Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Front Mol Neurosci ; 16: 1225847, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37664240

RESUMO

A challenge for central nervous system (CNS) tissue analysis in neuroscience research has been the difficulty to codetect and colocalize gene and protein expression in the same tissue. Given the importance of identifying gene expression relative to proteins of interest, for example, cell-type specific markers, we aimed to develop a protocol to optimize their codetection. RNAscope fluorescent in situ hybridization (FISH) combined with immunohistochemistry (IHC) in fixed (CNS) tissue sections allows for reliable quantification of gene transcripts of interest within IHC-labeled cells. This paper describes a new method for simultaneous visualization of FISH and IHC in thicker (14-µm), fixed tissue samples, using spinal cord sections. This method's effectiveness is shown by the cell-type-specific quantification of two genes, namely the proinflammatory cytokine interleukin-1beta (IL-1b) and the inflammasome NLR family pyrin domain containing 3 (NLRP3). These genes are challenging to measure accurately using immunohistochemistry (IHC) due to the nonspecificity of available antibodies and the hard-to-distinguish, dot-like visualizations of the labeled proteins within the tissue. These measurements were carried out in spinal cord sections after unilateral chronic constriction injury of the sciatic nerve to induce neuroinflammation in the spinal cord. RNAscope is used to label transcripts of genes of interest and IHC is used to label cell-type specific antigens (IBA1 for microglia, NeuN for neurons). This combination allowed for labeled RNA transcripts to be quantified within cell-type specific boundaries using confocal microscopy and standard image analysis methods. This method makes it easy to answer empirical questions that are intractable with standard IHC or in situ hybridization alone. The method, which has been optimized for spinal cord tissue and to minimize tissue preparation time and costs, is described in detail from tissue collection to image analysis. Further, the relative expression changes in inflammatory genes NLRP3 and IL-1b in spinal cord microglia vs. neurons of somatotopically relevant laminae are described for the first time.

2.
Eur J Neurosci ; 58(7): 3630-3649, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37715507

RESUMO

Foraging is a universal behaviour that has co-evolved with predation pressure. We investigated the role of the bed nucleus of the stria terminalis (BNST) GABA neurons in robotic and live predator threat processing and their consequences in post-threat encounter foraging. Both robotic and live predator interactions increased BNST GABA neuron activity. Mice were trained to procure food in a laboratory-based foraging apparatus in which food pellets were placed at incrementally greater distances from a nest zone. After mice learned to forage, they were exposed to a robotic or live predator threat, while BNST GABA neurons were chemogenetically inhibited. Post-robotic threat encounter, mice spent more time in the nest zone, but other foraging parameters were unchanged compared with pre-encounter behaviour. Inhibition of BNST GABA neurons had no effect on foraging behaviour post-robotic threat encounter. Following live predator exposure, control mice spent significantly more time in the nest zone, increased their latency to successfully forage, and significantly altered their overall foraging performance. Inhibition of BNST GABA neurons during live predator exposure prevented changes in foraging behaviour from developing after a live predator threat. BNST GABA neuron inhibition did not alter foraging behaviour during robotic or live predator threats. We conclude that these results demonstrate that while both robotic and live predator encounters effectively intrude on foraging behaviour, the perceived risk and behavioural consequences of the threat are distinguishable. Additionally, BNST GABA neurons may play a role in the integration of prior innate predator threat experience that results in hypervigilance during post-encounter foraging behaviour.


Assuntos
Núcleos Septais , Camundongos , Animais , Neurônios GABAérgicos , Ansiedade
3.
Neuropsychopharmacology ; 48(13): 1889-1900, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37407648

RESUMO

A two-neuron model of ventral tegmental area (VTA) opioid function classically involves VTA GABA neuron regulation of VTA dopamine neurons via a mu-opioid receptor dependent inhibitory circuit. However, this model predates the discovery of a third major type of neuron in the VTA: glutamatergic neurons. We found that about one-quarter of VTA neurons expressing the mu-opioid receptor are glutamate neurons without molecular markers of GABA co-release. Glutamate-Mu opioid receptor neurons are largely distributed in the anterior VTA. The majority of remaining VTA mu-opioid receptor neurons are GABAergic neurons that are mostly within the posterior VTA and do not express molecular markers of glutamate co-release. Optogenetic stimulation of VTA glutamate neurons resulted in excitatory currents recorded from VTA dopamine neurons that were reduced by presynaptic activation of the mu-opioid receptor ex vivo, establishing a local mu-opioid receptor dependent excitatory circuit from VTA glutamate neurons to VTA dopamine neurons. This VTA glutamate to VTA dopamine pathway regulated dopamine release to the nucleus accumbens through mu-opioid receptor activity in vivo. Behaviorally, VTA glutamate calcium-related neuronal activity increased following oral oxycodone consumption during self-administration and response-contingent oxycodone-associated cues during abstinent reinstatement of drug-seeking behavior. Further, chemogenetic inhibition of VTA glutamate neurons reduced abstinent oral oxycodone-seeking behavior in male but not female mice. These results establish 1) a three-neuron model of VTA opioid function involving a mu-opioid receptor gated VTA glutamate neuron pathway to VTA dopamine neurons that controls dopamine release within the nucleus accumbens, and 2) that VTA glutamate neurons participate in opioid-seeking behavior.


Assuntos
Analgésicos Opioides , Área Tegmentar Ventral , Camundongos , Masculino , Animais , Área Tegmentar Ventral/metabolismo , Analgésicos Opioides/farmacologia , Analgésicos Opioides/metabolismo , Neurônios Dopaminérgicos/metabolismo , Ácido Glutâmico/metabolismo , Receptores Opioides mu/metabolismo , Oxicodona/metabolismo , Dopamina/metabolismo , Ácido gama-Aminobutírico/metabolismo
4.
bioRxiv ; 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-37066408

RESUMO

A unique population of ventral tegmental area (VTA) neurons co-transmits glutamate and GABA as well as functionally signals rewarding and aversive outcomes. However, the circuit inputs to VTA VGluT2+VGaT+ neurons are unknown, limiting our understanding of the functional capabilities of these neurons. To identify the inputs to VTA VGluT2+VGaT+ neurons, we coupled monosynaptic rabies tracing with intersectional genetic targeting of VTA VGluT2+VGaT+ neurons in mice. We found that VTA VGluT2+VGaT+ neurons received diverse brain-wide inputs. The largest numbers of monosynaptic inputs to VTA VGluT2+VGaT+ neurons were from superior colliculus, lateral hypothalamus, midbrain reticular nucleus, and periaqueductal gray, whereas the densest inputs relative to brain region volume were from dorsal raphe nucleus, lateral habenula, and ventral tegmental area. Based on these and prior data, we hypothesized that lateral hypothalamus and superior colliculus inputs were glutamatergic neurons. Optical activation of glutamatergic lateral hypothalamus neurons robustly activated VTA VGluT2+VGaT+ neurons regardless of stimulation frequency and resulted in flee-like ambulatory behavior. In contrast, optical activation of glutamatergic superior colliculus neurons activated VTA VGluT2+VGaT+ neurons for a brief period of time at high stimulation frequency and resulted in head rotation and arrested ambulatory behavior (freezing). For both pathways, behaviors induced by stimulation were uncorrelated with VTA VGluT2+VGaT+ neuron activity. However, stimulation of glutamatergic lateral hypothalamus neurons, but not glutamatergic superior colliculus neurons, was associated with VTA VGluT2+VGaT+ footshock-induced activity. We interpret these results such that inputs to VTA VGluT2+VGaT+ neurons may integrate diverse signals related to the detection and processing of motivationally-salient outcomes. Further, VTA VGluT2+VGaT+ neurons may signal threat-related outcomes, possibly via input from lateral hypothalamus glutamate neurons, but not threat-induced behavioral kinematics.

5.
bioRxiv ; 2023 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-36865159

RESUMO

Foraging is a universal behavior that has co-evolved with predation pressure. We investigated the role of bed nucleus of the stria terminalis (BNST) GABA neurons in robotic and live predator threat processing and their consequences in post-threat encounter foraging. Mice were trained to procure food in a laboratory-based foraging apparatus in which food pellets were placed at discrete and incrementally greater distances from a nest zone. After mice learned to forage, they were exposed to either a robotic or live predator threat, while BNST GABA neurons were chemogenetically inhibited. Post-robotic threat encounter, mice spent more time in the nest zone, but other foraging parameters were unchanged compared to pre-encounter behavior. Inhibition of BNST GABA neurons had no effect on foraging behavior post-robotic threat encounter. Following live predator exposure, control mice spent significantly more time in the nest zone, increased their latency to successfully forage, and their overall foraging performance was significantly a ltered. I nhibition o f BNST GABA neurons during live predator exposure prevented changes in foraging behavior from developing after live predator threat. BNST GABA neuron inhibition did not alter foraging behavior during robotic or live predator threat. We conclude that while both robotic and live predator encounter effectively intrude on foraging behavior, the perceived risk and behavioral consequence of the threats are distinguishable. Additionally, BNST GABA neurons may play a role in the integration of prior innate predator threat experience that results in hypervigilance during post-encounter foraging behavior.

6.
Neuropsychopharmacology ; 48(3): 498-507, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36076018

RESUMO

Stress-linked disorders are more prevalent in women than in men and differ in their clinical presentation. Thus, investigating sex differences in factors that promote susceptibility or resilience to stress outcomes, and the circuit elements that mediate their effects, is important. In male rats, instrumental control over stressors engages a corticostriatal system involving the prelimbic cortex (PL) and dorsomedial striatum (DMS) that prevent many of the sequelae of stress exposure. Interestingly, control does not buffer against stress outcomes in females, and here, we provide evidence that the instrumental controlling response in females is supported instead by the dorsolateral striatum (DLS). Additionally, we used in vivo microdialysis, fluorescent in situ hybridization, and receptor subtype pharmacology to examine the contribution of prefrontal dopamine (DA) to the differential impact of behavioral control. Although both sexes preferentially expressed D1 receptor mRNA in PL GABAergic neurons, there were robust sex differences in the dynamic properties of prefrontal DA during controllable stress. Behavioral control potently attenuated stress-induced DA efflux in males, but not females, who showed a sustained DA increase throughout the entire stress session. Importantly, PL D1 receptor blockade (SCH 23390) shifted the proportion of striatal activity from the DLS to the DMS in females and produced the protective effects of behavioral control. These findings suggest a sex-selective mechanism in which elevated DA in the PL biases instrumental responding towards prefrontal-independent striatal circuitry, thereby eliminating the protective impact of coping with stress.


Assuntos
Controle Comportamental , Dopamina , Ratos , Feminino , Masculino , Animais , Dopamina/farmacologia , Hibridização in Situ Fluorescente , Córtex Pré-Frontal , Neostriado/metabolismo , Corpo Estriado/metabolismo , Receptores de Dopamina D1/metabolismo
7.
Mol Psychiatry ; 2022 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-36437312

RESUMO

Exposure to trauma is a risk factor for the development of a number of mood disorders, and may enhance vulnerability to future adverse life events. Recent data demonstrate that ventral tegmental area (VTA) neurons expressing the vesicular glutamate transporter 2 (VGluT2) signal and causally contribute to behaviors that involve aversive or threatening stimuli. However, it is unknown whether VTA VGluT2 neurons regulate transsituational outcomes of stress and whether these neurons are sensitive to stressor controllability. This work adapted an operant mouse paradigm to examine the impact of stressor controllability on VTA VGluT2 neuron function as well as the role of VTA VGluT2 neurons in mediating transsituational stressor outcomes. Uncontrollable (inescapable) stress, but not physically identical controllable (escapable) stress, produced social avoidance and exaggerated fear in male mice. Uncontrollable stress in females led to exploratory avoidance of a novel brightly lit environment. Both controllable and uncontrollable stressors increased VTA VGluT2 neuronal activity, and chemogenetic silencing of VTA VGluT2 neurons prevented the behavioral sequelae of uncontrollable stress in male and female mice. Further, we show that stress activates multiple genetically-distinct subtypes of VTA VGluT2 neurons, especially those that are VGluT2+VGaT+, as well as lateral habenula neurons receiving synaptic input from VTA VGluT2 neurons. Our results provide causal evidence that mice can be used for identifying stressor controllability circuitry and that VTA VGluT2 neurons contribute to transsituational stressor outcomes, such as social avoidance, exaggerated fear, or anxiety-like behavior that are observed within trauma-related disorders.

8.
eNeuro ; 9(2)2022.
Artigo em Inglês | MEDLINE | ID: mdl-35396257

RESUMO

Tools for refined cell-specific targeting have significantly contributed to understanding the characteristics and dynamics of distinct cellular populations by brain region. While advanced cell-labeling methods have accelerated the field of neuroscience, specifically in brain mapping, there remains a need to quantify and analyze the data. Here, by modifying a toolkit that localizes electrodes to brain regions (SHARP-Track; Slice Histology Alignment, Registration, and Probe-Track analysis), we introduce a post-imaging analysis tool to map histological images to established mouse brain atlases called SHARCQ (Slice Histology Alignment, Registration, and Cell Quantification). The program requires MATLAB, histological images, and either a manual or automatic cell count of the unprocessed images. SHARCQ simplifies the post-imaging analysis pipeline with a step-by-step GUI. We demonstrate that SHARCQ can be applied for a variety of mouse brain images, regardless of histology technique. In addition, SHARCQ rectifies discrepancies in mouse brain region borders between atlases by allowing the user to select between the Allen Brain Atlas or the digitized and modified Franklin-Paxinos Atlas for quantifying cell counts by region. SHARCQ produces quantitative and qualitative data, including counts of brain-wide region populations and a 3D model of registered cells within the atlas space. In summary, SHARCQ was designed as a neuroscience post-imaging analysis tool for cell-to-brain registration and quantification with a simple, accessible interface. All code is open-source and available for download (https://github.com/wildrootlab/SHARCQ).


Assuntos
Encéfalo , Processamento de Imagem Assistida por Computador , Animais , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico/métodos , Técnicas Histológicas , Processamento de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética , Camundongos , Fluxo de Trabalho
10.
J Neurosci ; 41(25): 5471-5486, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34001626

RESUMO

Ventral tegmental area (VTA) glutamate neurons signal and participate in reward and aversion-based behaviors. However, the neurochemical mechanisms that underlie how these neurons contribute to motivated behaviors is unknown. We used a combination of optical sensors to identify how distinct neurochemical inputs to VTA glutamate neurons participate in motivated behavior within female and male transgenic mice. Activity of glutamate inputs to VTA glutamate neurons increased for both reward-predicting and aversion-predicting cues and aversive outcomes, but subpopulations of glutamate inputs were increased or decreased by reward. For both reward and aversion-based cues and outcomes, activity of GABA inputs to VTA glutamate neurons mostly decreased. GCaMP recordings showed overall population increases in VTA glutamate neuron intracellular calcium during reward and aversion-based cues and outcomes. Electrophysiological recordings of VTA VGluT2 neurons showed that glutamate receptor activation increases firing while loss of excitation via glutamate receptor blockade decreases firing. GABA-A receptor activation decreased VTA glutamate neuron firing but GABA-A receptor blockade did not significantly change VTA glutamate neuron firing. Electrophysiological recordings in coordination with our sensor data suggest that glutamate inputs strongly regulate VTA glutamate neuron participation in diverse motivated behaviors.SIGNIFICANCE STATEMENT Glutamate and GABA are the primary excitatory and inhibitory neurotransmitters of the nervous system. However, identifying how these neurotransmitters regulate motivated behavior has remained challenging because of a lack of tools (1) capable of measuring neurotransmission at the temporal scale of motivated behaviors and (2) capable of capturing chemical signaling onto genetically-distinct neuronal populations. We have overcome these obstacles by implementing genetically-encoded fluorescent indicators to monitor both glutamate and GABA input dynamics exclusively to ventral tegmental area (VTA) glutamate neurons during reward and aversion-based behaviors. We identify that glutamate and GABA inputs to VTA glutamate neurons differentially and dynamically signal reward and aversion-based cues and outcomes. This research provides foundational evidence that links distinct neurotransmitters to motivated behaviors regulated by VTA glutamate neurons.


Assuntos
Comportamento Animal/fisiologia , Ácido Glutâmico/metabolismo , Neurônios/metabolismo , Recompensa , Área Tegmentar Ventral/metabolismo , Animais , Sinais (Psicologia) , Feminino , Masculino , Camundongos , Motivação/fisiologia , Transdução de Sinais/fisiologia
11.
Cell Rep ; 32(9): 108094, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32877676

RESUMO

Ventral tegmental area (VTA) neurons play roles in reward and aversion. We recently discovered that the VTA has neurons that co-transmit glutamate and GABA (glutamate-GABA co-transmitting neurons), transmit glutamate without GABA (glutamate-transmitting neurons), or transmit GABA without glutamate (GABA-transmitting neurons). However, the functions of these VTA cell types in motivated behavior are unclear. To identify the functions of these VTA cell types, we combine recombinase mouse lines with INTRSECT2.0 vectors to selectively target these neurons. We find that VTA cell types have unique signaling patterns for reward, aversion, and learned cues. Whereas VTA glutamate-transmitting neurons signal cues predicting reward, VTA GABA-transmitting neurons signal cues predicting the absence of reward, and glutamate-GABA co-transmitting neurons signal rewarding and aversive outcomes without signaling learned cues related to those outcomes. Thus, we demonstrate that genetically defined subclasses of VTA glutamate and GABA neurons signal different aspects of motivated behavior.


Assuntos
Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Motivação/genética , Área Tegmentar Ventral/fisiopatologia , Animais , Humanos , Masculino , Camundongos , Transdução de Sinais
12.
Addict Biol ; 25(6): e12828, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-31489746

RESUMO

A significant portion of prescription opioid users self-administer orally rather than intravenously. Animal models of opioid addiction have demonstrated that intravenous cues are sufficient to cause drug seeking. However, intravenous models may not characterize oral users, and the preference to self-administer orally appears to be partially influenced by the user's sex. Our objectives were to determine whether oral opioid-associated cues are sufficient for relapse and whether sex differences exist in relapse susceptibility. Mice orally self-administered escalating doses of oxycodone under postprandial (prefed) or non-postprandial (no prefeeding) conditions. Both sexes demonstrated cue-induced reinstatement following abstinence. In separate mice, we found that oral oxycodone cues were sufficient to reinstate extinguished oral oxycodone-seeking behavior following abstinence without prior postprandial or water self-administration training. During self-administration, we incidentally found that female mice earned significantly more mg/kg oxycodone than male mice. Follow-up studies indicated sex differences in psychomotor stimulation and plasma oxycodone/oxymorphone following oral oxycodone administration. In addition, gonadal studies were performed in which we found divergent responses where ovariectomy-enhanced and orchiectomy-suppressed oral self-administration. While the suppressive effects of orchiectomy were identified across doses and postprandial conditions, the enhancing effects of ovariectomy were selective to non-postprandial conditions. These studies establish that (a) oral drug cues are sufficient to cause reinstatement that is independent of prandial conditions and water-seeking behavior, (b) earned oral oxycodone is larger in female mice compared with male mice potentially through differences in psychomotor stimulation and drug metabolism, and (c) gonadectomy produces divergent effects on oral oxycodone self-administration between sexes.


Assuntos
Comportamento de Procura de Droga , Transtornos Relacionados ao Uso de Opioides/psicologia , Oxicodona/administração & dosagem , Abuso Oral de Substâncias/psicologia , Administração Oral , Animais , Comportamento Animal/efeitos dos fármacos , Sinais (Psicologia) , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxicodona/sangue , Autoadministração , Fatores Sexuais
14.
iScience ; 2: 51-62, 2018 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-29888759

RESUMO

Ventral tegmental area (VTA) neurons play roles in reward and aversion. The VTA has, in addition to dopamine neurons, glutamatergic neurons expressing VGluT2. Here, by determining the firing patterns of VTA-VGluT2 neurons expressing channelrhodopsin 2, we identified a major subpopulation of VTA-VGluT2 neurons whose firing rates decreased or were unchanged during sucrose consumption and increased during facial airpuff presentation. We identified a small subpopulation of VTA-VGluT2 neurons whose firing rates increased in response to both rewarding and aversive stimuli. We also found that the changes in firing rate of some VTA-VGluT2 neurons were greater following reward delivery compared with reward omission, whereas others did not differ. We conclude that VTA-VGluT2 neurons are responsive to aversive stimuli, but subpopulations of VTA-VGluT2 neurons are differentially affected by sucrose reward. Reward-responsive subpopulations of VTA-VGluT2 neurons are also divided into those affected by reward expectation alone or the real-time delivery of reward.

15.
Cell Rep ; 23(12): 3465-3479, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29924991

RESUMO

For decades, it has been thought that glutamate and GABA are released by distinct neurons. However, some mouse neurons innervating the lateral habenula (LHb) co-release glutamate and GABA. Here, we mapped the distribution of neurons throughout the rat brain that co-express vesicular transporters for the accumulation of glutamate (VGluT2) or GABA (VGaT) and for GABA synthesis (GAD). We found concentrated groups of neurons that co-express VGluT2, VGaT, and GAD mRNAs within subdivisions of the ventral tegmental area (VTA), entopeduncular (EPN), and supramammillary (SUM) nuclei. Single axon terminals established by VTA, EPN, or SUM neurons form a common synaptic architecture involving asymmetric (putative excitatory) and symmetric (putative inhibitory) synapses. Within the LHb, which receives co-transmitted glutamate and GABA from VTA and EPN, VGluT2 and VGaT are distributed on separate synaptic vesicles. We conclude that single axon terminals from VGluT2 and VGaT co-expressing neurons co-transmit glutamate and GABA from distinct synaptic vesicles at independent synapses.


Assuntos
Encéfalo/metabolismo , Neurônios GABAérgicos/metabolismo , Glutamatos/metabolismo , Sinapses/metabolismo , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Núcleo Entopeduncular/metabolismo , Glutamato Descarboxilase/metabolismo , Masculino , Modelos Biológicos , Ratos Sprague-Dawley , Sinapses/ultraestrutura , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura , Distribuição Tecidual , Área Tegmentar Ventral/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/metabolismo
16.
Cell Rep ; 21(7): 1757-1769, 2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29141211

RESUMO

The lateral habenula (LHb) is a brain structure that participates in cognitive and emotional processing and has been implicated in several mental disorders. Although one of the largest inputs to the LHb originates in the lateral preoptic area (LPO), little is known about how the LPO participates in the regulation of LHb function. Here, we provide evidence that the LPO exerts bivalent control over the LHb through the convergent transmission of LPO glutamate and Î³-aminobutyric acid (GABA) onto single LHb neurons. In vivo, both LPO-glutamatergic and LPO-GABAergic inputs to the LHb are activated by aversive stimuli, and their predictive cues yet produce opposing behaviors when stimulated independently. These results support a model wherein the balanced response of converging LPO-glutamate and LPO-GABA are necessary for a normal response to noxious stimuli, and an imbalance in LPO→LHb glutamate or GABA results in the type of aberrant processing that may underlie mental disorders.


Assuntos
Ácido Glutâmico/metabolismo , Habenula/metabolismo , Área Pré-Óptica/metabolismo , Transmissão Sináptica , Ácido gama-Aminobutírico/metabolismo , Animais , Habenula/citologia , Habenula/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/fisiologia , Área Pré-Óptica/citologia , Área Pré-Óptica/fisiologia , Ratos , Ratos Sprague-Dawley
17.
Pharmacol Biochem Behav ; 162: 3-21, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28647565

RESUMO

The cytology and connections of the lateral habenula (LHb) are reviewed. The habenula is first introduced, after which the cytology of the LHb is discussed mainly with reference to cell types, general topography and descriptions of subnuclei. An overview of LHb afferent connections is given followed by some details about the projections to LHb from a number of structures. An overview of lateral habenula efferent connections is given followed by some details about the projections from LHb to a number of structures. In considering the afferent and efferent connections of the LHb some attention is given to the relative validity of regarding it as a bi-partite structure featuring 'limbic' and 'pallidal' parts. The paper ends with some concluding remarks about the relative place of the LHb in adaptive behaving.


Assuntos
Adaptação Fisiológica/fisiologia , Habenula/citologia , Habenula/fisiologia , Rede Nervosa/citologia , Rede Nervosa/fisiologia , Animais , Humanos
18.
Sci Rep ; 6: 30615, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27477243

RESUMO

The rodent ventral tegmental area (VTA) and substantia nigra pars compacta (SNC) contain dopamine neurons intermixed with glutamate neurons (expressing vesicular glutamate transporter 2; VGluT2), which play roles in reward and aversion. However, identifying the neuronal compositions of the VTA and SNC in higher mammals has remained challenging. Here, we revealed VGluT2 neurons within the VTA and SNC of nonhuman primates and humans by simultaneous detection of VGluT2 mRNA and tyrosine hydroxylase (TH; for identification of dopamine neurons). We found that several VTA subdivisions share similar cellular compositions in nonhuman primates and humans; their rostral linear nuclei have a high prevalence of VGluT2 neurons lacking TH; their paranigral and parabrachial pigmented nuclei have mostly TH neurons, and their parabrachial pigmented nuclei have dual VGluT2-TH neurons. Within nonhuman primates and humans SNC, the vast majority of neurons are TH neurons but VGluT2 neurons were detected in the pars lateralis subdivision. The demonstration that midbrain dopamine neurons are intermixed with glutamate or glutamate-dopamine neurons from rodents to humans offers new opportunities for translational studies towards analyzing the roles that each of these neurons play in human behavior and in midbrain-associated illnesses such as addiction, depression, schizophrenia, and Parkinson's disease.


Assuntos
Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Mesencéfalo/citologia , Neurônios/metabolismo , Animais , Callithrix , Humanos , Masculino , Mesencéfalo/metabolismo , Parte Compacta da Substância Negra/citologia , Parte Compacta da Substância Negra/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
19.
J Chem Neuroanat ; 73: 33-42, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26763116

RESUMO

The ventral tegmental area (VTA) is an evolutionarily conserved structure that has roles in reward-seeking, safety-seeking, learning, motivation, and neuropsychiatric disorders such as addiction and depression. The involvement of the VTA in these various behaviors and disorders is paralleled by its diverse signaling mechanisms. Here we review recent advances in our understanding of neuronal diversity in the VTA with a focus on cell phenotypes that participate in 'multiplexed' neurotransmission involving distinct signaling mechanisms. First, we describe the cellular diversity within the VTA, including neurons capable of transmitting dopamine, glutamate or GABA as well as neurons capable of multiplexing combinations of these neurotransmitters. Next, we describe the complex synaptic architecture used by VTA neurons in order to accommodate the transmission of multiple transmitters. We specifically cover recent findings showing that VTA multiplexed neurotransmission may be mediated by either the segregation of dopamine and glutamate into distinct microdomains within a single axon or by the integration of glutamate and GABA into a single axon terminal. In addition, we discuss our current understanding of the functional role that these multiplexed signaling pathways have in the lateral habenula and the nucleus accumbens. Finally, we consider the putative roles of VTA multiplexed neurotransmission in synaptic plasticity and discuss how changes in VTA multiplexed neurons may relate to various psychopathologies including drug addiction and depression.


Assuntos
Neurônios/fisiologia , Transdução de Sinais , Área Tegmentar Ventral/fisiologia , Animais , Comportamento/fisiologia , Depressão/fisiopatologia , Dopamina/fisiologia , Habenula/fisiologia , Humanos , Neurotransmissores/fisiologia , Núcleo Accumbens/fisiologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transmissão Sináptica , Área Tegmentar Ventral/citologia , Ácido gama-Aminobutírico/fisiologia
20.
Prog Neurobiol ; 130: 29-70, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25857550

RESUMO

The ventral pallidum (VP) plays a critical role in the processing and execution of motivated behaviors. Yet this brain region is often overlooked in published discussions of the neurobiology of mental health (e.g., addiction, depression). This contributes to a gap in understanding the neurobiological mechanisms of psychiatric disorders. This review is presented to help bridge the gap by providing a resource for current knowledge of VP anatomy, projection patterns and subregional circuits, and how this organization relates to the function of VP neurons and ultimately behavior. For example, ventromedial (VPvm) and dorsolateral (VPdl) VP subregions receive projections from nucleus accumbens shell and core, respectively. Inhibitory GABAergic neurons of the VPvm project to mediodorsal thalamus, lateral hypothalamus, and ventral tegmental area, and this VP subregion helps discriminate the appropriate conditions to acquire natural rewards or drugs of abuse, consume preferred foods, and perform working memory tasks. GABAergic neurons of the VPdl project to subthalamic nucleus and substantia nigra pars reticulata, and this VP subregion is modulated by, and is necessary for, drug-seeking behavior. Additional circuits arise from nonGABAergic neuronal phenotypes that are likely to excite rather than inhibit their targets. These subregional and neuronal phenotypic circuits place the VP in a unique position to process motivationally relevant stimuli and coherent adaptive behaviors.


Assuntos
Prosencéfalo Basal/fisiologia , Comportamento Animal/fisiologia , Neurônios GABAérgicos/citologia , Motivação/fisiologia , Atividade Motora/fisiologia , Núcleo Accumbens/fisiologia , Animais , Prosencéfalo Basal/anatomia & histologia , Humanos , Núcleo Accumbens/anatomia & histologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA