461-8474, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34446574

RESUMO

α7 nicotinic acetylcholine receptors (nAChRs) are widely expressed in the brain where they promote fast cholinergic synaptic transmission and serve important neuromodulatory functions. However, their high permeability to Ca2+ also predisposes them to contribute to disease states. Here, using transfected HEK-tsa cells and primary cultured hippocampal neurons from male and female rats, we demonstrate that two proteins called Ly6h and NACHO compete for access to α7 subunits, operating together but in opposition to maintain α7 assembly and activity within a narrow range that is optimal for neuronal function and viability. Using mixed gender human temporal cortex and cultured hippocampal neurons from rats we further show that this balance is perturbed during Alzheimer's disease (AD) because of amyloid ß (Aß)-driven reduction in Ly6h, with severe reduction leading to increased phosphorylated tau and α7-mediated neurotoxicity. Ly6h release into human CSF is also correlated with AD severity. Thus, Ly6h links cholinergic signaling, Aß and phosphorylated tau and may serve as a novel marker for AD progression.SIGNIFICANCE STATEMENT One of the earliest and most persistent hypotheses regarding Alzheimer's disease (AD) attributes cognitive impairment to loss of cholinergic signaling. More recently, interest has focused on crucial roles for amyloid ß (Aß) and phosphorylated tau in Alzheimer's pathogenesis. Here, we demonstrate that these elements are linked by Ly6h and its counterpart, NACHO, functioning in opposition to maintain assembly of nicotinic acetylcholine receptors (nAChRs) within the physiological range. Our data suggests that Aß shifts the balance away from Ly6h and toward NACHO, resulting in increased assembly of Ca2+-permeable nAChRs and thus a conversion of basal cholinergic to neurotoxic signaling.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular , Células Cultivadas , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Lobo Temporal/metabolismo , Lobo Temporal/patologia
6.
Curr Biol ; 29(22): R1181-R1184, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31743673

RESUMO

Sleep deprivation has long been known to impair cognition, but it has been difficult to distinguish whether loss or disruption of sleep is responsible. Now it appears that merely interrupting sleep is sufficient to interfere with memory formation.


Assuntos
Serotonina , Privação do Sono , Animais , Cognição , Drosophila , Memória
7.
Neuron ; 104(5): 947-959.e5, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31629603

RESUMO

Insect olfactory receptors operate as ligand-gated ion channels that directly transduce odor stimuli into electrical signals. However, in the absence of any known intermediate transduction steps, it remains unclear whether and how these ionotropic inputs are amplified in olfactory receptor neurons (ORNs). Here, we find that amplification occurs in the Drosophila courtship-promoting ORNs through Pickpocket 25 (PPK25), a member of the degenerin/epithelial sodium channel family (DEG/ENaC). Pharmacological and genetic manipulations indicate that, in Or47b and Ir84a ORNs, PPK25 mediates Ca2+-dependent signal amplification via an intracellular calmodulin-binding motif. Additionally, hormonal signaling upregulates PPK25 expression to determine the degree of amplification, with striking effects on male courtship. Together, these findings advance our understanding of sensory neurobiology by identifying an amplification mechanism compatible with ionotropic signaling. Moreover, this study offers new insights into DEG/ENaC activation by highlighting a novel means of regulation that is likely conserved across species.


Assuntos
Proteínas de Drosophila/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Comportamento Sexual Animal/fisiologia , Olfato/fisiologia , Canais de Sódio/metabolismo , Animais , Corte , Drosophila melanogaster , Masculino
8.
Physiology (Bethesda) ; 33(5): 317-327, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30109824

RESUMO

The functions of sleep remain a mystery. Yet they must be important since sleep is highly conserved, and its chronic disruption is associated with various metabolic, psychiatric, and neurodegenerative disorders. This review will cover our evolving understanding of the mechanisms by which sleep is controlled and the complex relationship between sleep and disease states.


Assuntos
Doenças Neurodegenerativas/fisiopatologia , Transtornos do Sono-Vigília/fisiopatologia , Sono/fisiologia , Animais , Humanos
9.
Curr Biol ; 26(20): R1073-R1087, 2016 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-27780049

RESUMO

Despite decades of intense study, the functions of sleep are still shrouded in mystery. The difficulty in understanding these functions can be at least partly attributed to the varied manifestations of sleep in different animals. Daily sleep duration can range from 4-20 hrs among mammals, and sleep can manifest throughout the brain, or it can alternate over time between cerebral hemispheres, depending on the species. Ecological factors are likely to have shaped these and other sleep behaviors during evolution by altering the properties of conserved arousal circuits in the brain. Nonetheless, core functions of sleep are likely to have arisen early and to have persisted to the present day in diverse organisms. This review will discuss the evolutionary forces that may be responsible for phylogenetic differences in sleep and the potential core functions that sleep fulfills.


Assuntos
Evolução Biológica , Invertebrados/fisiologia , Sono , Vertebrados/fisiologia , Animais
10.
Sci Rep ; 6: 25252, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27143646

RESUMO

Traumatic brain injury (TBI) is a major cause of morbidity and mortality worldwide. In addition, there has been a growing appreciation that even repetitive, milder forms of TBI (mTBI) can have long-term deleterious consequences to neural tissues. Hampering our understanding of genetic and environmental factors that influence the cellular and molecular responses to injury has been the limited availability of effective genetic model systems that could be used to identify the key genes and pathways that modulate both the acute and long-term responses to TBI. Here we report the development of a severe and mild-repetitive TBI model using Drosophila. Using this system, key features that are typically found in mammalian TBI models were also identified in flies, including the activation of inflammatory and autophagy responses, increased Tau phosphorylation and neuronal defects that impair sleep-related behaviors. This novel injury paradigm demonstrates the utility of Drosophila as an effective tool to validate genetic and environmental factors that influence the whole animal response to trauma and to identify prospective therapies needed for the treatment of TBI.


Assuntos
Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/fisiopatologia , Modelos Animais de Doenças , Drosophila , Animais
11.
Neuropharmacology ; 107: 262-270, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27033596

RESUMO

Endogenous circadian (∼24 h) clocks regulate key physiological and cognitive processes via rhythmic expression of clock genes. The main circadian pacemaker is the hypothalamic suprachiasmatic nucleus (SCN). Mood disorders, including bipolar disorder (BD), are commonly associated with disturbed circadian rhythms. Dopamine (DA) contributes to mania in BD and has direct impact on clock gene expression. Therefore, we hypothesized that high levels of DA during episodes of mania contribute to disturbed circadian rhythms in BD. The mood stabilizer valproic acid (VPA) also affects circadian rhythms. Thus, we further hypothesized that VPA normalizes circadian disturbances caused by elevated levels of DA. To test these hypotheses, we examined locomotor rhythms and circadian gene cycling in mice with reduced expression of the dopamine transporter (DAT-KD mice), which results in elevated DA levels and mania-like behavior. We found that elevated DA signaling lengthened the circadian period of behavioral rhythms in DAT-KD mice and clock gene expression rhythms in SCN explants. In contrast, we found that VPA shortened circadian period of behavioral rhythms in DAT-KD mice and clock gene expression rhythms in SCN explants, hippocampal cell lines, and human fibroblasts from BD patients. Thus, DA and VPA have opposing effects on circadian period. To test whether the impact of VPA on circadian rhythms contributes to its behavioral effects, we fed VPA to DAT-deficient Drosophila with and without functioning circadian clocks. Consistent with our hypothesis, we found that VPA had potent activity-suppressing effects in hyperactive DAT-deficient flies with intact circadian clocks. However, these effects were attenuated in DAT-deficient flies in which circadian clocks were disrupted, suggesting that VPA functions partly through the circadian clock to suppress activity. Here, we provide in vivo and in vitro evidence across species that elevated DA signaling lengthens the circadian period, an effect remediated by VPA treatment. Hence, VPA may exert beneficial effects on mood by normalizing lengthened circadian rhythm period in subjects with elevated DA resulting from reduced DAT.


Assuntos
Antimaníacos/farmacologia , Ritmo Circadiano/efeitos dos fármacos , Dopamina/metabolismo , Locomoção/efeitos dos fármacos , Ácido Valproico/farmacologia , Animais , Antimaníacos/uso terapêutico , Células Cultivadas , Ritmo Circadiano/fisiologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/deficiência , Drosophila , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Transtornos do Humor/tratamento farmacológico , Transtornos do Humor/metabolismo , Ácido Valproico/uso terapêutico
12.
PLoS One ; 11(2): e0148215, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26828958

RESUMO

Ly6 proteins are endogenous prototoxins found in most animals. They show striking structural and functional parallels to snake α-neurotoxins, including regulation of ion channels and cholinergic signaling. However, the structural contributions of Ly6 proteins to regulation of effector molecules is poorly understood. This question is particularly relevant to the Ly6 protein QUIVER/SLEEPLESS (QVR/SSS), which has previously been shown to suppress excitability and synaptic transmission by upregulating potassium (K) channels and downregulating nicotinic acetylcholine receptors (nAChRs) in wake-promoting neurons to facilitate sleep in Drosophila. Using deletion mutagenesis, co-immunoprecipitations, ion flux assays, surface labeling and confocal microscopy, we demonstrate that only loop 2 is required for many of the previously described properties of SSS in transfected cells, including interactions with K channels and nAChRs. Collectively our data suggest that QVR/SSS, and by extension perhaps other Ly6 proteins, target effector molecules using limited protein motifs. Mapping these motifs may be useful in rational design of drugs that mimic or suppress Ly6-effector interactions to modulate nervous system function.


Assuntos
Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Mutagênese/genética , Canais de Potássio/química , Canais de Potássio/genética , Receptores Nicotínicos/metabolismo , Deleção de Sequência/genética , Superfamília Shaker de Canais de Potássio/metabolismo , Animais , Membrana Celular/metabolismo , Proteínas de Membrana , Modelos Moleculares , Ligação Proteica , Transporte Proteico , Relação Estrutura-Atividade
13.
Curr Biol ; 25(22): 2928-38, 2015 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-26526372

RESUMO

Sleep is thought to be controlled by two main processes: a circadian clock that primarily regulates sleep timing and a homeostatic mechanism that detects and responds to sleep need. Whereas abundant experimental evidence suggests that sleep need increases with time spent awake, the contributions of different brain arousal systems have not been assessed independently of each other to determine whether certain neural circuits, rather than waking per se, selectively contribute to sleep homeostasis. Using the fruit fly, Drosophila melanogaster, we found that sustained thermogenetic activation of three independent neurotransmitter systems promoted nighttime wakefulness. However, only sleep deprivation resulting from activation of cholinergic neurons was sufficient to elicit subsequent homeostatic recovery sleep, as assessed by multiple behavioral criteria. In contrast, sleep deprivation resulting from activation of octopaminergic neurons suppressed homeostatic recovery sleep, indicating that wakefulness can be dissociated from accrual of sleep need. Neurons that promote sleep homeostasis were found to innervate the central brain and motor control regions of the thoracic ganglion. Blocking activity of these neurons suppressed recovery sleep but did not alter baseline sleep, further differentiating between neural control of sleep homeostasis and daily fluctuations in the sleep/wake cycle. Importantly, selective activation of wake-promoting neurons without engaging the sleep homeostat impaired subsequent short-term memory, thus providing evidence that neural circuits that regulate sleep homeostasis are important for behavioral plasticity. Together, our data suggest a neural circuit model involving distinct populations of wake-promoting neurons, some of which are involved in homeostatic control of sleep and cognition.


Assuntos
Drosophila melanogaster/fisiologia , Sono/fisiologia , Animais , Nível de Alerta/fisiologia , Encéfalo/fisiologia , Ritmo Circadiano , Homeostase/fisiologia , Memória de Curto Prazo/fisiologia , Modelos Animais , Neurônios/fisiologia , Receptores de Amina Biogênica/fisiologia , Vigília/fisiologia
14.
J Biol Chem ; 290(40): 24509-18, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26276394

RESUMO

α4ß2 nicotinic acetylcholine receptors (nAChRs) are abundantly expressed throughout the central nervous system and are thought to be the primary target of nicotine, the main addictive substance in cigarette smoking. Understanding the mechanisms by which these receptors are regulated may assist in developing compounds to selectively interfere with nicotine addiction. Here we report previously unrecognized modulatory properties of members of the Ly6 protein family on α4ß2 nAChRs. Using a FRET-based Ca(2+) flux assay, we found that the maximum response of α4ß2 receptors to agonist was strongly inhibited by Ly6h and Lynx2 but potentiated by Ly6g6e. The mechanisms underlying these opposing effects appear to be fundamentally distinct. Receptor inhibition by Lynx2 was accompanied by suppression of α4ß2 expression at the cell surface, even when assays were preceded by chronic exposure of cells to an established chaperone, nicotine. Receptor inhibition by Lynx2 also was resistant to pretreatment with extracellular phospholipase C, which cleaves lipid moieties like those that attach Ly6 proteins to the plasma membrane. In contrast, potentiation of α4ß2 activity by Ly6g6e was readily reversible by pretreatment with phospholipase C. Potentiation was also accompanied by slowing of receptor desensitization and an increase in peak currents. Collectively our data support roles for Lynx2 and Ly6g6e in intracellular trafficking and allosteric potentiation of α4ß2 nAChRs, respectively.


Assuntos
Antígenos Ly/metabolismo , Glicoproteínas de Membrana/metabolismo , Neuropeptídeos/metabolismo , Receptores Nicotínicos/metabolismo , Sítio Alostérico , Animais , Biotinilação , Cálcio/química , Membrana Celular/metabolismo , Transferência Ressonante de Energia de Fluorescência , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Lipídeos/química , Camundongos , Nicotina/química , Agonistas Nicotínicos/farmacologia , Técnicas de Patch-Clamp , Ligação Proteica , Transporte Proteico , Fumar/efeitos adversos , Fosfolipases Tipo C/metabolismo
15.
PLoS One ; 10(7): e0132768, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26182057

RESUMO

Multiple neurological disorders are characterized by the abnormal accumulation of protein aggregates and the progressive impairment of complex behaviors. Our Drosophila studies demonstrate that middle-aged wild-type flies (WT, ~4-weeks) exhibit a marked accumulation of neural aggregates that is commensurate with the decline of the autophagy pathway. However, enhancing autophagy via neuronal over-expression of Atg8a (Atg8a-OE) reduces the age-dependent accumulation of aggregates. Here we assess basal locomotor activity profiles for single- and group-housed male and female WT flies and observed that only modest behavioral changes occurred by 4-weeks of age, with the noted exception of group-housed male flies. Male flies in same-sex social groups exhibit a progressive increase in nighttime activity. Infrared videos show aged group-housed males (4-weeks) are engaged in extensive bouts of courtship during periods of darkness, which is partly repressed during lighted conditions. Together, these nighttime courtship behaviors were nearly absent in young WT flies and aged Atg8a-OE flies. Previous studies have indicated a regulatory role for olfaction in male courtship partner choice. Coincidently, the mRNA expression profiles of several olfactory genes decline with age in WT flies; however, they are maintained in age-matched Atg8a-OE flies. Together, these results suggest that middle-aged male flies develop impairments in olfaction, which could contribute to the dysregulation of courtship behaviors during dark time periods. Combined, our results demonstrate that as Drosophila age, they develop early behavior defects that are coordinate with protein aggregate accumulation in the nervous system. In addition, the nighttime activity behavior is preserved when neuronal autophagy is maintained (Atg8a-OE flies). Thus, environmental or genetic factors that modify autophagic capacity could have a positive impact on neuronal aging and complex behaviors.


Assuntos
Envelhecimento/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas Heterotriméricas de Ligação ao GTP/genética , Olfato/genética , Envelhecimento/metabolismo , Animais , Autofagia/genética , Ritmo Circadiano/genética , Corte , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Feminino , Regulação da Expressão Gênica , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Masculino , Atividade Motora , Neurônios/metabolismo , Neurônios/patologia , Agregados Proteicos , Fatores Sexuais
16.
PLoS Genet ; 11(7): e1005344, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26131556

RESUMO

Many protein-coding genes identified by genome sequencing remain without functional annotation or biological context. Here we define a novel protein-coding gene, Nmf9, based on a forward genetic screen for neurological function. ENU-induced and genome-edited null mutations in mice produce deficits in vestibular function, fear learning and circadian behavior, which correlated with Nmf9 expression in inner ear, amygdala, and suprachiasmatic nuclei. Homologous genes from unicellular organisms and invertebrate animals predict interactions with small GTPases, but the corresponding domains are absent in mammalian Nmf9. Intriguingly, homozygotes for null mutations in the Drosophila homolog, CG45058, show profound locomotor defects and premature death, while heterozygotes show striking effects on sleep and activity phenotypes. These results link a novel gene orthology group to discrete neurological functions, and show conserved requirement across wide phylogenetic distance and domain level structural changes.


Assuntos
Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Proteínas de Drosophila/genética , Medo/fisiologia , Proteínas do Tecido Nervoso/genética , Vestíbulo do Labirinto/patologia , Tonsila do Cerebelo/metabolismo , Animais , Sequência de Bases , Comportamento Animal/fisiologia , Drosophila melanogaster/genética , Feminino , Deleção de Genes , Locomoção/genética , Masculino , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Análise de Sequência de DNA , Fatores Sexuais , Sono/genética , Sono/fisiologia , Núcleo Supraquiasmático/metabolismo , Testes de Função Vestibular , Vestíbulo do Labirinto/fisiologia
17.
Curr Biol ; 25(6): R234-R236, 2015 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-25784043

RESUMO

A new study in fruit flies suggests modulation of neural activity links sleep and Alzheimer's disease. Both sleep loss and amyloid beta increase neural excitability, which reinforces the accumulation of amyloid beta and shortens lifespan.


Assuntos
Degeneração Neural/fisiopatologia , Sono/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/fisiologia , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Humanos , Degeneração Neural/patologia , Privação do Sono/patologia , Privação do Sono/fisiopatologia
18.
J Neurosci ; 35(8): 3420-30, 2015 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25716842

RESUMO

α7 nAChRs are expressed widely throughout the brain, where they are important for synaptic signaling, gene transcription, and plastic changes that regulate sensory processing, cognition, and neural responses to chronic nicotine exposure. However, the mechanisms by which α7 nAChRs are regulated are poorly understood. Here we show that trafficking of α7-subunits is controlled by endogenous membrane-associated prototoxins in the Ly6 family. In particular, we find that Ly6h reduces cell-surface expression and calcium signaling by α7 nAChRs. We detect Ly6h in several rat brain regions, including the hippocampus, where we find it is both necessary and sufficient to limit the magnitude of α7-mediated currents. Consistent with such a regulatory function, knockdown of Ly6h in rat hippocampal pyramidal neurons enhances nicotine-induced potentiation of glutamatergic mEPSC amplitude, which is known to be mediated by α7 signaling. Collectively our data suggest a novel cellular role for Ly6 proteins in regulating nAChRs, which may be relevant to plastic changes in the nervous system including rewiring of glutamatergic circuitry during nicotine addiction.


Assuntos
Potenciais Pós-Sinápticos Excitadores , Potenciação de Longa Duração , Glicoproteínas de Membrana/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Sequência de Aminoácidos , Animais , Sinalização do Cálcio , Células Cultivadas , Ácido Glutâmico/farmacologia , Células HEK293 , Hipocampo/citologia , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Camundongos , Potenciais Pós-Sinápticos em Miniatura , Dados de Sequência Molecular , Agonistas Nicotínicos/farmacologia , Transporte Proteico , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Células Piramidais/fisiologia
19.
Curr Biol ; 24(6): 621-9, 2014 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-24613312

RESUMO

BACKGROUND: Although sleep is conserved throughout evolution, the molecular basis of its control is still largely a mystery. We previously showed that the quiver/sleepless (qvr/sss) gene encodes a membrane-tethered protein that is required for normal sleep in Drosophila. SLEEPLESS (SSS) protein functions, at least in part, by upregulating the levels and open probability of Shaker (Sh) potassium channels to suppress neuronal excitability and enable sleep. Consistent with this proposed mechanism, loss-of-function mutations in Sh phenocopy qvr/sss-null mutants. However, sleep is more genetically modifiable in Sh than in qvr/sss mutants, suggesting that SSS may regulate additional molecules to influence sleep. RESULTS: Here we show that SSS also antagonizes nicotinic acetylcholine receptors (nAChRs) to reduce synaptic transmission and promote sleep. Mimicking this antagonism with the nAChR inhibitor mecamylamine or by RNAi knockdown of specific nAChR subunits is sufficient to restore sleep to qvr/sss mutants. Regulation of nAChR activity by SSS occurs posttranscriptionally, since the levels of nAChR mRNAs are unchanged in qvr/sss mutants. Regulation of nAChR activity by SSS may in fact be direct, since SSS forms a stable complex with and antagonizes nAChR function in transfected cells. Intriguingly, lynx1, a mammalian homolog of SSS, can partially restore normal sleep to qvr/sss mutants, and lynx1 can form stable complexes with Shaker-type channels and nAChRs. CONCLUSIONS: Together, our data point to an evolutionarily conserved, bifunctional role for SSS and its homologs in controlling excitability and synaptic transmission in fundamental processes of the nervous system such as sleep.


Assuntos
Neurônios Colinérgicos/fisiologia , Proteínas de Drosophila/fisiologia , Proteínas de Membrana/fisiologia , Sono/fisiologia , Transmissão Sináptica/fisiologia , Animais , Drosophila melanogaster , Técnicas de Silenciamento de Genes , Corpos Pedunculados/fisiologia , Receptores Nicotínicos/fisiologia , Superfamília Shaker de Canais de Potássio/fisiologia , Vigília/fisiologia
20.
Mol Metab ; 3(1): 42-54, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24567903

RESUMO

Obesity has dramatically increased in prevalence, making it essential to understand its accompanying metabolic changes. Modeling diet-induced obesity in Drosophila melanogaster (fruit flies), we elucidated transcriptional and metabolic changes in w (1118) flies on a high-fat diet (HFD). Mass spectrometry-based metabolomics revealed altered fatty acid, amino acid, and carbohydrate metabolism with HFD. Microarray analysis uncovered transcriptional changes in nitrogen metabolism, including CG9510, homolog of human argininosuccinate lyase (ASL). CG9510 knockdown in flies phenocopied traits observed with HFD, namely increased triglyceride levels and decreased cold tolerance. Restoration of CG9510 expression ameliorated observed negative consequences of HFD. Metabolomic analysis of CG9510 knockdown flies confirmed functional similarity to ASL, regulating the balance of carbon and nitrogen metabolism. In summary, we found that HFD suppresses CG9510 expression, a gene required for proper triglyceride storage and stress tolerance. These results draw an important link between regulation of amino acid metabolism and the response to diet-induced obesity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...