Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
PLoS Genet ; 20(1): e1011054, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38236837

RESUMO

Living in dynamic environments such as the social domain, where interaction with others determines the reproductive success of individuals, requires the ability to recognize opportunities to obtain natural rewards and cope with challenges that are associated with achieving them. As such, actions that promote survival and reproduction are reinforced by the brain reward system, whereas coping with the challenges associated with obtaining these rewards is mediated by stress-response pathways, the activation of which can impair health and shorten lifespan. While much research has been devoted to understanding mechanisms underlying the way by which natural rewards are processed by the reward system, less attention has been given to the consequences of failure to obtain a desirable reward. As a model system to study the impact of failure to obtain a natural reward, we used the well-established courtship suppression paradigm in Drosophila melanogaster as means to induce repeated failures to obtain sexual reward in male flies. We discovered that beyond the known reduction in courtship actions caused by interaction with non-receptive females, repeated failures to mate induce a stress response characterized by persistent motivation to obtain the sexual reward, reduced male-male social interaction, and enhanced aggression. This frustrative-like state caused by the conflict between high motivation to obtain sexual reward and the inability to fulfill their mating drive impairs the capacity of rejected males to tolerate stressors such as starvation and oxidative stress. We further show that sensitivity to starvation and enhanced social arousal is mediated by the disinhibition of a small population of neurons that express receptors for the fly homologue of neuropeptide Y. Our findings demonstrate for the first time the existence of social stress in flies and offers a framework to study mechanisms underlying the crosstalk between reward, stress, and reproduction in a simple nervous system that is highly amenable to genetic manipulation.


Assuntos
Drosophila melanogaster , Neuropeptídeos , Comportamento Sexual Animal , Humanos , Animais , Feminino , Masculino , Drosophila melanogaster/genética , Comportamento Sexual Animal/fisiologia , Reprodução/genética , Recompensa , Neurônios/metabolismo
2.
Cell ; 137(5): 949-60, 2009 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-19464045

RESUMO

The consequences of alcohol use disorders (AUDs) are devastating to individuals and society, yet few treatments are currently available. To identify genes regulating the behavioral effects of ethanol, we conducted a genetic screen in Drosophila and identified a mutant, happyhour (hppy), due to its increased resistance to the sedative effects of ethanol. Hppy protein shows strong homology to mammalian Ste20 family kinases of the GCK-1 subfamily. Genetic and biochemical experiments revealed that the epidermal growth factor (EGF)-signaling pathway regulates ethanol sensitivity in Drosophila and that Hppy functions as an inhibitor of the pathway. Acute pharmacological inhibition of the EGF receptor (EGFR) in adult animals altered acute ethanol sensitivity in both flies and mice and reduced ethanol consumption in a preclinical rat model of alcoholism. Inhibitors of the EGFR or components of its signaling pathway are thus potential pharmacotherapies for AUDs.


Assuntos
Transtornos Induzidos por Álcool/metabolismo , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Receptores ErbB/metabolismo , Etanol/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Animais , Cruzamentos Genéticos , Dopamina/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/química , Drosophila melanogaster/genética , Feminino , Insulina/metabolismo , Masculino , Camundongos , Mutação , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética
3.
Annu Rev Neurosci ; 36: 121-38, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23642133

RESUMO

Animal models have been widely used to gain insight into the mechanisms underlying the acute and long-term effects of alcohol exposure. The fruit fly Drosophila melanogaster encounters ethanol in its natural habitat and possesses many adaptations that allow it to survive and thrive in ethanol-rich environments. Several assays to study ethanol-related behaviors in flies, ranging from acute intoxication to self-administration and reward, have been developed in the past 20 years. These assays have provided the basis for studying the physiological and behavioral effects of ethanol and for identifying genes mediating these effects. In this review we describe the ecological relationship between flies and ethanol, the effects of ethanol on fly development and behavior, the use of flies as a model for alcohol addiction, and the interaction between ethanol and social behavior. We discuss these advances in the context of their utility to help decipher the mechanisms underlying the diverse effects of ethanol, including those that mediate ethanol dependence and addiction in humans.


Assuntos
Alcoolismo/fisiopatologia , Evolução Biológica , Modelos Animais de Doenças , Etanol/efeitos adversos , Alcoolismo/psicologia , Animais , Drosophila melanogaster , Etanol/administração & dosagem , Humanos , Recompensa , Autoadministração
4.
Proc Natl Acad Sci U S A ; 115(5): 1099-1104, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29339481

RESUMO

Multiple studies have investigated the mechanisms of aggressive behavior in Drosophila; however, little is known about the effects of chronic fighting experience. Here, we investigated if repeated fighting encounters would induce an internal state that could affect the expression of subsequent behavior. We trained wild-type males to become winners or losers by repeatedly pairing them with hypoaggressive or hyperaggressive opponents, respectively. As described previously, we observed that chronic losers tend to lose subsequent fights, while chronic winners tend to win them. Olfactory conditioning experiments showed that winning is perceived as rewarding, while losing is perceived as aversive. Moreover, the effect of chronic fighting experience generalized to other behaviors, such as gap-crossing and courtship. We propose that in response to repeatedly winning or losing aggressive encounters, male flies form an internal state that displays persistence and generalization; fight outcomes can also have positive or negative valence. Furthermore, we show that the activities of the PPL1-γ1pedc dopaminergic neuron and the MBON-γ1pedc>α/ß mushroom body output neuron are required for aversion to an olfactory cue associated with losing fights.


Assuntos
Agressão/fisiologia , Comportamento Animal/fisiologia , Drosophila melanogaster/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Análise por Conglomerados , Comportamento Competitivo , Cruzamentos Genéticos , Feminino , Masculino , Memória , Movimento , Neurônios/metabolismo , Odorantes , Bulbo Olfatório , Assunção de Riscos , Fatores de Tempo
5.
J Exp Biol ; 223(Pt 24)2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33268534

RESUMO

Social interactions pivot on an animal's experiences, internal states and feedback from others. This complexity drives the need for precise descriptions of behavior to dissect the fine detail of its genetic and neural circuit bases. In laboratory assays, male Drosophila melanogaster reliably exhibit aggression, and its extent is generally measured by scoring lunges, a feature of aggression in which one male quickly thrusts onto his opponent. Here, we introduce an explicit approach to identify both the onset and reversals in hierarchical status between opponents and observe that distinct aggressive acts reproducibly precede, concur or follow the establishment of dominance. We find that lunges are insufficient for establishing dominance. Rather, lunges appear to reflect the dominant state of a male and help in maintaining his social status. Lastly, we characterize the recurring and escalating structure of aggression that emerges through subsequent reversals in dominance. Collectively, this work provides a framework for studying the complexity of agonistic interactions in male flies, enabling its neurogenetic basis to be understood with precision.


Assuntos
Agressão , Drosophila melanogaster , Animais , Comportamento Animal , Drosophila melanogaster/genética , Hierarquia Social , Masculino , Predomínio Social
6.
Proc Natl Acad Sci U S A ; 114(38): E8091-E8099, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28874527

RESUMO

In their classic experiments, Olds and Milner showed that rats learn to lever press to receive an electric stimulus in specific brain regions. This led to the identification of mammalian reward centers. Our interest in defining the neuronal substrates of reward perception in the fruit fly Drosophila melanogaster prompted us to develop a simpler experimental approach wherein flies could implement behavior that induces self-stimulation of specific neurons in their brains. The high-throughput assay employs optogenetic activation of neurons when the fly occupies a specific area of a behavioral chamber, and the flies' preferential occupation of this area reflects their choosing to experience optogenetic stimulation. Flies in which neuropeptide F (NPF) neurons are activated display preference for the illuminated side of the chamber. We show that optogenetic activation of NPF neuron is rewarding in olfactory conditioning experiments and that the preference for NPF neuron activation is dependent on NPF signaling. Finally, we identify a small subset of NPF-expressing neurons located in the dorsomedial posterior brain that are sufficient to elicit preference in our assay. This assay provides the means for carrying out unbiased screens to map reward neurons in flies.


Assuntos
Proteínas de Drosophila/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Neuropeptídeos/genética
7.
BMC Biol ; 17(1): 30, 2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30967153

RESUMO

BACKGROUND: Epigenetic mechanisms play fundamental roles in brain function and behavior and stressors such as social isolation can alter animal behavior via epigenetic mechanisms. However, due to cellular heterogeneity, identifying cell-type-specific epigenetic changes in the brain is challenging. Here, we report the first use of a modified isolation of nuclei tagged in specific cell type (INTACT) method in behavioral epigenetics of Drosophila melanogaster, a method we call mini-INTACT. RESULTS: Using ChIP-seq on mini-INTACT purified dopaminergic nuclei, we identified epigenetic signatures in socially isolated and socially enriched Drosophila males. Social experience altered the epigenetic landscape in clusters of genes involved in transcription and neural function. Some of these alterations could be predicted by expression changes of four transcription factors and the prevalence of their binding sites in several clusters. These transcription factors were previously identified as activity-regulated genes, and their knockdown in dopaminergic neurons reduced the effects of social experience on sleep. CONCLUSIONS: Our work enables the use of Drosophila as a model for cell-type-specific behavioral epigenetics and establishes that social environment shifts the epigenetic landscape in dopaminergic neurons. Four activity-related transcription factors are required in dopaminergic neurons for the effects of social environment on sleep.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Drosophila melanogaster/genética , Epigênese Genética/genética , Genética Comportamental/métodos , Meio Social , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Epigenômica/métodos , Masculino , Modelos Animais , Sono/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
J Neurosci ; 35(26): 9638-47, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26134647

RESUMO

The ability to use environmental cues to predict rewarding events is essential to survival. The basolateral amygdala (BLA) plays a central role in such forms of associative learning. Aberrant cue-reward learning is thought to underlie many psychopathologies, including addiction, so understanding the underlying molecular mechanisms can inform strategies for intervention. The transcriptional regulator LIM-only 4 (LMO4) is highly expressed in pyramidal neurons of the BLA, where it plays an important role in fear learning. Because the BLA also contributes to cue-reward learning, we investigated the role of BLA LMO4 in this process using Lmo4-deficient mice and RNA interference. Lmo4-deficient mice showed a selective deficit in conditioned reinforcement. Knockdown of LMO4 in the BLA, but not in the nucleus accumbens, recapitulated this deficit in wild-type mice. Molecular and electrophysiological studies identified a deficit in dopamine D2 receptor signaling in the BLA of Lmo4-deficient mice. These results reveal a novel, LMO4-dependent transcriptional program within the BLA that is essential to cue-reward learning.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aprendizagem por Associação/fisiologia , Comportamento de Escolha/fisiologia , Sinais (Psicologia) , Proteínas com Domínio LIM/metabolismo , Recompensa , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Complexo Nuclear Basolateral da Amígdala/citologia , Condicionamento Operante/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Proteínas com Domínio LIM/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/fisiologia , Técnicas de Patch-Clamp , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Sacarose/administração & dosagem
9.
Proc Natl Acad Sci U S A ; 110(52): 21153-8, 2013 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-24324162

RESUMO

The neural circuits that mediate behavioral choice evaluate and integrate information from the environment with internal demands and then initiate a behavioral response. Even circuits that support simple decisions remain poorly understood. In Drosophila melanogaster, oviposition on a substrate containing ethanol enhances fitness; however, little is known about the neural mechanisms mediating this important choice behavior. Here, we characterize the neural modulation of this simple choice and show that distinct subsets of dopaminergic neurons compete to either enhance or inhibit egg-laying preference for ethanol-containing food. Moreover, activity in α'ß' neurons of the mushroom body and a subset of ellipsoid body ring neurons (R2) is required for this choice. We propose a model where competing dopaminergic systems modulate oviposition preference to adjust to changes in natural oviposition substrates.


Assuntos
Comportamento de Escolha/fisiologia , Neurônios Dopaminérgicos/fisiologia , Drosophila melanogaster/fisiologia , Etanol/metabolismo , Oviposição/fisiologia , Análise de Variância , Animais , Neurônios Dopaminérgicos/metabolismo , Feminino , Fermentação , Frutas/metabolismo , Corpos Pedunculados/fisiologia
10.
Proc Natl Acad Sci U S A ; 109(51): 21087-92, 2012 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-23213244

RESUMO

In mammalian and insect models of ethanol intoxication, low doses of ethanol stimulate locomotor activity whereas high doses induce sedation. Sex differences in acute ethanol responses, which occur in humans, have not been characterized in Drosophila. In this study, we find that male flies show increased ethanol hyperactivity and greater resistance to ethanol sedation compared with females. We show that the sex determination gene transformer (tra) acts in the developing nervous system, likely through regulation of fruitless (fru), to at least partially mediate the sexual dimorphism in ethanol sedation. Although pharmacokinetic differences may contribute to the increased sedation sensitivity of females, neuronal tra expression regulates ethanol sedation independently of ethanol pharmacokinetics. We also show that acute activation of fru-expressing neurons affects ethanol sedation, further supporting a role for fru in regulating this behavior. Thus, we have characterized previously undescribed sex differences in behavioral responses to ethanol, and implicated fru in mediating a subset of these differences.


Assuntos
Drosophila melanogaster/metabolismo , Etanol/farmacocinética , Regulação da Expressão Gênica , Caracteres Sexuais , Intoxicação Alcoólica , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Cruzamentos Genéticos , Modelos Animais de Doenças , Feminino , Masculino , Modelos Estatísticos , Neurônios/efeitos dos fármacos , Fatores Sexuais
11.
J Neurosci ; 33(9): 4044-54, 2013 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-23447613

RESUMO

In the fruit fly Drosophila melanogaster, as in mammals, acute exposure to a high dose of ethanol leads to stereotypical behavioral changes beginning with increased activity, followed by incoordination, loss of postural control, and eventually, sedation. The mechanism(s) by which ethanol impacts the CNS leading to ethanol-induced sedation and the genes required for normal sedation sensitivity remain largely unknown. Here we identify the gene apontic (apt), an Myb/SANT-containing transcription factor that is required in the nervous system for normal sensitivity to ethanol sedation. Using genetic and behavioral analyses, we show that apt mediates sensitivity to ethanol sedation by acting in a small set of neurons that express Corazonin (Crz), a neuropeptide likely involved in the physiological response to stress. The activity of Crz neurons regulates the behavioral response to ethanol, as silencing and activating these neurons affects sedation sensitivity in opposite ways. Furthermore, this effect is mediated by Crz, as flies with reduced crz expression show reduced sensitivity to ethanol sedation. Finally, we find that both apt and crz are rapidly upregulated by acute ethanol exposure. Thus, we have identified two genes and a small set of peptidergic neurons that regulate sensitivity to ethanol-induced sedation. We propose that Apt regulates the activity of Crz neurons and/or release of the neuropeptide during ethanol exposure.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Etanol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipnóticos e Sedativos/farmacologia , Neurônios/efeitos dos fármacos , Neuropeptídeos/metabolismo , Sistemas Neurossecretores/citologia , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Comportamento Animal/efeitos dos fármacos , Encéfalo/citologia , Proteínas de Ligação a DNA/genética , Relação Dose-Resposta a Droga , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Metamorfose Biológica/genética , Mutação/genética , Neurônios/metabolismo , Neuropeptídeos/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/genética
12.
J Neurosci ; 33(19): 8134-43, 2013 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-23658154

RESUMO

In both mammalian and insect models of ethanol intoxication, high doses of ethanol induce motor impairment and eventually sedation. Sensitivity to the sedative effects of ethanol is inversely correlated with risk for alcoholism. However, the genes regulating ethanol sensitivity are largely unknown. Based on a previous genetic screen in Drosophila for ethanol sedation mutants, we identified a novel gene, tank (CG15626), the homolog of the mammalian tumor suppressor EI24/PIG8, which has a strong role in regulating ethanol sedation sensitivity. Genetic and behavioral analyses revealed that tank acts in the adult nervous system to promote ethanol sensitivity. We localized the function of tank in regulating ethanol sensitivity to neurons within the pars intercerebralis that have not been implicated previously in ethanol responses. We show that acutely manipulating the activity of all tank-expressing neurons, or of pars intercerebralis neurons in particular, alters ethanol sensitivity in a sexually dimorphic manner, since neuronal activation enhanced ethanol sedation in males, but not females. Finally, we provide anatomical evidence that tank-expressing neurons form likely synaptic connections with neurons expressing the neural sex determination factor fruitless (fru), which have been implicated recently in the regulation of ethanol sensitivity. We suggest that a functional interaction with fru neurons, many of which are sexually dimorphic, may account for the sex-specific effect induced by activating tank neurons. Overall, we have characterized a novel gene and corresponding set of neurons that regulate ethanol sensitivity in Drosophila.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Mutação/genética , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Análise de Variância , Animais , Animais Geneticamente Modificados , Proteínas Reguladoras de Apoptose/genética , Comportamento Animal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Canais Iônicos , Biologia Molecular , Proteínas Nucleares/genética , Interferência de RNA/fisiologia , Canal de Cátion TRPA1 , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo
13.
J Neurosci ; 31(40): 14134-41, 2011 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-21976498

RESUMO

Previously, we showed that the mouse LIM-domain only 4 (Lmo4) gene, which encodes a protein containing two zinc-finger LIM domains that interact with various DNA-binding transcription factors, attenuates behavioral sensitivity to repeated cocaine administration. Here we show that transcription of anaplastic lymphoma kinase (Alk) is repressed by LMO4 in the striatum and that Alk promotes the development of cocaine sensitization and conditioned place preference, a measure of cocaine reward. Since LMO4 is known to interact with estrogen receptor α (ERα) at the promoters of target genes, we investigated whether Alk expression might be controlled by a similar mechanism. We found that LMO4 and ERα are associated with the Alk promoter by chromatin immunoprecipitation and that Alk is an estrogen-responsive gene in the striatum. Moreover, we show that ERα knock-out mice exhibit enhanced cocaine sensitization and conditioned place preference and an increase in Alk expression in the nucleus accumbens. These data define a novel regulatory network involved in behavioral responses to cocaine. Interestingly, sex differences in several behavioral responses to cocaine in humans and rodents have been described, and estrogen is thought to mediate some of these differences. Our data suggest that estrogen regulation of Alk may be one mechanism responsible for sexually dimorphic responses to cocaine.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Cocaína/farmacologia , Receptor alfa de Estrogênio/biossíntese , Marcação de Genes/métodos , Proteínas com Domínio LIM/biossíntese , Receptores Proteína Tirosina Quinases/biossíntese , Recompensa , Transcrição Gênica/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Quinase do Linfoma Anaplásico , Animais , Animais Recém-Nascidos , Células Cultivadas , Cocaína/metabolismo , Receptor alfa de Estrogênio/genética , Células HEK293 , Humanos , Proteínas com Domínio LIM/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Receptores Proteína Tirosina Quinases/genética , Transcrição Gênica/efeitos dos fármacos
14.
J Neurosci ; 31(3): 1139-48, 2011 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-21248138

RESUMO

In both mammalian and insect models of ethanol-induced behavior, low doses of ethanol stimulate locomotion. However, the mechanisms of the stimulant effects of ethanol on the CNS are mostly unknown. We have identified tao, encoding a serine-threonine kinase of the Ste20 family, as a gene necessary for ethanol-induced locomotor hyperactivity in Drosophila. Mutations in tao also affect behavioral responses to cocaine and nicotine, making flies resistant to the effects of both drugs. We show that tao function is required during the development of the adult nervous system and that tao mutations cause defects in the development of central brain structures, including the mushroom body. Silencing of a subset of mushroom body neurons is sufficient to reduce ethanol-induced hyperactivity, revealing the mushroom body as an important locus mediating the stimulant effects of ethanol. We also show that mutations in par-1 suppress both the mushroom body morphology and behavioral phenotypes of tao mutations and that the phosphorylation state of the microtubule-binding protein Tau can be altered by RNA interference knockdown of tao, suggesting that tao and par-1 act in a pathway to control microtubule dynamics during neural development.


Assuntos
Proteínas de Drosophila/metabolismo , Etanol/farmacologia , Atividade Motora/fisiologia , Corpos Pedunculados/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Western Blotting , Drosophila , Hipercinese/induzido quimicamente , Hipercinese/metabolismo , Imuno-Histoquímica , Metamorfose Biológica , Atividade Motora/efeitos dos fármacos , Corpos Pedunculados/efeitos dos fármacos , Neurônios/metabolismo
15.
Hum Genet ; 131(6): 959-75, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22350798

RESUMO

Animal studies have been instrumental in providing knowledge about the molecular and neural mechanisms underlying drug addiction. Recently, the fruit fly Drosophila melanogaster has become a valuable system to model not only the acute stimulating and sedating effects of drugs but also their more complex rewarding properties. In this review, we describe the advantages of using the fly to study drug-related behavior, provide a brief overview of the behavioral assays used, and review the molecular mechanisms and neural circuits underlying drug-induced behavior in flies. Many of these mechanisms have been validated in mammals, suggesting that the fly is a useful model to understand the mechanisms underlying addiction.


Assuntos
Comportamento Animal/fisiologia , Modelos Animais de Doenças , Drosophila melanogaster , Etanol/toxicidade , Transtornos Relacionados ao Uso de Substâncias/genética , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transmissão Sináptica/fisiologia , Anfetaminas/efeitos adversos , Animais , Comportamento Animal/efeitos dos fármacos , Cocaína/efeitos adversos , Vias Neurais/efeitos dos fármacos , Nicotina/efeitos adversos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética
16.
Proc Natl Acad Sci U S A ; 106(49): 20924-9, 2009 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-19923438

RESUMO

It has long been known that heavy alcohol consumption leads to neuropathology and neuronal death. While the response of neurons to an ethanol insult is strongly influenced by genetic background, the underlying mechanisms are poorly understood. Here, we show that even a single intoxicating exposure to ethanol causes non-cell-autonomous apoptotic death specifically of Drosophila olfactory neurons, which is accompanied by a loss of a behavioral response to the smell of ethanol and a blackening of the third antennal segment. The Drosophila homolog of glycogen synthase kinase-3 (GSK-3)beta, Shaggy, is required for ethanol-induced apoptosis. Consistent with this requirement, the GSK-3beta inhibitor lithium protects against the neurotoxic effects of ethanol, indicating the possibility for pharmacological intervention in cases of alcohol-induced neurodegeneration. Ethanol-induced death of olfactory neurons requires both their neural activity and functional NMDA receptors. This system will allow the investigation of the genetic and molecular basis of ethanol-induced apoptosis in general and provide an understanding of the molecular role of GSK-3beta in programmed cell death.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/enzimologia , Etanol/toxicidade , Quinase 3 da Glicogênio Sintase/metabolismo , Neurotoxinas/toxicidade , Neurônios Receptores Olfatórios/enzimologia , Neurônios Receptores Olfatórios/patologia , Animais , Morte Celular/efeitos dos fármacos , Drosophila melanogaster/efeitos dos fármacos , Eletricidade , Inativação Gênica/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta , Lítio/farmacologia , Neurônios Receptores Olfatórios/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Homologia de Sequência de Aminoácidos , Volatilização/efeitos dos fármacos
17.
Proc Natl Acad Sci U S A ; 106(27): 11352-7, 2009 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-19541615

RESUMO

Selection of appropriate oviposition sites is essential for progeny survival and fitness in generalist insect species, such as Drosophila melanogaster, yet little is known about the mechanisms regulating how environmental conditions and innate adult preferences are evaluated and balanced to yield the final substrate choice for egg-deposition. Female D. melanogaster are attracted to food containing acetic acid (AA) as an oviposition substrate. However, our observations reveal that this egg-laying preference is a complex process, as it directly opposes an otherwise strong, default behavior of positional avoidance for the same food. We show that 2 distinct sensory modalities detect AA. Attraction to AA-containing food for the purpose of egg-laying relies on the gustatory system, while positional repulsion depends primarily on the olfactory system. Similarly, distinct central brain regions are involved in AA attraction and repulsion. Given this unique situation, in which a single environmental stimulus yields 2 opposing behavioral outputs, we propose that the interaction of egg-laying attraction and positional aversion for AA provides a powerful model for studying how organisms balance competing behavioral drives and integrate signals involved in choice-like processes.


Assuntos
Ácido Acético/farmacologia , Aprendizagem da Esquiva/efeitos dos fármacos , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/fisiologia , Modelos Biológicos , Oviposição/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Ração Animal , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Condutos Olfatórios/efeitos dos fármacos , Paladar/efeitos dos fármacos
18.
J Neurosci ; 30(26): 8830-40, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20592205

RESUMO

There is considerable interest in the regulation of sensorimotor gating, since deficits in this process could play a critical role in the symptoms of schizophrenia and other psychiatric disorders. Sensorimotor gating is often studied in humans and rodents using the prepulse inhibition of the acoustic startle response (PPI) model, in which an acoustic prepulse suppresses behavioral output to a startle-inducing stimulus. However, the molecular and neural mechanisms underlying PPI are poorly understood. Here, we show that a regulatory pathway involving protein phosphatase 2A (PP2A), glycogen synthase kinase 3 beta (GSK3beta), and their downstream target, the M-type potassium channel, regulates PPI. Mice (Mus musculus) carrying a hypomorphic allele of Ppp2r5delta, encoding a regulatory subunit of PP2A, show attenuated PPI. This PPP2R5delta reduction increases the phosphorylation of GSK3beta at serine 9, which inactivates GSK3beta, indicating that PPP2R5delta positively regulates GSK3beta activity in the brain. Consistently, genetic and pharmacological manipulations that reduce GSK3beta function attenuate PPI. The M-type potassium channel subunit, KCNQ2, is a putative GSK3beta substrate. Genetic reduction of Kcnq2 also reduces PPI, as does systemic inhibition of M-channels with linopirdine. Importantly, both the GSK3 inhibitor 3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)1H-pyrrole-2,5-dione (SB216763) and linopirdine reduce PPI when directly infused into the medial prefrontal cortex (mPFC). Whole-cell electrophysiological recordings of mPFC neurons show that SB216763 and linopirdine have similar effects on firing, and GSK3 inhibition occludes the effects of M-channel inhibition. These data support a previously uncharacterized mechanism by which PP2A/GSK3beta signaling regulates M-type potassium channel activity in the mPFC to modulate sensorimotor gating.


Assuntos
Percepção Auditiva/fisiologia , Encéfalo/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Inibição Psicológica , Canais de Potássio/metabolismo , Proteína Fosfatase 2/metabolismo , Sequência de Aminoácidos , Animais , Percepção Auditiva/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fosforilação , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Proteína Fosfatase 2/genética , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/fisiologia , Transdução de Sinais
19.
Alcohol Clin Exp Res ; 35(9): 1600-6, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21599714

RESUMO

BACKGROUND: Previous work from our laboratory demonstrated a role for the Drosophila Lim-only (dLmo) gene in regulating behavioral responses to cocaine. Herein, we examined whether dLmo influences the flies' sensitivity to ethanol's sedating effects. We also investigated whether 1 of the mammalian homologs of dLmo, Lmo3, is involved in behavioral responses to ethanol in mice. METHODS: To examine dLmo function in ethanol-induced sedation, mutant flies with reduced or increased dLmo expression were tested using the loss of righting (LOR) assay. To determine whether mouse Lmo3 regulates behavioral responses to ethanol, we generated transgenic mice expressing a short-hairpin RNA targeting Lmo3 for RNA interference-mediated knockdown by lentiviral infection of single cell embryos. Adult founder mice, expressing varying amounts of Lmo3 in the brain, were tested using ethanol loss-of-righting-reflex (LORR) and 2-bottle choice ethanol consumption assays. RESULTS: We found that in flies, reduced dLmo activity increased sensitivity to ethanol-induced sedation, whereas increased expression of dLmo led to increased resistance to ethanol-induced sedation. In mice, reduced levels of Lmo3 were correlated with increased sedation time in the LORR test and decreased ethanol consumption in the 2-bottle choice protocol. CONCLUSIONS: These data describe a novel and conserved role for Lmo genes in flies and mice in behavioral responses to ethanol. These studies also demonstrate the feasibility of rapidly translating findings from invertebrate systems to mammalian models of alcohol abuse by combining RNA interference in transgenic mice and behavioral testing.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Depressores do Sistema Nervoso Central/farmacologia , Proteínas de Drosophila/genética , Etanol/farmacologia , Proteínas de Homeodomínio/genética , Proteínas com Domínio LIM/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Depressores do Sistema Nervoso Central/farmacocinética , Modelos Animais de Doenças , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Etanol/farmacocinética , Feminino , Genótipo , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/metabolismo , Hipnóticos e Sedativos/análise , Proteínas com Domínio LIM/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Interferente Pequeno/genética
20.
Nature ; 436(7052): 845-7, 2005 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-16094367

RESUMO

Repeated alcohol consumption leads to the development of tolerance, simply defined as an acquired resistance to the physiological and behavioural effects of the drug. This tolerance allows increased alcohol consumption, which over time leads to physical dependence and possibly addiction. Previous studies have shown that Drosophila develop ethanol tolerance, with kinetics of acquisition and dissipation that mimic those seen in mammals. This tolerance requires the catecholamine octopamine, the functional analogue of mammalian noradrenaline. Here we describe a new gene, hangover, which is required for normal development of ethanol tolerance. hangover flies are also defective in responses to environmental stressors, such as heat and the free-radical-generating agent paraquat. Using genetic epistasis tests, we show that ethanol tolerance in Drosophila relies on two distinct molecular pathways: a cellular stress pathway defined by hangover, and a parallel pathway requiring octopamine. hangover encodes a large nuclear zinc-finger protein, suggesting a role in nucleic acid binding. There is growing recognition that stress, at both the cellular and systemic levels, contributes to drug- and addiction-related behaviours in mammals. Our studies suggest that this role may be conserved across evolution.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/genética , Tolerância a Medicamentos/genética , Etanol/farmacologia , Estresse Fisiológico/genética , Estresse Fisiológico/fisiopatologia , Alcoolismo/genética , Alcoolismo/fisiopatologia , Animais , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Resposta ao Choque Térmico/genética , Resposta ao Choque Térmico/fisiologia , Mutação/genética , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA