Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
J Neurosci ; 43(36): 6280-6296, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37591737

RESUMO

The hypothalamic melanocortin system is critically involved in sensing stored energy and communicating this information throughout the brain, including to brain regions controlling motivation and emotion. This system consists of first-order agouti-related peptide (AgRP) and pro-opiomelanocortin (POMC) neurons located in the hypothalamic arcuate nucleus and downstream neurons containing the melanocortin-3 (MC3R) and melanocortin-4 receptor (MC4R). Although extensive work has characterized the function of downstream MC4R neurons, the identity and function of MC3R-containing neurons are poorly understood. Here, we used neuroanatomical and circuit manipulation approaches in mice to identify a novel pathway linking hypothalamic melanocortin neurons to melanocortin-3 receptor neurons located in the paraventricular thalamus (PVT) in male and female mice. MC3R neurons in PVT are innervated by hypothalamic AgRP and POMC neurons and are activated by anorexigenic and aversive stimuli. Consistently, chemogenetic activation of PVT MC3R neurons increases anxiety-related behavior and reduces feeding in hungry mice, whereas inhibition of PVT MC3R neurons reduces anxiety-related behavior. These studies position PVT MC3R neurons as important cellular substrates linking energy status with neural circuitry regulating anxiety-related behavior and represent a promising potential target for diseases at the intersection of metabolism and anxiety-related behavior such as anorexia nervosa.SIGNIFICANCE STATEMENT Animals must constantly adapt their behavior to changing internal and external challenges, and impairments in appropriately responding to these challenges are a hallmark of many neuropsychiatric disorders. Here, we demonstrate that paraventricular thalamic neurons containing the melanocortin-3 receptor respond to energy-state-related information and external challenges to regulate anxiety-related behavior in mice. Thus, these neurons represent a potential target for understanding the neurobiology of disorders at the intersection of metabolism and psychiatry such as anorexia nervosa.


Assuntos
Melanocortinas , Pró-Opiomelanocortina , Animais , Feminino , Masculino , Camundongos , Proteína Relacionada com Agouti , Ansiedade , Homeostase , Receptor Tipo 3 de Melanocortina , Tálamo
2.
J Neurochem ; 168(6): 995-1018, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38664195

RESUMO

Paraventricular thalamus (PVT) plays important roles in the regulation of emotion and motivation through connecting many brain structures including the midbrain and the limbic system. Although acetylcholine (ACh) neurons of the midbrain were reported to send projections to PVT, little is known about how cholinergic signaling regulates PVT neurons. Here, we used both RNAscope and slice patch-clamp recordings to characterize cholinergic receptor expression and ACh modulation of PVT neurons in mice. We found ACh excited a majority of anterior PVT (aPVT) neurons but predominantly inhibited posterior PVT (pPVT) neurons. Compared to pPVT with more inhibitory M2 receptors, aPVT expressed higher levels of all excitatory receptor subtypes including nicotinic α4, α7, and muscarinic M1 and M3. The ACh-induced excitation was mimicked by nicotine and antagonized by selective blockers for α4ß2 and α7 nicotinic ACh receptor (nAChR) subtypes as well as selective antagonists for M1 and M3 muscarinic ACh receptors (mAChR). The ACh-induced inhibition was attenuated by selective M2 and M4 mAChR receptor antagonists. Furthermore, we found ACh increased the frequency of excitatory postsynaptic currents (EPSCs) on a majority of aPVT neurons but decreased EPSC frequency on a larger number of pPVT neurons. In addition, ACh caused an acute increase followed by a lasting reduction in inhibitory postsynaptic currents (IPSCs) on PVT neurons of both subregions. Together, these data suggest that multiple AChR subtypes coordinate a differential modulation of ACh on aPVT and pPVT neurons.


Assuntos
Acetilcolina , Camundongos Endogâmicos C57BL , Neurônios , Animais , Camundongos , Acetilcolina/metabolismo , Acetilcolina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Masculino , Núcleos da Linha Média do Tálamo/efeitos dos fármacos , Núcleos da Linha Média do Tálamo/fisiologia , Receptores Colinérgicos/metabolismo , Feminino , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia
3.
Br J Anaesth ; 132(2): 334-342, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38044237

RESUMO

BACKGROUND: Delayed emergence from general anaesthesia poses a significant perioperative safety hazard. Subanaesthetic doses of ketamine not only deepen anaesthesia but also accelerate recovery from isoflurane anaesthesia; however, the mechanisms underlying this phenomenon remain elusive. Esketamine exhibits a more potent receptor affinity and fewer adverse effects than ketamine and exhibits shorter recovery times after brief periods of anaesthesia. As the paraventricular thalamus (PVT) plays a pivotal role in regulating wakefulness, we studied its role in the emergence process during combined esketamine and isoflurane anaesthesia. METHODS: The righting reflex and cortical electroencephalography were used as measures of consciousness in mice during isoflurane anaesthesia with coadministration of esketamine. The expression of c-Fos was used to determine neuronal activity changes in PVT neurones after esketamine administration. The effect of esketamine combined with isoflurane anaesthesia on PVT glutamatergic (PVTGlu) neuronal activity was monitored by fibre photometry, and chemogenetic technology was used to manipulate PVTGlu neuronal activity. RESULTS: A low dose of esketamine (5 mg kg-1) accelerated emergence from isoflurane general anaesthesia (474 [30] s vs 544 [39] s, P=0.001). Esketamine (5 mg kg-1) increased PVT c-Fos expression (508 [198] vs 258 [87], P=0.009) and enhanced the population activity of PVTGlu neurones (0.03 [1.7]% vs 6.9 [3.4]%, P=0.002) during isoflurane anaesthesia (1.9 [5.7]% vs -5.1 [5.3]%, P=0.016) and emergence (6.1 [6.2]% vs -1.1 [5.0]%, P=0.022). Chemogenetic suppression of PVTGlu neurones abolished the arousal-promoting effects of esketamine (459 [33] s vs 596 [33] s, P<0.001). CONCLUSIONS: Our results suggest that esketamine promotes recovery from isoflurane anaesthesia by activating PVTGlu neurones. This mechanism could explain the rapid arousability exhibited upon treatment with a low dose of esketamine.


Assuntos
Anestésicos Inalatórios , Isoflurano , Ketamina , Tálamo , Animais , Camundongos , Anestesia Geral , Anestésicos Inalatórios/farmacologia , Isoflurano/farmacologia , Ketamina/farmacologia , Tálamo/efeitos dos fármacos
4.
J Neurosci ; 42(16): 3473-3483, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35273082

RESUMO

Decisions to act while pursuing goals in the presence of danger must be made quickly but safely. Premature decisions risk injury or death, whereas postponing decisions risk goal loss. Here we show how mice resolve these competing demands. Using microstructural behavioral analyses, we identified the spatiotemporal dynamics of approach-avoidance decisions under motivational conflict in male mice. Then we used cognitive modeling to show that these dynamics reflect the speeded decision-making mechanisms used by humans and nonhuman primates, with mice trading off decision speed for safety of choice when danger loomed. Using calcium imaging in paraventricular thalamus and optogenetic inhibition of the prelimbic cortex to paraventricular thalamus pathway, we show that this speed-safety trade off occurs because increases in paraventricular thalamus activity increase decision caution, thereby increasing approach-avoid decision times in the presence of danger. Our findings demonstrate that a discrete brain circuit involving the paraventricular thalamus and its prefrontal input adjusts decision caution during motivational conflict, trading off decision speed for decision safety when danger is close. We identify the corticothalamic pathway as central to cognitive control during decision-making under conflict.SIGNIFICANCE STATEMENT Foraging animals balance the need to seek food and energy against the conflicting needs to avoid injury and predation. This competition is fundamental to survival but rarely has a stable, correct solution. Here we show that approach-avoid decisions under motivational conflict involve strategic adjustments in decision caution controlled via a top-down corticothalamic pathway from the prelimbic cortex to the paraventricular thalamus. We identify a novel corticothalamic mechanism for cognitive control that is applicable across a range of motivated behaviors and mark paraventricular thalamus and its prefrontal cortical input as targets to remediate the deficits in decision caution characteristic of unsafe and impulsive choices.


Assuntos
Motivação , Tálamo , Animais , Tomada de Decisões/fisiologia , Comportamento Impulsivo , Masculino , Camundongos , Córtex Pré-Frontal , Recompensa
5.
J Neurosci ; 42(19): 3949-3964, 2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35387870

RESUMO

Oxytocin receptors (OTR) have been found in the paraventricular thalamus (PVT) for the regulation of feeding and maternal behaviors. However, the functional projections of OTR-expressing PVT neurons remain largely unknown. Here, we used chemogenetic and optogenetic tools to test the role of OTR-expressing PVT neurons and their projections in the regulation of food intake in both male and female OTR-Cre mice. We found chemogenetic activation of OTR-expressing PVT neurons promoted food seeking under trials with a progressive ratio schedule of reinforcement. Using Feeding Experimentation Devices for real-time meal measurements, we found chemogenetic activation of OTR-expressing PVT neurons increased meal frequency but not cumulative food intake because of a compensatory decrease in meal sizes. In combination with anterograde neural tracing and slice patch-clamp recordings, we found optogenetic stimulation of PVT OTR terminals excited neurons in the posterior basolateral amygdala (pBLA) and nucleus accumbens core (NAcC) as well as local PVT neurons through monosynaptic glutamatergic transmissions. Photostimulation of OTR-expressing PVT-NAcC projections promoted food-seeking, whereas selective activation of PVT-pBLA projections produced little effect on feeding. In contrast to selective activation of OTR terminals, photostimulation of a broader population of glutamatergic PVT terminals exerted direct excitation followed by indirect lateral inhibition on neurons in both NAcC and anterior basolateral amygdala. Together, these results suggest that OTR-expressing PVT neurons are a distinct population of PVT glutamate neurons that regulate feeding motivation through projections to NAcC.SIGNIFICANCE STATEMENT The paraventricular thalamus plays an important role in the regulation of feeding motivation. However, because of the diversity of paraventricular thalamic neurons, the specific neuron types promoting food motivation remain elusive. In this study, we provide evidence that oxytocin receptor-expressing neurons are a specific group of glutamate neurons that primarily project to the nucleus accumbens core and posterior amygdala. We found that activation of these neurons promotes the motivation for food reward and increases meal frequency through projections to the nucleus accumbens core but not the posterior amygdala. As a result, we postulate that oxytocin receptor-expressing neurons in the paraventricular thalamus and their projections to the nucleus accumbens core mainly regulate feeding motivation but not food consumption.


Assuntos
Núcleo Accumbens , Receptores de Ocitocina , Animais , Feminino , Ácido Glutâmico , Masculino , Camundongos , Motivação , Neurônios/metabolismo , Núcleo Accumbens/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Tálamo/fisiologia
6.
Br J Anaesth ; 130(6): 698-708, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36828739

RESUMO

BACKGROUND: The neuronal mechanisms underlying propofol-induced modulation of consciousness are poorly understood. Neuroimaging studies suggest a potential role for non-specific thalamic nuclei in propofol-induced loss of consciousness. We investigated the contribution of the paraventricular thalamus (PVT), a midline thalamic nucleus that has been implicated in arousal control and general anaesthesia with inhaled anaesthetics, to loss and recovery of consciousness during propofol anaesthesia. METHODS: Polysomnographic recordings and righting reflex test were used to determine the transitions of loss and recovery of righting reflex, used as a measure of consciousness in mice, during propofol anaesthesia in mice under conditions mimicking clinical propofol administration. PVT neuronal activities were monitored using fibre photometry and regulated using optogenetic and chemogenetic methods. RESULTS: Population activities of PVT glutamatergic neurones began to decrease before propofol-induced loss of consciousness and rapidly increased to a peak at the onset of recovery of consciousness. Chemogenetic inhibition of PVT calretinin-expressing (PVTCR) neurones shortened onset (from 176 [35] to 127 [26] s; P=0.001) and prolonged return (from 1568 [611] to 3126 [1616] s; P=0.002) of righting reflex. Conversely, chemogenetic activation of PVTCR neurones exerted opposite effects. Furthermore, optogenetic silencing of PVTCR neurones accelerated transitions to loss of consciousness (from 205 [35] to 158 [44] s; P=0.027) and slowed transitions to recovery of consciousness (from 230 [78] to 370 [99] s; P=0.041). During a steady period of unconsciousness maintained with continuous propofol infusion, brief optical activation of PVTCR neurones restored cortical activity and arousal with a latency of about 5 s. CONCLUSIONS: The paraventricular thalamus contributes to the control of consciousness transitions in propofol anaesthesia in mice. This provides a potential neuroanatomical target for controlling consciousness to reduce anaesthetic dose requirements and side effects.


Assuntos
Propofol , Camundongos , Animais , Propofol/efeitos adversos , Estado de Consciência , Anestésicos Intravenosos/efeitos adversos , Tálamo , Inconsciência/induzido quimicamente , Anestesia Geral/métodos
7.
Ecotoxicol Environ Saf ; 262: 115205, 2023 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-37392660

RESUMO

Bisphenol A (BPA), a ubiquitous endocrine disrupting chemical, is widely used in household plastic products. Large amounts of evidence indicate prenatal and postnatal BPA exposure causes neurodevelopmental disorders such as anxiety and autism. However, the neuronal mechanisms underlying the neurotoxic effects of adulthood BPA exposure remain poorly understood. Here, we provided evidences that adult mice treated with BPA (0.45 mg/kg/day) during 3 weeks exhibited sex-specific anxiety like behaviors. We demonstrated that the BPA-induced anxiety in male mice, but not in female mice, was closely associated with hyperactivity of glutamatergic neurons in the paraventricular thalamus (PVT). Acute chemogenetic activation of PVT glutamatergic neurons caused similar effects on anxiety as observed in male mice exposed to BPA. In contrast, acute chemogenetic inhibition of PVT glutamatergic neurons reduced BPA-induced anxiety in male mice. Concomitantly, the BPA-induced anxiety was related with a down-regulation of alpha-1D adrenergic receptor in the PVT. Taken together, the present study indicated a previously unknown target region in the brain for neurotoxic effects of BPA on anxiety and implicated a possible molecular mechanism of action.

8.
J Physiol ; 600(12): 2877-2895, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35648134

RESUMO

The regulation of food intake and energy balance relies on the dynamic integration of exteroceptive and interoceptive signals monitoring nutritional, metabolic, cognitive, and emotional states. The paraventricular thalamus (PVT) is a central hub that, by integrating sensory, metabolic, and emotional states, may contribute to the regulation of feeding and homeostatic/allostatic processes. However, the underlying PVT circuits still remain elusive. Here, we aimed at unravelling the role of catecholaminergic (CA) inputs to the PVT in scaling feeding and metabolic efficiency. First, using region-specific retrograde disruption of CA projections, we show that PVT CA inputs mainly arise from the hindbrain, notably the locus coeruleus (LC) and the nucleus tractus solitarius. Second, taking advantage of integrative calorimetric measurements of metabolic efficiency, we reveal that CA inputs to the PVT scale adaptive feeding and metabolic responses in environmental, behavioural, physiological, and metabolic stress-like contexts. Third, we show that hindbrainTH →PVT inputs contribute to modulating the activity of PVT as well as lateral and dorsomedial hypothalamic neurons. In conclusion, the present study, by assessing the key role of CA inputs to the PVT in scaling homeostatic/allostatic regulations of feeding patterns, reveals the integrative and converging hindbrainTH →PVT paths that contribute to whole-body metabolic adaptations in stress-like contexts. KEY POINTS: The paraventricular thalamus (PVT) is known to receive projections from the hindbrain. Here, we confirm and further extend current knowledge on the existence of hindbrainTH →PVT catecholaminergic inputs, notably from the locus coeruleus and the nucleus tractus solitarius, with the nucleus tractus solitarius representing the main source. Disruption of hindbrainTH →PVT inputs contributes to the modulation of PVT neuron activity. HindbrainTH →PVT inputs scale feeding strategies in environmental, behavioural, physiological, and metabolic stress-like contexts. HindbrainTH →PVT inputs participate in regulating metabolic efficiency and nutrient partitioning in stress-like contexts. HindbrainTH →PVT inputs, directly and/or indirectly, contribute to modulating the downstream activity of lateral and dorsomedial hypothalamic neurons.


Assuntos
Núcleo Solitário , Tálamo , Comportamento Alimentar/fisiologia , Hipotálamo , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular , Tálamo/fisiologia
9.
Neurobiol Dis ; 174: 105890, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36220611

RESUMO

The dysregulation of neuronal networks contributes to the etiology of psychiatric diseases, including anxiety. However, the neural circuits underlying anxiety symptoms remain unidentified. We observed acute restraint stress activating excitatory neurons in the paraventricular thalamus (PVT). Activation of PVT neurons caused anxious behaviors, whereas suppression of PVT neuronal activity induced an anxiolytic effect, achieved by using a chemogenetic method. Moreover, we found that the PVT neurons showed plentiful neuronal projections to the bed nucleus of the stria terminalis (BNST). Activation of PVT-BNST neural projections increased the susceptibility of stress-induced anxiety-related behaviors, and inhibition of this neural circuit produced anxiolysis. The insular cortex (IC) is an important upstream region projecting to PVT. Activation of IC-PVT neuronal projections enhanced susceptibility to stress induced anxious behaviors. Inhibiting this neural circuit suppressed anxious behaviors. Moreover, anterograde monosynaptic tracing results showed that the IC exerts strong neuronal projections to PVT, forming synaptic connections with its neurons, and these neurons throw extensive neuronal fibers to form synapse with BNST neurons. Finally, our results showed that ablation of neurons in PVT receiving monosynaptic input from IC attenuated the anxiety-related phenotypes induced by activating IC neurons. Lesions of the neurons in BNST synaptic origination from PVT blocked the anxiety-related phenotypes induced by activating PVT neurons. Our findings indicate that the PVT is a crucial anxiety-regulating nucleus, and the IC-PVT-BNST neural projection is an essential pathway affecting anxiety morbidity and treatment.


Assuntos
Núcleos Septais , Núcleos Septais/fisiologia , Córtex Insular , Tálamo , Ansiedade , Neurônios , Vias Neurais/fisiologia
10.
J Physiol ; 599(21): 4883-4900, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34510418

RESUMO

Paraventricular thalamus (PVT) is a midline thalamic area that receives dense GABA projections from zona incerta (ZI) for the regulation of feeding behaviours. Activation of central serotonin (5-HT) signalling is known to inhibit food intake. Although previous studies have reported both 5-HT fibres and receptors in the PVT, it remains unknown how 5-HT regulates PVT neurons and whether PVT 5-HT signalling is involved in the control of food intake. Using slice patch-clamp recordings in combination with optogenetics, we found that 5-HT not only directly excited PVT neurons by activating 5-HT7 receptors to modulate hyperpolarization-activated cyclic nucleotide-gated (HCN) channels but also disinhibited these neurons by acting on presynaptic 5-HT1A receptors to reduce GABA inhibition. Specifically, 5-HT depressed photostimulation-evoked inhibitory postsynaptic currents (eIPSCs) in PVT neurons innervated by channelrhodopsin-2-positive GABA axons from ZI. Using paired-pulse photostimulation, we found 5-HT increased paired-pulse ratios of eIPSCs, suggesting 5-HT decreases ZI-PVT GABA release. Furthermore, we found that exposure to a high-fat-high-sucrose diet for 2 weeks impaired both 5-HT inhibition of ZI-PVT GABA transmission and 5-HT excitation of PVT neurons. Using retrograde tracer in combination with immunocytochemistry and slice electrophysiology, we found that PVT-projecting dorsal raphe neurons expressed 5-HT and were inhibited by food deprivation. Together, our study reveals the mechanism by which 5-HT activates PVT neurons through both direct excitation and indirect disinhibition from the ZI. The downregulation in 5-HT excitation and disinhibition of PVT neurons may contribute to the development of overeating and obesity after chronic high-fat diet. KEY POINTS: Serotonin (5-HT) depolarizes and excites paraventricular thalamus (PVT) neurons. 5-HT7 receptors are responsible for 5-HT excitation of PVT neurons and the coupling of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels to 5-HT receptors in part mediates the excitatory effect of 5-HT. 5-HT depresses the frequency of spontaneous inhibitory but not excitatory postsynaptic currents in PVT neurons. 5-HT1A receptors contribute to the depressive effect of 5-HT on inhibitory transmissions. 5-HT inhibits GABA release from zona incerta (ZI) GABA terminals in PVT. Chronic high-fat diet not only impairs 5-HT inhibition of the ZI-PVT GABA transmission but also downregulates 5-HT excitation of PVT neurons. PVT-projecting dorsal raphe neurons express 5-HT and are inhibited by food deprivation.


Assuntos
Serotonina , Zona Incerta , Potenciais Pós-Sinápticos Excitadores , Neurônios , Tálamo
11.
Brain Behav Immun ; 95: 36-44, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33540073

RESUMO

Many temperate zone animals exhibit seasonal rhythms in physiology and behavior, including seasonal cycles of reproduction, energetics, stress responsiveness, and immune function, among many others. These rhythms are driven by seasonal changes in the duration of pineal melatonin secretion. The neural melatonin target tissues that mediate several of these rhythms have been identified, though the target(s) mediating melatonin's regulation of glucocorticoid secretion, immune cell numbers, and bacterial killing capacity remain unspecified. The present results indicate that one melatonin target tissue, the paraventricular nucleus of the thalamus (PVT), is necessary for the expression of these seasonal rhythms. Thus, while radiofrequency ablations of the PVT failed to alter testicular and body mass response to short photoperiod exposure, they did block the effect of short day lengths on cortisol secretion and bacterial killing efficacy. These results are consistent with the independent regulation by separate neural circuits of several physiological traits that vary seasonally in mammals.


Assuntos
Melatonina , Glândula Pineal , Animais , Ritmo Circadiano , Fotoperíodo , Estações do Ano , Tálamo
12.
J Neurosci ; 39(25): 4945-4958, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-30979815

RESUMO

Decision-making often involves motivational conflict because of the competing demands of approach and avoidance for a common resource: behavior. This conflict must be resolved as a necessary precursor for adaptive behavior. Here we show a role for the paraventricular thalamus (PVT) in behavioral control during motivational conflict. We used Pavlovian counterconditioning in male rats to establish a conditioned stimulus (CS) as a signal for reward (or danger) and then transformed the same CS into a signal for danger (or reward). After such training, the CS controls conflicting appetitive and aversive behaviors. To assess PVT involvement in conflict, we injected an adeno-associated virus (AAV) expressing the genetically encoded Ca2+ indicator GCaMP and used fiber photometry to record population PVT Ca2+ signals. We show distinct profiles of responsivity across the anterior-posterior axis of PVT during conflict, including an ordinal relationship between posterior PVT CS responses and behavior strength. To study the causal role of PVT in behavioral control during conflict, we injected AAV expressing the inhibitory hM4Di DREADD and determined the effects of chemogenetic PVT inhibition on behavior. We show that chemogenetic inhibition across the anterior-posterior axis of the PVT, but not anterior or posterior PVT alone, disrupts arbitration between appetitive and aversive behaviors when they are in conflict but has no effect when these behaviors are assessed in isolation. Together, our findings identify PVT as central to behavioral control during motivational conflict.SIGNIFICANCE STATEMENT Animals, including humans, approach attractive stimuli and avoid aversive ones. However, they frequently face conflict when the demands of approach and avoidance are incompatible. Resolution of this conflict is fundamental to adaptive behavior. Here we show a role for the paraventricular thalamus, a nucleus of the dorsal midline thalamus, in the arbitration of appetitive and aversive behavior during motivational conflict.


Assuntos
Condicionamento Operante/fisiologia , Tomada de Decisões/fisiologia , Núcleos da Linha Média do Tálamo/fisiologia , Motivação/fisiologia , Animais , Sinais (Psicologia) , Masculino , Atividade Motora/fisiologia , Ratos , Ratos Sprague-Dawley , Recompensa
13.
J Neurosci ; 37(11): 3018-3029, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28193686

RESUMO

Foraging animals balance the need to seek food and energy against the accompanying dangers of injury and predation. To do so, they rely on learning systems encoding reward and danger. Whereas much is known about these separate learning systems, little is known about how they interact to shape and guide behavior. Here we show a key role for the rat paraventricular nucleus of the thalamus (PVT), a nucleus of the dorsal midline thalamus, in this interaction. First, we show behavioral competition between reward and danger: the opportunity to seek food reward negatively modulates expression of species-typical defensive behavior. Then, using a chemogenetic approach expressing the inhibitory hM4Di designer receptor exclusively activated by a designer drug in PVT neurons, we show that the PVT is central to this behavioral competition. Chemogenetic PVT silencing biases behavior toward either defense or reward depending on the experimental conditions, but does not consistently favor expression of one over the other. This bias could not be attributed to changes in fear memory retrieval, learned safety, or memory interference. Rather, our results demonstrate that the PVT is essential for balancing conflicting behavioral tendencies toward danger and reward, enabling adaptive responding under this basic selection pressure.SIGNIFICANCE STATEMENT Among the most basic survival problems faced by animals is balancing the need to seek food and energy against the accompanying dangers of injury and predation. Although much is known about the brain mechanisms that underpin learning about reward and danger, little is known about how these interact to solve basic survival problems. Here we show competition between defensive (to avoid predatory detection) and approach (to obtain food) behavior. We show that the paraventricular thalamus, a nucleus of the dorsal midline thalamus, is integral to this behavioral competition. The paraventricular thalamus balances the competing behavioral demands of danger and reward, enabling adaptive responding under this selection pressure.


Assuntos
Aprendizagem da Esquiva/fisiologia , Tomada de Decisões/fisiologia , Mecanismos de Defesa , Núcleos da Linha Média do Tálamo/fisiologia , Rede Nervosa/fisiologia , Recompensa , Animais , Masculino , Ratos , Ratos Sprague-Dawley
14.
Adv Exp Med Biol ; 1099: 101-114, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30306518

RESUMO

The prevalence of chronic pain increases with age. The pain occurrence in the elderly ranges from 25% to 80% in different countries. Ischemic heart disease is also prevailing in the aged people. Restored blood flow quickly rescues myocardium but also causes ischemia-reperfusion (IR) injury. Brief episodes of ischemia at a distant organ could reduce the myocardial reperfusion injury. This is called remote ischemic preconditioning (RIPC) cardioprotection. Several circulating factors and neurogenic signals contribute to the cardioprotection by RIPC. Preinfarction angina, a form of chest pain, is associated with significant cardioprotection in myocardial infarction patients. Activation of peripheral nociception also induces cardioprotection against IR injury via neurogenic pathway. It is possible that angina also induces nociceptive signal pathway to provide cardioprotection. It is unclear whether pre-existing chronic pain will also have a cardioprotection effect. We recently reported chronic neuropathic pain attenuates cardiac IR injury in mice. ERK activation in anterior nucleus of paraventricular thalamus (PVA) is required for this remote cardioprotection. Direct activation of PVA neurons also provides cardioprotection against cardiac IR injury. Chronic neuropathic pain-induced cardioprotection requires activation of parasympathetic nerves. This review summarizes the potential interaction of chronic pain and cardiac IR injury.


Assuntos
Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Neuralgia , Animais , Coração , Humanos , Camundongos , Sistema Nervoso Parassimpático
15.
J Neurosci ; 36(12): 3567-78, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27013685

RESUMO

Midline thalamus is implicated in linking visceral and exteroceptive sensory information with behavior. However, whether neuronal activity is modulated with temporal precision by cues and actions in real time is unknown. Using single-neuron recording and a Pavlovian visual-cue/liquid-reward association task in rats, we discovered phasic responses to sensory cues, appropriately timed to modify information processing in output targets, as well as tonic modulations within and between trials that were differentially reward modulated, which may have distinct arousal functions. Many of the cue-responsive neurons also responded to repetitive licks, consistent with sensorimotor integration. Further, some lick-related neurons were activated only by the first rewarded lick and only if that lick were also part of a conditioned response sequence initiated earlier, consistent with binding action decisions to their ensuing outcome. This rich repertoire of responses provides electrophysiological evidence for midline thalamus as a site of complex information integration for reward-mediated behavior. SIGNIFICANCE STATEMENT: Disparate brain circuits are involved in sensation, movement, and reward information. These must interact in order for the relationships between cues, actions, and outcomes to be learned. We found that responses of single neurons in midline thalamus to sensory cues are increased when associated with reward. This output may amplify similar signals generated in parallel by the dopamine system. In addition, some neurons coded a three-factor decision in which the neuron fired only if there was a movement, if it was the first one after the reward becoming available, and if it was part of a sequence triggered in response to a preceding cue. These data highlight midline thalamus as an important node integrating multiple types of information for linking sensation, actions, and rewards.


Assuntos
Núcleos da Linha Média do Tálamo/fisiologia , Movimento/fisiologia , Neurônios/fisiologia , Desempenho Psicomotor/fisiologia , Recompensa , Língua/fisiologia , Animais , Sinais (Psicologia) , Masculino , Ratos , Ratos Wistar
16.
Addict Biol ; 22(1): 58-69, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26223289

RESUMO

The paraventricular nucleus of the thalamus (PVT) appears to participate in drug addiction. Recent evidence in rats shows that ethanol drinking is increased by orexin/hypocretin (OX) afferents from the hypothalamus, acting specifically in the anterior (aPVT) rather than posterior (pPVT) PVT subregion. The present study sought to identify neuropeptides transcribed within the PVT, which themselves might contribute to ethanol drinking and possibly mediate the actions of OX. We discovered that substance P (SP) in the aPVT can stimulate intermittent-access ethanol drinking, similar to OX, and that SP receptor [neurokinin 1 receptor/tachykinin receptor 1 (NK1R)] antagonists in this subregion reduce ethanol drinking. As with OX, this effect is site specific, with SP in the pPVT or dorsal third ventricle having no effect on ethanol drinking, and it is behaviorally specific, with SP in the aPVT reducing the drinking of sucrose and stimulating it in the pPVT. A close relationship between SP and OX was demonstrated by a stimulatory effect of local OX injection on SP mRNA and peptide levels, specifically in the aPVT but not pPVT, and a stimulatory effect of OX on SP expression in isolated thalamic neurons, reflecting postsynaptic actions. A functional relationship between OX and SP in the aPVT is suggested by our additional finding that ethanol drinking induced by OX is blocked by a local NK1R antagonist administered at a sub-threshold dose. These results, suggesting that SP in the aPVT mediates the stimulatory effect of OX on ethanol drinking, identify a new role for SP in the control of this behavior.


Assuntos
Comportamento Animal , Etanol/administração & dosagem , Hipotálamo/metabolismo , Orexinas/metabolismo , Substância P/metabolismo , Núcleos Talâmicos/metabolismo , Animais , Depressores do Sistema Nervoso Central/administração & dosagem , Masculino , Modelos Animais , Neurotransmissores/metabolismo , Ratos , Ratos Long-Evans
17.
Adv Sci (Weinh) ; : e2401855, 2024 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-38973158

RESUMO

Clinically, chronic pain and depression often coexist in multiple diseases and reciprocally reinforce each other, which greatly escalates the difficulty of treatment. The neural circuit mechanism underlying the chronic pain/depression comorbidity remains unclear. The present study reports that two distinct subregions in the paraventricular thalamus (PVT) play different roles in this pathological process. In the first subregion PVT posterior (PVP), glutamatergic neurons (PVPGlu) send signals to GABAergic neurons (VLPAGGABA) in the ventrolateral periaqueductal gray (VLPAG), which mediates painful behavior in comorbidity. Meanwhile, in another subregion PVT anterior (PVA), glutamatergic neurons (PVAGlu) send signals to the nucleus accumbens D1-positive neurons and D2-positive neurons (NAcD1→D2), which is involved in depression-like behavior in comorbidity. This study demonstrates that the distinct thalamo-subcortical circuits PVPGlu→VLPAGGABA and PVAGlu→NAcD1→D2 mediated painful behavior and depression-like behavior following spared nerve injury (SNI), respectively, which provides the circuit-based potential targets for preventing and treating comorbidity.

18.
CNS Neurosci Ther ; 30(4): e14534, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-37994678

RESUMO

AIMS: Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder, but its pathogenesis remains incompletely understood, particularly the involvements of central nervous system sensitization in colorectal visceral pain. Our study was to investigate whether the paraventricular thalamus (PVT) projected to the insular cortex (IC) to regulate colorectal visceral pain in neonatal colonic inflammation (NCI) mice and underlying mechanisms. METHODS: We applied optogenetic, chemogenetic, or pharmacological approaches to manipulate the glutamatergicPVT-IC pathway. Fiber photometry was used to assess neuronal activity. Electromyography activities in response to colorectal distension (CRD) were measured to evaluate the colorectal visceral pain. RESULTS: NCI enhanced c-Fos expression and calcium activity upon CRD in the ICGlu, and optogenetic manipulation of them altered colorectal visceral pain responses accordingly. Viral tracing indicated that the PVTGlu projected to the ICGlu. Optogenetic manipulation of PVTGlu changed colorectal visceral pain responses. Furthermore, selective optogenetic modulation of PVT projections in the IC influenced colorectal visceral pain, which was reversed by chemogenetic manipulation of downstream ICGlu. CONCLUSIONS: This study identified a novel PVT-IC neural circuit playing a critical role in colorectal visceral pain in a mouse model of IBS.


Assuntos
Neoplasias Colorretais , Síndrome do Intestino Irritável , Dor Visceral , Animais , Camundongos , Dor Visceral/metabolismo , Síndrome do Intestino Irritável/metabolismo , Córtex Insular , Tálamo , Inflamação
19.
Neuron ; 112(5): 772-785.e9, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38141605

RESUMO

Lack of behavioral suppression typifies substance use disorders, yet the neural circuit underpinnings of drug-induced behavioral disinhibition remain unclear. Here, we employ deep-brain two-photon calcium imaging in heroin self-administering mice, longitudinally tracking adaptations within a paraventricular thalamus to nucleus accumbens behavioral inhibition circuit from the onset of heroin use to reinstatement. We find that select thalamo-accumbal neuronal ensembles become profoundly hypoactive across the development of heroin seeking and use. Electrophysiological experiments further reveal persistent adaptations at thalamo-accumbal parvalbumin interneuronal synapses, whereas functional rescue of these synapses prevents multiple triggers from initiating reinstatement of heroin seeking. Finally, we find an enrichment of µ-opioid receptors in output- and cell-type-specific paraventricular thalamic neurons, which provide a mechanism for heroin-induced synaptic plasticity and behavioral disinhibition. These findings reveal key circuit adaptations that underlie behavioral disinhibition in opioid dependence and further suggest that recovery of this system would reduce relapse susceptibility.


Assuntos
Heroína , Transtornos Relacionados ao Uso de Opioides , Ratos , Camundongos , Animais , Heroína/farmacologia , Ratos Sprague-Dawley , Autoadministração/métodos , Neurônios , Núcleo Accumbens/fisiologia
20.
Elife ; 122023 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-36867023

RESUMO

The paraventricular nucleus of the thalamus (PVT) is known to regulate various cognitive and behavioral processes. However, while functional diversity among PVT circuits has often been linked to cellular differences, the molecular identity and spatial distribution of PVT cell types remain unclear. To address this gap, here we used single nucleus RNA sequencing (snRNA-seq) and identified five molecularly distinct PVT neuronal subtypes in the mouse brain. Additionally, multiplex fluorescent in situ hybridization of top marker genes revealed that PVT subtypes are organized by a combination of previously unidentified molecular gradients. Lastly, comparing our dataset with a recently published single-cell sequencing atlas of the thalamus yielded novel insight into the PVT's connectivity with the cortex, including unexpected innervation of auditory and visual areas. This comparison also revealed that our data contains a largely non-overlapping transcriptomic map of multiple midline thalamic nuclei. Collectively, our findings uncover previously unknown features of the molecular diversity and anatomical organization of the PVT and provide a valuable resource for future investigations.


Assuntos
Núcleo Hipotalâmico Paraventricular , Tálamo , Ratos , Camundongos , Animais , Hibridização in Situ Fluorescente , Ratos Sprague-Dawley , Vias Neurais/fisiologia , Núcleos da Linha Média do Tálamo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA