Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Elife ; 92020 06 22.
Article in English | MEDLINE | ID: mdl-32568072

ABSTRACT

Among the brainstem raphe nuclei, the dorsal raphe nucleus (DR) contains the greatest number of Pet1-lineage neurons, a predominantly serotonergic group distributed throughout DR subdomains. These neurons collectively regulate diverse physiology and behavior and are often therapeutically targeted to treat affective disorders. Characterizing Pet1 neuron molecular heterogeneity and relating it to anatomy is vital for understanding DR functional organization, with potential to inform therapeutic separability. Here we use high-throughput and DR subdomain-targeted single-cell transcriptomics and intersectional genetic tools to map molecular and anatomical diversity of DR-Pet1 neurons. We describe up to fourteen neuron subtypes, many showing biased cell body distributions across the DR. We further show that P2ry1-Pet1 DR neurons - the most molecularly distinct subtype - possess unique efferent projections and electrophysiological properties. These data complement and extend previous DR characterizations, combining intersectional genetics with multiple transcriptomic modalities to achieve fine-scale molecular and anatomic identification of Pet1 neuron subtypes.


Subject(s)
Dorsal Raphe Nucleus/anatomy & histology , Mice/anatomy & histology , Mice/genetics , Neurons , Transcriptome , Animals , Dorsal Raphe Nucleus/metabolism , Female , Gene Expression Profiling , Male , Mice, Inbred C57BL , Neurons/metabolism , Single-Cell Analysis , Transcription Factors/metabolism
2.
Curr Biol ; 29(13): 2145-2156.e5, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31231050

ABSTRACT

Pathological aggression is commonly associated with psychiatric and neurological disorders and can impose a substantial burden and cost on human society. Serotonin (5HT) has long been implicated in the regulation of aggression in a wide variety of animal species. In Drosophila, a small group of serotonergic neurons selectively modulates the escalation of aggression. Here, we identified downstream targets of serotonergic input-two types of neurons with opposing roles in aggression control. The dendritic fields of both neurons converge on a single optic glomerulus LC12, suggesting a key pathway linking visual input to the aggression circuitry. The first type is an inhibitory GABAergic neuron: its activation leads to a decrease in aggression. The second neuron type is excitatory: its silencing reduces and its activation increases aggression. RNA sequencing (RNA-seq) profiling of this neuron type identified that it uses acetylcholine as a neurotransmitter and likely expresses 5HT1A, short neuropeptide F receptor (sNPFR), and the resistant to dieldrin (RDL) category of GABA receptors. Knockdown of RDL receptors in these neurons increases aggression, suggesting the possibility of a direct crosstalk between the inhibitory GABAergic and the excitatory cholinergic neurons. Our data show further that neurons utilizing serotonin, GABA, ACh, and short neuropeptide F interact in the LC12 optic glomerulus. Parallel cholinergic and GABAergic pathways descending from this sensory integration area may be key elements in fine-tuning the regulation of aggression.


Subject(s)
Cholinergic Neurons/physiology , Drosophila melanogaster/physiology , GABAergic Neurons/physiology , Serotonergic Neurons/physiology , Serotonin/metabolism , Aggression/physiology , Animals
3.
PLoS One ; 10(9): e0138181, 2015.
Article in English | MEDLINE | ID: mdl-26383635

ABSTRACT

To study the molecular mechanism of complex biological systems, it is important to be able to artificially manipulate gene expression in desired target sites with high precision. Based on the light dependent binding of cryptochrome 2 and a cryptochrome interacting bHLH protein, we developed a split lexA transcriptional activation system for use in Drosophila that allows regulation of gene expression in vivo using blue light or two-photon excitation. We show that this system offers high spatiotemporal resolution by inducing gene expression in tissues at various developmental stages. In combination with two-photon excitation, gene expression can be manipulated at precise sites in embryos, potentially offering an important tool with which to examine developmental processes.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Light , Optogenetics , Animals , Animals, Genetically Modified , Cells, Cultured , Cryptochromes/genetics , Cryptochromes/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/radiation effects , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/radiation effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Neurons/metabolism , Transcriptional Activation/radiation effects , Transgenes/radiation effects
4.
Curr Biol ; 24(22): 2700-7, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25447998

ABSTRACT

Monoamine serotonin (5HT) has been linked to aggression for many years across species. However, elaboration of the neurochemical pathways that govern aggression has proven difficult because monoaminergic neurons also regulate other behaviors. There are approximately 100 serotonergic neurons in the Drosophila nervous system, and they influence sleep, circadian rhythms, memory, and courtship. In the Drosophila model of aggression, the acute shut down of the entire serotonergic system yields flies that fight less, whereas induced activation of 5HT neurons promotes aggression. Using intersectional genetics, we restricted the population of 5HT neurons that can be reproducibly manipulated to identify those that modulate aggression. Although similar approaches were used recently to find aggression-modulating dopaminergic and Fru(M)-positive peptidergic neurons, the downstream anatomical targets of the neurons that make up aggression-controlling circuits remain poorly understood. Here, we identified a symmetrical pair of serotonergic PLP neurons that are necessary for the proper escalation of aggression. Silencing these neurons reduced aggression in male flies, and activating them increased aggression in male flies. GFP reconstitution across synaptic partners (GRASP) analyses suggest that 5HT-PLP neurons form contacts with 5HT1A receptor-expressing neurons in two distinct anatomical regions of the brain. Activation of these 5HT1A receptor-expressing neurons, in turn, caused reductions in aggression. Our studies, therefore, suggest that aggression may be held in check, at least in part, by inhibitory input from 5HT1A receptor-bearing neurons, which can be released by activation of the 5HT-PLP neurons.


Subject(s)
Aggression/physiology , Behavior, Animal/physiology , Drosophila/physiology , Serotonergic Neurons/physiology , Animals , Animals, Genetically Modified/physiology , Drosophila/genetics , Transgenes
5.
Fly (Austin) ; 8(4): 200-5, 2014.
Article in English | MEDLINE | ID: mdl-25923771

ABSTRACT

All species of animals display aggression in order to obtain resources such as territories, mates, or food. Appropriate displays of aggression rely on the correct identification of a potential competitor, an evaluation of the environmental signals, and the physiological state of the animal. With a hard-wired circuitry involving fixed numbers of neurons, neuromodulators like serotonin offer adaptive flexibility in behavioral responses without changing the "hard-wiring". In a recent report, we combined intersectional genetics, quantitative behavioral assays and morphological analyses to identify single serotonergic neurons that modulate the escalation of aggression. We found anatomical target areas within the brain where these neurons appear to form synaptic contacts with 5HT1A receptor-expressing neurons, and then confirmed the likelihood of those connections on a functional level. In this Extra View article, we offer an extended discussion of these recent findings and elaborate on how they can link a cellular and functional mapping of an aggression-regulating circuit at a single-cell resolution level.


Subject(s)
Aggression/physiology , Drosophila/metabolism , Serotonergic Neurons/physiology , Serotonin/metabolism , Animals , Brain/anatomy & histology , Brain/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Gene Expression Regulation/physiology , Male , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism
6.
Proc Natl Acad Sci U S A ; 110(15): 6151-6, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23530210

ABSTRACT

Monoamines, including dopamine (DA), have been linked to aggression in various species. However, the precise role or roles served by the amine in aggression have been difficult to define because dopaminergic systems influence many behaviors, and all can be altered by changing the function of dopaminergic neurons. In the fruit fly, with the powerful genetic tools available, small subsets of brain cells can be reliably manipulated, offering enormous advantages for exploration of how and where amine neurons fit into the circuits involved with aggression. By combining the GAL4/upstream activating sequence (UAS) binary system with the Flippase (FLP) recombination technique, we were able to restrict the numbers of targeted DA neurons down to a single-cell level. To explore the function of these individual dopaminergic neurons, we inactivated them with the tetanus toxin light chain, a genetically encoded inhibitor of neurotransmitter release, or activated them with dTrpA1, a temperature-sensitive cation channel. We found two sets of dopaminergic neurons that modulate aggression, one from the T1 cluster and another from the PPM3 cluster. Both activation and inactivation of these neurons resulted in an increase in aggression. We demonstrate that the presynaptic terminals of the identified T1 and PPM3 dopaminergic neurons project to different parts of the central complex, overlapping with the receptor fields of DD2R and DopR DA receptor subtypes, respectively. These data suggest that the two types of dopaminergic neurons may influence aggression through interactions in the central complex region of the brain involving two different DA receptor subtypes.


Subject(s)
Aggression/drug effects , Brain/pathology , Dopaminergic Neurons/physiology , Drosophila/physiology , Animals , Brain Mapping/methods , Dopamine/metabolism , Drosophila/genetics , Enhancer Elements, Genetic , Female , Gene Library , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Male , Neurons/metabolism , Neurotransmitter Agents/metabolism , Temperature
7.
PLoS One ; 5(5): e10806, 2010 May 24.
Article in English | MEDLINE | ID: mdl-20520823

ABSTRACT

Dopamine (DA) and serotonin (5HT) are reported to serve important roles in aggression in a wide variety of animals. Previous investigations of 5HT function in adult Drosophila behavior have relied on pharmacological manipulations, or on combinations of genetic tools that simultaneously target both DA and 5HT neurons. Here, we generated a transgenic line that allows selective, direct manipulation of serotonergic neurons and asked whether DA and 5HT have separable effects on aggression. Quantitative morphological examination demonstrated that our newly generated tryptophan hydroxylase (TRH)-Gal4 driver line was highly selective for 5HT-containing neurons. This line was used in conjunction with already available Gal4 driver lines that target DA or both DA and 5HT neurons to acutely alter the function of aminergic systems. First, we showed that acute impairment of DA and 5HT neurotransmission using expression of a temperature sensitive form of dynamin completely abolished mid- and high-level aggression. These flies did not escalate fights beyond brief low-intensity interactions and therefore did not yield dominance relationships. We showed next that manipulation of either 5HT or DA neurotransmission failed to duplicate this phenotype. Selective disruption of 5HT neurotransmission yielded flies that fought, but with reduced ability to escalate fights, leading to fewer dominance relationships. Acute activation of 5HT neurons using temperature sensitive dTrpA1 channel expression, in contrast, resulted in flies that escalated fights faster and that fought at higher intensities. Finally, acute disruption of DA neurotransmission produced hyperactive flies that moved faster than controls, and rarely engaged in any social interactions. By separately manipulating 5HT- and DA- neuron systems, we collected evidence demonstrating a direct role for 5HT in the escalation of aggression in Drosophila.


Subject(s)
Aggression , Aging/psychology , Drosophila melanogaster/physiology , Serotonin/metabolism , Synaptic Transmission , Animals , Courtship , Dopamine/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Green Fluorescent Proteins/metabolism , Male , Mutation/genetics , Neurons/metabolism , Phenotype , Tryptophan Hydroxylase/metabolism
8.
Physiol Behav ; 90(2-3): 438-49, 2007 Feb 28.
Article in English | MEDLINE | ID: mdl-17118411

ABSTRACT

Previous studies showed that bilateral lesions of the male ferret's preoptic area/anterior hypothalamus (POA/AH), centered in the sexually dimorphic nuclei present in this region, caused subjects to seek out a same-sex male, as opposed to a female conspecific. Male subjects with POA/AH lesions (which were also castrated and given estradiol) displayed female-typical receptive behavior in response to neck gripping by a stimulus male, implying that subjects' approaches to a same-sex conspecific were sexually motivated. We asked whether the effect of POA/AH lesions on males' partner preference reflects a shift in the central processing of body odorant cues so that males come to display a female-typical preference to approach male body odorants. Sexually experienced male ferrets in which electrolytic lesions of the POA/AH caused bilateral damage to the sexually dimorphic male nucleus (MN) resembled sham-operated females by preferring to approach body odors emitted from anesthetized male as opposed to female stimulus ferrets confined in the goal boxes of a Y-maze. This lesion-induced shift in odor preference was correlated with a significant increase in the ability of soiled male bedding to induce a Fos response in the medial POA of males with bilateral damage to the MN-POA/AH. No such partner preference or neural Fos responses were seen in sham-operated males or in other groups of males with POA/AH lesions that either caused unilateral damage or no damage to the MN-POA/AH. Male-typical hypothalamic processing of conspecifics' body odorants may determine males' normal preference to seek out odors emitted by female conspecifics, leading to mating and successful reproduction.


Subject(s)
Ferrets/physiology , Hypothalamus, Anterior/physiology , Mating Preference, Animal/physiology , Sex Characteristics , Smell/physiology , Analysis of Variance , Animals , Female , Hypothalamus, Anterior/metabolism , Male , Pheromones/physiology , Preoptic Area/metabolism , Preoptic Area/physiology , Proto-Oncogene Proteins c-fos/metabolism , Sex Differentiation/physiology
9.
Neuropsychopharmacology ; 30(2): 278-85, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15257307

ABSTRACT

Although mu-opioid receptors have been extensively investigated for their role in drug reinforcement, little is known about the contribution of these receptors to the acute and sensitized locomotor response to cocaine. In this study mu-opioid receptor involvement in acute cocaine-induced locomotor activity and in the development of cocaine-induced behavioral sensitization was evaluated using mu-opioid receptor knockout mice and chronic naltrexone (NTX) pretreatment as models. In addition, co-administration of the specific mu-opioid receptor antagonist CTOP with repeated saline or cocaine injections was used to establish the involvement of mu-opioid receptors in sensitization to the locomotor stimulant effects of cocaine. The acute locomotor response to cocaine (3, 10, 20, or 30 mg/kg i.p.) of mu-opioid receptor knockout or chronic NTX pretreated mice was not different from the cocaine response of their respective controls. With respect to cocaine-induced behavioral sensitization, induced by daily injections of 20 mg/kg cocaine for 11 subsequent days, mu-opioid receptor knockout mice developed behavioral sensitization to the locomotor stimulant effects of cocaine (challenge 10 mg/kg i.p.) comparable to wild-type littermates and the mu-opioid receptor antagonist CTOP did not affect cocaine-induced sensitization either. However, mice that were pretreated with NTX exhibited augmented cocaine-induced behavioral sensitization relative to placebo pretreated controls, which may be ascribed to increased delta-opioid receptor levels as has been described for chronic NTX pretreated mice. The present findings suggest that mu-opioid receptors are not required for the acute locomotor response to cocaine nor are they essential for the development of cocaine-induced behavioral sensitization.


Subject(s)
Behavior, Animal/drug effects , Cocaine/pharmacology , Motor Activity/drug effects , Receptors, Opioid, mu/drug effects , Somatostatin/analogs & derivatives , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, delta/drug effects , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, kappa/metabolism , Somatostatin/pharmacology , Stimulation, Chemical
10.
Life Sci ; 72(13): 1437-44, 2003 Feb 14.
Article in English | MEDLINE | ID: mdl-12535712

ABSTRACT

We experimented on inbred C57BL/6J strain mice who experienced social stress caused by defeat in inter-male confrontations for 20 days. From the fifth fight on, some mice were injected with ipsapirone (3 mg/kg), and some with buspirone (1 mg/kg) on a daily basis, for 14 days. Post-treatment behavior was examined in the plus-maze, partition, and Porsolt forced swim test (Porsolt's test). Each of these drugs had anxiolytic effects in the plus-maze, suggesting that they reduce state anxiety. Neither had any effect in the partition test, which provides further support to the hypothesis that normally the C57BL/6J strain mice have a high level of trait anxiety and for that reason they did not respond to the drugs. Chronic treatment with neither drug had any effect in the Porsolt's test. It is proposed that ipsapirone and buspirone fail to alleviate the depressive-like behaviors in the C57BL/6J mice because of a high level of trait anxiety, which might be inherent to this mouse strain.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Buspirone/therapeutic use , Pyrimidines/therapeutic use , Social Environment , Stress, Psychological/drug therapy , Stress, Psychological/psychology , Animals , Behavior, Animal/drug effects , Depression/drug therapy , Depression/psychology , Interpersonal Relations , Male , Mice , Mice, Inbred C57BL , Social Dominance
11.
Neurosci Lett ; 321(1-2): 25-8, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11872248

ABSTRACT

Serotonin transporter (SERT) and monoamine oxidase A (MAOA) mRNA levels in the raphe nuclei area of the midbrain were measured by the multiplex reverse transcription-polymerase chain reaction method in male mice with repeated experience of social victories (winners) and defeats (losers) in ten daily agonistic confrontations. Experiments revealed enhanced SERT and MAOA mRNA levels in the losers compared with the winners and controls. It has been supposed that SERT and MAOA genes are involved in enhancement of serotonin inactivation in response to the increase of serotonergic activity shown earlier in the losers. A positive correlation between MAOA and SERT mRNA levels in the raphe nuclei area of the midbrain was shown.


Subject(s)
Agonistic Behavior/physiology , Carrier Proteins/genetics , Membrane Glycoproteins/genetics , Membrane Transport Proteins , Monoamine Oxidase/genetics , Nerve Tissue Proteins , RNA, Messenger/metabolism , Raphe Nuclei/enzymology , Serotonin/metabolism , Up-Regulation/physiology , Acute Disease , Animals , Behavior, Animal/physiology , Chronic Disease , Depression/enzymology , Depression/physiopathology , Male , Mice , Mice, Inbred CBA , Neurons/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Serotonin Plasma Membrane Transport Proteins , Stress, Physiological/enzymology , Stress, Physiological/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...