Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Addict Biol ; 28(8): e13304, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37500483

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ritmo Circadiano , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Sueño , Neuronas/metabolismo
2.
bioRxiv ; 2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36778487

RESUMEN

Alcohol tolerance is a simple form of behavioral 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 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 comprised 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.

3.
J Neurosci ; 43(12): 2210-2220, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36750369

RESUMEN

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.


Asunto(s)
Alcoholismo , Proteínas de Drosophila , Animales , Masculino , Drosophila/metabolismo , Sirtuina 1/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Etanol/farmacología , Alcoholismo/metabolismo , Cuerpos Pedunculados/metabolismo , Drosophila melanogaster/genética , Receptores Citoplasmáticos y Nucleares/metabolismo
4.
Elife ; 102021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-34018925

RESUMEN

Thirst is a motivational state that drives behaviors to obtain water for fluid homeostasis. We identified two types of central brain interneurons that regulate thirsty water seeking in Drosophila, that we term the Janu neurons. Janu-GABA, a local interneuron in the subesophageal zone, is activated by water deprivation and is specific to thirsty seeking. Janu-AstA projects from the subesophageal zone to the superior medial protocerebrum, a higher order processing area. Janu-AstA signals with the neuropeptide Allatostatin A to promote water seeking and to inhibit feeding behavior. NPF (Drosophila NPY) neurons are postsynaptic to Janu-AstA for water seeking and feeding through the AstA-R2 galanin-like receptor. NPF neurons use NPF to regulate thirst and hunger behaviors. Flies choose Janu neuron activation, suggesting that thirsty seeking up a humidity gradient is rewarding. These findings identify novel central brain circuit elements that coordinate internal state drives to selectively control motivated seeking behavior.


Asunto(s)
Encéfalo/fisiología , Ingestión de Líquidos , Drosophila melanogaster/fisiología , Conducta Alimentaria , Neuronas GABAérgicas/fisiología , Hambre , Interneuronas/fisiología , Sed , Animales , Animales Modificados Genéticamente , Encéfalo/citología , Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Neuronas GABAérgicas/metabolismo , Interneuronas/metabolismo , Inhibición Neural , Neuropéptido Y/metabolismo , Oligopéptidos/metabolismo , Receptores de Neuropéptido/metabolismo
5.
Genetics ; 213(4): 1465-1478, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31619445

RESUMEN

Caenorhabditis elegans larval development requires the function of the two Canal-Associated Neurons (CANs): killing the CANs by laser microsurgery or disrupting their development by mutating the gene ceh-10 results in early larval arrest. How these cells promote larval development, however, remains a mystery. In screens for mutations that bypass CAN function, we identified the gene kin-29, which encodes a member of the Salt-Inducible Kinase (SIK) family and a component of a conserved pathway that regulates various C. elegans phenotypes. Like kin-29 loss, gain-of-function mutations in genes that may act upstream of kin-29 or growth in cyclic-AMP analogs bypassed ceh-10 larval arrest, suggesting that a conserved adenylyl cyclase/PKA pathway inhibits KIN-29 to promote larval development, and that loss of CAN function results in dysregulation of KIN-29 and larval arrest. The adenylyl cyclase ACY-2 mediates CAN-dependent larval development: acy-2 mutant larvae arrested development with a similar phenotype to ceh-10 mutants, and the arrest phenotype was suppressed by mutations in kin-29 ACY-2 is expressed predominantly in the CANs, and we provide evidence that the acy-2 functions in the CANs to promote larval development. By contrast, cell-specific expression experiments suggest that kin-29 acts in both the hypodermis and neurons, but not in the CANs. Based on our findings, we propose two models for how ACY-2 activity in the CANs regulates KIN-29 in target cells.


Asunto(s)
Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , AMP Cíclico/metabolismo , Neuronas/metabolismo , Transducción de Señal , Adenilil Ciclasas/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Larva/crecimiento & desarrollo , Modelos Biológicos , Mutación/genética , Fenotipo , Dominios Proteicos , Tejido Subcutáneo , Regulación hacia Arriba
6.
Sci Rep ; 8(1): 5777, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636522

RESUMEN

Hunger evokes stereotypic behaviors that favor the discovery of nutrients. The neural pathways that coordinate internal and external cues to motivate foraging behaviors are only partly known. Drosophila that are food deprived increase locomotor activity, are more efficient in locating a discrete source of nutrition, and are willing to overcome adversity to obtain food. We developed a simple open field assay that allows flies to freely perform multiple steps of the foraging sequence, and we show that two distinct dopaminergic neural circuits regulate measures of foraging behaviors. One group, the PAM neurons, functions in food deprived flies while the other functions in well fed flies, and both promote foraging. These satiation state-dependent circuits converge on dopamine D1 receptor-expressing Kenyon cells of the mushroom body, where neural activity promotes foraging independent of satiation state. These findings provide evidence for active foraging in well-fed flies that is separable from hunger-driven foraging.


Asunto(s)
Drosophila/fisiología , Privación de Alimentos , Locomoción , Cuerpos Pedunculados/metabolismo , Neuronas/metabolismo , Saciedad , Animales , Dopamina/metabolismo , Drosophila/metabolismo , Conducta Alimentaria , Masculino , Vías Nerviosas
7.
Cell Rep ; 22(7): 1647-1656, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29444420

RESUMEN

Ethanol is the most common drug of abuse. It exerts its behavioral effects by acting on widespread neural circuits; however, its impact on glial cells is less understood. We show that Drosophila perineurial glia are critical for ethanol tolerance, a simple form of behavioral plasticity. The perineurial glia form the continuous outer cellular layer of the blood-brain barrier and are the interface between the brain and the circulation. Ethanol tolerance development requires the A kinase anchoring protein Akap200 specifically in perineurial glia. Akap200 tightly coordinates protein kinase A, actin, and calcium signaling at the membrane to control tolerance. Furthermore, ethanol causes a structural remodeling of the actin cytoskeleton and perineurial membrane topology in an Akap200-dependent manner, without disrupting classical barrier functions. Our findings reveal an active molecular signaling process in the cells at the blood-brain interface that permits a form of behavioral plasticity induced by ethanol.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Conducta Animal/efectos de los fármacos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Etanol/toxicidad , Proteínas de la Membrana/metabolismo , Neuroglía/metabolismo , Nervios Periféricos/patología , Actinas/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Drosophila melanogaster/efectos de los fármacos , Mutación/genética , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo
8.
Neuroscience ; 348: 191-200, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28215745

RESUMEN

Abnormal buildup of the microtubule associated protein tau is a major pathological hallmark of Alzheimer's disease (AD) and various tauopathies. The mechanisms by which pathological tau accumulates and spreads throughout the brain remain largely unknown. Previously, we demonstrated that a restoration of the major astrocytic glutamate transporter, GLT1, ameliorated a buildup of tau pathology and rescued cognition in a mouse model of AD. We hypothesized that aberrant extracellular glutamate and abnormal neuronal excitatory activities promoted tau pathology. In the present study, we investigated genetic interactions between tau and the GLT1 homolog dEaat1 in Drosophila melanogaster. Neuronal-specific overexpression of human wildtype tau markedly shortened lifespan and impaired motor behavior. RNAi depletion of dEaat1 in astrocytes worsened these phenotypes, whereas overexpression of dEaat1 improved them. However, the synaptic neuropil appeared unaffected, and we failed to detect any major neuronal loss with tau overexpression in combination with dEaat1 depletion. To mimic glutamate-induced aberrant excitatory input in neurons, repeated depolarization of neurons via transgenic TrpA1 was applied to the adult Drosophila optic nerves, and we examined the change of tau deposits. Repeated depolarization significantly increased the accumulation of tau in these neurons. We propose that increased neuronal excitatory activity exacerbates tau-mediated neuronal toxicity and behavioral deficits.


Asunto(s)
Astrocitos/metabolismo , Proteínas de Drosophila/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Proteínas tau/metabolismo , Animales , Transporte Biológico , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Drosophila melanogaster
9.
G3 (Bethesda) ; 6(12): 4217-4226, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27760793

RESUMEN

Sleep is an essential behavioral state of rest that is regulated by homeostatic drives to ensure a balance of sleep and activity, as well as independent arousal mechanisms in the central brain. Dopamine has been identified as a critical regulator of both sleep behavior and arousal. Here, we present results of a genetic screen that selectively restored the Dopamine Receptor (DopR/DopR1/dumb) to specific neuroanatomical regions of the adult Drosophila brain to assess requirements for DopR in sleep behavior. We have identified subsets of the mushroom body that utilizes DopR in daytime sleep regulation. These data are supported by multiple examples of spatially restricted genetic rescue data in discrete circuits of the mushroom body, as well as immunohistochemistry that corroborates the localization of DopR protein within mushroom body circuits. Independent loss of function data using an inducible RNAi construct in the same specific circuits also supports a requirement for DopR in daytime sleep. Additional circuit activation of discrete DopR+ mushroom body neurons also suggests roles for these subpopulations in sleep behavior. These conclusions support a new separable function for DopR in daytime sleep regulation within the mushroom body. This daytime regulation is independent of the known role of DopR in nighttime sleep, which is regulated within the Fan-Shaped Body (FSB). This study provides new neuroanatomical loci for exploration of dopaminergic sleep functions in Drosophila, and expands our understanding of sleep regulation during the day vs. night.


Asunto(s)
Drosophila/fisiología , Receptores Dopaminérgicos/genética , Sueño/genética , Animales , Animales Modificados Genéticamente , Conducta Animal , Encéfalo/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Técnicas de Inactivación de Genes , Pruebas Genéticas , Genotipo , Masculino , Cuerpos Pedunculados/metabolismo , Mutación
10.
J Neurosci ; 36(19): 5241-51, 2016 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-27170122

RESUMEN

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.


Asunto(s)
Intoxicación Alcohólica/metabolismo , Alcoholismo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Histona Desacetilasas/metabolismo , Terminales Presinápticos/metabolismo , Recompensa , Sirtuinas/metabolismo , Intoxicación Alcohólica/genética , Alcoholismo/genética , Animales , Drosophila/genética , Drosophila/fisiología , Proteínas de Drosophila/genética , Histona Desacetilasas/genética , Histonas/metabolismo , Cuerpos Pedunculados/metabolismo , Terminales Presinápticos/fisiología , Sirtuinas/genética , Sinapsinas/genética , Sinapsinas/metabolismo , Transcriptoma
11.
Biomed J ; 38(6): 496-509, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27013449

RESUMEN

The neural circuitry and molecules that control the rewarding properties of food and drugs of abuse appear to partially overlap in the mammalian brain. This has raised questions about the extent of the overlap and the precise role of specific circuit elements in reward and in other behaviors associated with feeding regulation and drug responses. The much simpler brain of invertebrates including the fruit fly Drosophila, offers an opportunity to make high-resolution maps of the circuits and molecules that govern behavior. Recent progress in Drosophila has revealed not only some common substrates for the actions of drugs of abuse and for the regulation of feeding, but also a remarkable level of conservation with vertebrates for key neuromodulatory transmitters. We speculate that Drosophila may serve as a model for distinguishing the neural mechanisms underlying normal and pathological motivational states that will be applicable to mammals.


Asunto(s)
Conducta Alimentaria/efectos de los fármacos , Drogas Ilícitas/farmacología , Animales , Dopamina/fisiología , Drosophila , Modelos Animales , Octopamina/fisiología , Tiramina/fisiología
12.
PLoS One ; 7(12): e50594, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23227189

RESUMEN

Neuronal signal transduction by the JNK MAP kinase pathway is altered by a broad array of stimuli including exposure to the widely abused drug ethanol, but the behavioral relevance and the regulation of JNK signaling is unclear. Here we demonstrate that JNK signaling functions downstream of the Sterile20 kinase family gene tao/Taok3 to regulate the behavioral effects of acute ethanol exposure in both the fruit fly Drosophila and mice. In flies tao is required in neurons to promote sensitivity to the locomotor stimulant effects of acute ethanol exposure and to establish specific brain structures. Reduced expression of key JNK pathway genes substantially rescued the structural and behavioral phenotypes of tao mutants. Decreasing and increasing JNK pathway activity resulted in increased and decreased sensitivity to the locomotor stimulant properties of acute ethanol exposure, respectively. Further, JNK expression in a limited pattern of neurons that included brain regions implicated in ethanol responses was sufficient to restore normal behavior. Mice heterozygous for a disrupted allele of the homologous Taok3 gene (Taok3Gt) were resistant to the acute sedative effects of ethanol. JNK activity was constitutively increased in brains of Taok3Gt/+ mice, and acute induction of phospho-JNK in brain tissue by ethanol was occluded in Taok3Gt/+ mice. Finally, acute administration of a JNK inhibitor conferred resistance to the sedative effects of ethanol in wild-type but not Taok3Gt/+ mice. Taken together, these data support a role of a TAO/TAOK3-JNK neuronal signaling pathway in regulating sensitivity to acute ethanol exposure in flies and in mice.


Asunto(s)
Proteínas de Drosophila/fisiología , Etanol/farmacología , MAP Quinasa Quinasa 4/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Secuencia de Bases , Conducta Animal , Cartilla de ADN , Drosophila , Inmunohistoquímica , MAP Quinasa Quinasa 4/genética , Ratones , Ratones Endogámicos C57BL , Mutación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Fly (Austin) ; 5(3): 191-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21750412

RESUMEN

The relationship between alcohol consumption, sensitivity, and tolerance is an important question that has been addressed in humans and rodent models. Studies have shown that alcohol consumption and risk of abuse may correlate with (1) increased sensitivity to the stimulant effects of alcohol, (2) decreased sensitivity to the depressant effects of alcohol, and (3) increased alcohol tolerance. However, many conflicting results have been observed. To complement these studies, we utilized a different organism and approach to analyze the relationship between ethanol consumption and other ethanol responses. Using a set of 20 Drosophila melanogaster mutants that were isolated for altered ethanol sensitivity, we measured ethanol-induced hyperactivity, ethanol sedation, sedation tolerance, and ethanol consumption preference. Ethanol preference showed a strong positive correlation with ethanol tolerance, consistent with some rodent and human studies, but not with ethanol hyperactivity or sedation. No pairwise correlations were observed between ethanol hyperactivity, sedation, and tolerance. The evolutionary conservation of the relationship between tolerance and ethanol consumption in flies, rodents, and humans indicates that there are fundamental biological mechanisms linking specific ethanol responses.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Intoxicación Alcohólica/genética , Depresores del Sistema Nervioso Central/farmacología , Drosophila melanogaster/genética , Etanol/farmacología , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/metabolismo , Preferencias Alimentarias , Hipercinesia/inducido químicamente , Masculino , Factores de Transcripción/metabolismo
14.
G3 (Bethesda) ; 1(7): 627-35, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22384374

RESUMEN

Alcohol use disorders are influenced by many interacting genetic and environmental factors. Highlighting this complexity is the observation that large genome-wide association experiments have implicated many genes with weak statistical support. Experimental model systems, cell culture and animal, have identified many genes and pathways involved in ethanol response, but their applicability to the development of alcohol use disorders in humans is undetermined. To overcome the limitations of any single experimental system, the analytical strategy used here was to identify genes that exert common phenotypic effects across multiple experimental systems. Specifically, we (1) performed a mouse linkage analysis to identify quantitative trait loci that influence ethanol-induced ataxia; (2) performed a human genetic association analysis of the mouse-identified loci against ethanol-induced body sway, a phenotype that is not only comparable to the mouse ethanol-ataxia phenotype but is also a genetically influenced endophenotype of alcohol use disorders; (3) performed behavioral genetic experiments in Drosophila showing that fly homologs of GPC5, the member of the glypican gene family implicated by both the human and mouse genetic analyses, influence the fly's response to ethanol; and (4) discovered data from the literature demonstrating that the genetically implicated gene's expression is not only temporally and spatially consistent with involvement in ethanol-induced behaviors but is also modulated by ethanol. The convergence of these data provides strong support to the hypothesis that GPC5 is involved in cellular and organismal ethanol response and the etiology of alcohol use disorders in humans.

15.
PLoS One ; 5(4): e9954, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20376353

RESUMEN

Dopamine is a mediator of the stimulant properties of drugs of abuse, including ethanol, in mammals and in the fruit fly Drosophila. The neural substrates for the stimulant actions of ethanol in flies are not known. We show that a subset of dopamine neurons and their targets, through the action of the D1-like dopamine receptor DopR, promote locomotor activation in response to acute ethanol exposure. A bilateral pair of dopaminergic neurons in the fly brain mediates the enhanced locomotor activity induced by ethanol exposure, and promotes locomotion when directly activated. These neurons project to the central complex ellipsoid body, a structure implicated in regulating motor behaviors. Ellipsoid body neurons are required for ethanol-induced locomotor activity and they express DopR. Elimination of DopR blunts the locomotor activating effects of ethanol, and this behavior can be restored by selective expression of DopR in the ellipsoid body. These data tie the activity of defined dopamine neurons to D1-like DopR-expressing neurons to form a neural circuit that governs acute responding to ethanol.


Asunto(s)
Dopamina/fisiología , Proteínas de Drosophila/metabolismo , Etanol/farmacología , Locomoción/efectos de los fármacos , Neuronas/fisiología , Receptores Dopaminérgicos/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central , Drosophila , Actividad Motora
16.
Alcohol Clin Exp Res ; 34(2): 302-16, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19951294

RESUMEN

BACKGROUND: Increased ethanol intake, a major predictor for the development of alcohol use disorders, is facilitated by the development of tolerance to both the aversive and pleasurable effects of the drug. The molecular mechanisms underlying ethanol tolerance development are complex and are not yet well understood. METHODS: To identify genetic mechanisms that contribute to ethanol tolerance, we examined the time course of gene expression changes elicited by a single sedating dose of ethanol in Drosophila, and completed a behavioral survey of strains harboring mutations in ethanol-regulated genes. RESULTS: Enrichment for genes in metabolism, nucleic acid binding, olfaction, regulation of signal transduction, and stress suggests that these biological processes are coordinately affected by ethanol exposure. We also detected a coordinate up-regulation of genes in the Toll and Imd innate immunity signal transduction pathways. A multi-study comparison revealed a small set of genes showing similar regulation, including increased expression of 3 genes for serine biosynthesis. A survey of Drosophila strains harboring mutations in ethanol-regulated genes for ethanol sensitivity and tolerance phenotypes revealed roles for serine biosynthesis, olfaction, transcriptional regulation, immunity, and metabolism. Flies harboring deletions of the genes encoding the olfactory co-receptor Or83b or the sirtuin Sir2 showed marked changes in the development of ethanol tolerance. CONCLUSIONS: Our findings implicate novel roles for these genes in regulating ethanol behavioral responses.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Tolerancia a Medicamentos/genética , Etanol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Animales , Conducta Animal/efectos de los fármacos , Coenzima A Ligasas/genética , Drosophila , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Femenino , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Masculino , Actividad Motora/efectos de los fármacos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Odorantes/biosíntesis , Receptores Odorantes/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serpinas/biosíntesis , Serpinas/genética , Sirtuinas/biosíntesis , Sirtuinas/genética , Especificidad de la Especie
17.
Neuron ; 64(4): 522-36, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19945394

RESUMEN

Arousal is fundamental to many behaviors, but whether it is unitary or whether there are different types of behavior-specific arousal has not been clear. In Drosophila, dopamine promotes sleep-wake arousal. However, there is conflicting evidence regarding its influence on environmentally stimulated arousal. Here we show that loss-of-function mutations in the D1 dopamine receptor DopR enhance repetitive startle-induced arousal while decreasing sleep-wake arousal (i.e., increasing sleep). These two types of arousal are also inversely influenced by cocaine, whose effects in each case are opposite to, and abrogated by, the DopR mutation. Selective restoration of DopR function in the central complex rescues the enhanced stimulated arousal but not the increased sleep phenotype of DopR mutants. These data provide evidence for at least two different forms of arousal, which are independently regulated by dopamine in opposite directions, via distinct neural circuits.


Asunto(s)
Nivel de Alerta/fisiología , Proteínas de Drosophila/fisiología , Red Nerviosa/fisiología , Receptores de Dopamina D1/fisiología , Receptores Dopaminérgicos/fisiología , Animales , Drosophila melanogaster , Humanos , Masculino
18.
Proc Natl Acad Sci U S A ; 104(11): 4653-7, 2007 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-17360579

RESUMEN

Habituation is a universal form of nonassociative learning that results in the devaluation of sensory inputs that have little information content. Although habituation is found throughout nature and has been studied in many organisms, the underlying molecular mechanisms remain poorly understood. We performed a forward genetic screen in Drosophila to search for mutations that modified habituation of an olfactory-mediated locomotor startle response, and we isolated a mutation in the glycogen synthase kinase-3 (GSK-3) homolog Shaggy. Decreases in Shaggy levels blunted habituation, whereas increases promoted habituation. Additionally, habituation acutely regulated Shaggy by an inhibitory phosphorylation mechanism, suggesting that a signal transduction pathway that regulates Shaggy is engaged during habituation. Although shaggy mutations also affected circadian rhythm period, this requirement was genetically separable from its role in habituation. Thus, shaggy functions in different neuronal circuits to regulate behavioral plasticity to an olfactory startle and circadian rhythmicity.


Asunto(s)
Proteínas de Drosophila/fisiología , Glucógeno Sintasa Quinasa 3/fisiología , Habituación Psicofisiológica , Alelos , Animales , Conducta Animal , Ritmo Circadiano , Drosophila , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Masculino , Modelos Genéticos , Mutación , Fosforilación , Transducción de Señal , Olfato
19.
Nat Neurosci ; 10(2): 169-76, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17237778

RESUMEN

During nervous system development, a small number of conserved guidance cues and receptors regulate many axon trajectories. How could a limited number of cues and receptors regulate such complex projection patterns? One way is to modulate receptor function. Here we show that the Caenorhabditis elegans kinesin-related protein VAB-8L, which is necessary and sufficient for posterior cell and growth-cone migrations, directs these migrations by regulating the levels of the guidance receptor SAX-3 (also known as robo). Genetic experiments indicate that VAB-8L and the Rac guanine nucleotide exchange factor activity of UNC-73 (trio) increase the ability of the SLT-1 (slit) and UNC-6 (netrin) guidance pathways to promote posterior guidance. The observations of higher SAX-3 receptor abundance in animals with increasing amounts of VAB-8L, and of physical interactions between UNC-73 and both VAB-8L and the intracellular domain of the SAX-3, support a model whereby VAB-8L directs cell and growth-cone migrations by promoting localization of guidance receptors to the cell surface.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Movimiento Celular/fisiología , Conos de Crecimiento/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/embriología , Receptores Inmunológicos/metabolismo , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Diferenciación Celular/fisiología , Señales (Psicología) , Conos de Crecimiento/ultraestructura , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/citología , Sistema Nervioso/metabolismo , Netrinas , Receptores Inmunológicos/genética , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteínas Roundabout
20.
Alcohol Clin Exp Res ; 30(2): 214-21, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16441270

RESUMEN

In recent years, it has become clear that growth factors are not only critical for the development of the central nervous system (CNS) but may also be important contributors to other neuronal functions in the adult brain. This symposium, presented at the 2005 RSA meeting, discussed evidence to support the hypothesis that alterations in growth factor pathways produce dramatic changes in the effects of alcohol on the CNS. The 4 speakers showed that the behavioral effects of alcohol in the adult are regulated by 3 growth factors, insulin, glial cell line-derived neurotrophic factor (GDNF), and brain-derived neurotrophic factor (BDNF). Dr. Wolf from the Heberlein laboratory presented findings obtained from genetic manipulations in Drosophila melanogaster, demonstrating that the insulin pathway controls sensitivity to the intoxicating effects of alcohol. Marian Logrip from the Ron and Janak laboratories presented evidence obtained in rodents that low concentrations of alcohol increase the expression of BDNF in the brain to regulate alcohol consumption. Dr. Pandey showed that amygdalar BDNF regulates alcohol's anxiolytic effects and preference. Finally, Dr. Janak presented evidence that increases in the expression of GDNF in the midbrain reduce alcohol self-administration in rats.


Asunto(s)
Alcoholismo/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/fisiología , Encéfalo/fisiopatología , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Insulina/fisiología , Alcoholismo/genética , Animales , Mapeo Encefálico , Drosophila melanogaster , Humanos , Ratones , Ratas , Investigación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...