Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 308
Filtrar
1.
Schizophr Res ; 267: 113-121, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38531158

RESUMEN

A decreased expression of specific interneuron subtypes, containing either the calcium binding protein parvalbumin (PV) or the neurotransmitter somatostatin (SST), are observed in the cortex and hippocampus of both patients with schizophrenia and rodent models used to study the disorder. Moreover, preclinical studies suggest that this loss of inhibitory function is a key pathological mechanism underlying the symptoms of schizophrenia. Interestingly, decreased expression of Lhx6, a key transcriptional regulator specific to the development and migration of PV and SST interneurons, is seen in human postmortem studies and following multiple developmental disruptions used to model schizophrenia preclinically. These results suggest that disruptions in interneuron development in utero may contribute to the pathology of the disorder. To recapitulate decreased Lhx6 expression during development, we used in utero electroporation to introduce an Lhx6 shRNA plasmid and knockdown Lhx6 expression in the brains of rats on gestational day 17. We then examined schizophrenia-like neurophysiological and behavioral alterations in the offspring once they reached adulthood. In utero Lhx6 knockdown resulted in increased ventral tegmental area (VTA) dopamine neuron population activity and a sex-specific increase in locomotor response to a psychotomimetic, consistent with positive symptomology of schizophrenia. However, Lhx6 knockdown had no effect on social interaction or spatial working memory, suggesting behaviors associated with negative and cognitive symptom domains were unaffected. These results suggest that knockdown of Lhx6 during development results in neurophysiological and behavioral alterations consistent with the positive symptom domain of schizophrenia in adult rats.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas con Homeodominio LIM , Esquizofrenia , Factores de Transcripción , Animales , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Esquizofrenia/genética , Femenino , Masculino , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ratas , Embarazo , Técnicas de Silenciamiento del Gen , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/fisiopatología , Interneuronas/metabolismo , Interneuronas/fisiología , Ratas Sprague-Dawley , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Interferente Pequeño
2.
Glia ; 71(8): 1906-1920, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37017183

RESUMEN

Microglia participates in the modulation of pain signaling. The activation of microglia is suggested to play an important role in affective disorders that are related to a dysfunction of the mesocorticolimbic system (MCLS) and are commonly associated with chronic pain. Moreover, there is evidence that mu-opioid receptors (MORs), expressed in the MCLS, are involved in neuroinflammatory events, although the way by which they do it remains to be elucidated. In this study, we propose that MOR pharmacological activation within the MCLS activates and triggers the local release of proinflammatory cytokines and this pattern of activation is impacted by the presence of systemic inflammatory pain. To test this hypothesis, we used in vivo microdialysis coupled with flow cytometry to measure cytokines release in the nucleus accumbens and immunofluorescence of IBA1 in areas of the MCLS on a rat model of inflammatory pain. Interestingly, the treatment with DAMGO, a MOR agonist locally in the nucleus accumbens, triggered the release of the IL1α, IL1ß, and IL6 proinflammatory cytokines. Furthermore, MOR pharmacological activation in the ventral tegmental area (VTA) modified the levels of IBA1-positive cells in the VTA, prefrontal cortex, the nucleus accumbens and the amygdala in a dose-dependent way, without impacting mechanical nociception. Additionally, MOR blockade in the VTA prevents DAMGO-induced effects. Finally, we observed that systemic inflammatory pain altered the IBA1 immunostaining derived from MOR activation in the MSCLS. Altogether, our results indicate that the microglia-MOR relationship could be pivotal to unravel some inflammatory pain-induced comorbidities related to MCLS dysfunction.


Asunto(s)
Dolor Crónico , Microglía , Enfermedades Neuroinflamatorias , Corteza Prefrontal , Receptores Opioides mu , Área Tegmental Ventral , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/fisiopatología , Microglía/metabolismo , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/fisiopatología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Animales , Ratas , Modelos Animales de Enfermedad , Dolor Crónico/metabolismo , Dolor Crónico/fisiopatología , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiopatología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Microfilamentos/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Masculino , Femenino , Ratas Sprague-Dawley
3.
Science ; 377(6601): 63-72, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35771921

RESUMEN

In mice, social defeat stress (SDS), an ethological model for psychosocial stress, induces sleep. Such sleep could enable resilience, but how stress promotes sleep is unclear. Activity-dependent tagging revealed a subset of ventral tegmental area γ-aminobutyric acid (GABA)-somatostatin (VTAVgat-Sst) cells that sense stress and drive non-rapid eye movement (NREM) and REM sleep through the lateral hypothalamus and also inhibit corticotropin-releasing factor (CRF) release in the paraventricular hypothalamus. Transient stress enhances the activity of VTAVgat-Sst cells for several hours, allowing them to exert their sleep effects persistently. Lesioning of VTAVgat-Sst cells abolished SDS-induced sleep; without it, anxiety and corticosterone concentrations remained increased after stress. Thus, a specific circuit allows animals to restore mental and body functions by sleeping, potentially providing a refined route for treating anxiety disorders.


Asunto(s)
Resiliencia Psicológica , Sueño , Derrota Social , Estrés Psicológico , Área Tegmental Ventral , Animales , Hormona Liberadora de Corticotropina/metabolismo , Área Hipotalámica Lateral/fisiopatología , Ratones , Sueño REM , Somatostatina/metabolismo , Estrés Psicológico/fisiopatología , Área Tegmental Ventral/fisiopatología , Ácido gamma-Aminobutírico/metabolismo
4.
Nat Commun ; 13(1): 577, 2022 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-35102141

RESUMEN

Emotional stress is considered a severe pathogenetic factor of psychiatric disorders. However, the circuit mechanisms remain largely unclear. Using a three-chamber vicarious social defeat stress (3C-VSDS) model in mice, we here show that chronic emotional stress (CES) induces anxiety-like behavior and transient social interaction changes. Dopaminergic neurons of ventral tegmental area (VTA) are required to control this behavioral deficit. VTA dopaminergic neuron hyperactivity induced by CES is involved in the anxiety-like behavior in the innate anxiogenic environment. Chemogenetic activation of VTA dopaminergic neurons directly triggers anxiety-like behavior, while chemogenetic inhibition of these neurons promotes resilience to the CES-induced anxiety-like behavior. Moreover, VTA dopaminergic neurons receiving nucleus accumbens (NAc) projections are activated in CES mice. Bidirectional modulation of the NAc-VTA circuit mimics or reverses the CES-induced anxiety-like behavior. In conclusion, we propose that a NAc-VTA circuit critically establishes and regulates the CES-induced anxiety-like behavior. This study not only characterizes a preclinical model that is representative of the nuanced aspect of CES, but also provides insight to the circuit-level neuronal processes that underlie empathy-like behavior.


Asunto(s)
Ansiedad/fisiopatología , Conducta Animal/fisiología , Vías Nerviosas/fisiopatología , Núcleo Accumbens/fisiopatología , Distrés Psicológico , Derrota Social , Área Tegmental Ventral/fisiopatología , Animales , Dependovirus/fisiología , Depresión/fisiopatología , Depresión/psicología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas GABAérgicas/metabolismo , Integrasas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Sinapsis/metabolismo , Ácido gamma-Aminobutírico/metabolismo
5.
Brain Res ; 1778: 147779, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35007546

RESUMEN

Pain can be ignited by noxious chemical (e.g., acid), mechanical (e.g., pressure), and thermal (e.g., heat) stimuli and generated by the activation of sensory neurons and their axonal terminals called nociceptors in the periphery. Nociceptive information transmitted from the periphery is projected to the central nervous system (thalamus, somatosensory cortex, insular, anterior cingulate cortex, amygdala, periaqueductal grey, prefrontal cortex, etc.) to generate a unified experience of pain. Local field potential (LFP) recording is one of the neurophysiological tools to investigate the combined neuronal activity, ranging from several hundred micrometers to a few millimeters (radius), located around the embedded electrode. The advantage of recording LFP is that it provides stable simultaneous activities in various brain regions in response to external stimuli. In this study, differential LFP activities from the contralateral anterior cingulate cortex (ACC), ventral tegmental area (VTA), and bilateral amygdala in response to peripheral noxious formalin injection were recorded in anesthetized male rats. The results indicated increased power of delta, theta, alpha, beta, and gamma bands in the ACC and amygdala but no change of gamma-band in the right amygdala. Within the VTA, intensities of the delta, theta, and beta bands were only enhanced significantly after formalin injection. It was found that the connectivity (i.t. the coherence) among these brain regions reduced significantly under the formalin-induced nociception, which suggests a significant interruption within the brain. With further study, it will sort out the key combination of structures that will serve as the signature for pain state.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Ondas Encefálicas/fisiología , Giro del Cíngulo/fisiopatología , Dolor Nociceptivo/fisiopatología , Área Tegmental Ventral/fisiopatología , Animales , Modelos Animales de Enfermedad , Desinfectantes/farmacología , Fenómenos Electrofisiológicos , Formaldehído/farmacología , Inflamación/inducido químicamente , Ratas
6.
Exp Neurol ; 350: 113969, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34973962

RESUMEN

Gradual decline in cognitive and non-cognitive functions are considered clinical hallmarks of Alzheimer's Disease (AD). Post-mortem autoptic analysis shows the presence of amyloid ß deposits, neuroinflammation and severe brain atrophy. However, brain circuit alterations and cellular derailments, assessed in very early stages of AD, still remain elusive. The understanding of these early alterations is crucial to tackle defective mechanisms. In a previous study we proved that the Tg2576 mouse model of AD displays functional deficits in the dorsal hippocampus and relevant behavioural AD-related alterations. We had shown that these deficits in Tg2576 mice correlate with the precocious degeneration of dopamine (DA) neurons in the Ventral Tegmental Area (VTA) and can be restored by L-DOPA treatment. Due to the distinct functionality and connectivity of dorsal versus ventral hippocampus, here we investigated neuronal excitability and synaptic functionality in the ventral CA1 hippocampal sub-region of Tg2576 mice. We found an age-dependent alteration of cell excitability and firing in pyramidal neurons starting at 3 months of age, that correlates with reduced levels in the ventral CA1 of tyrosine hydroxylase - the rate-limiting enzyme of DA synthesis. Additionally, at odds with the dorsal hippocampus, we found no alterations in basal glutamatergic transmission and long-term plasticity of ventral neurons in 8-month old Tg2576 mice compared to age-matched controls. Last, we used computational analysis to model the early derailments of firing properties observed and hypothesize that the neuronal alterations found could depend on dysfunctional sodium and potassium conductances, leading to anticipated depolarization-block of action potential firing. The present study depicts that impairment of cell excitability and homeostatic control of firing in ventral CA1 pyramidal neurons is a prodromal feature in Tg2576 AD mice.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Región CA1 Hipocampal/fisiopatología , Fenómenos Electrofisiológicos , Células Piramidales , Potenciales de Acción , Envejecimiento , Animales , Dopaminérgicos/farmacología , Neuronas Dopaminérgicas , Femenino , Levodopa/farmacología , Masculino , Ratones , Ratones Transgénicos , Canales de Potasio , Canales de Sodio , Tirosina 3-Monooxigenasa/metabolismo , Área Tegmental Ventral/fisiopatología
7.
Neuropharmacology ; 202: 108859, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34710468

RESUMEN

Nicotine, the addictive component of tobacco, has bivalent rewarding and aversive properties. Recently, the lateral habenula (LHb), a structure that controls ventral tegmental area (VTA) dopamine (DA) function, has attracted attention as it is potentially involved in the aversive properties of drugs of abuse. Hitherto, the LHb-modulation of nicotine-induced VTA neuronal activity in vivo is unknown. Using standard single-extracellular recording in anesthetized rats, we observed that intravenous administration of nicotine hydrogen tartrate (25-800 µg/kg i.v.) caused a dose-dependent increase in the basal firing rate of the LHb neurons of nicotine-naïve rats. This effect underwent complete desensitization in chronic nicotine (6 mg/kg/day for 14 days)-treated animals. As previously reported, acute nicotine induced an increase in the VTA DA neuronal firing rate. Interestingly, only neurons located medially (mVTA) but not laterally (latVTA) within the VTA were responsive to acute nicotine. This pattern of activation was reversed by chronic nicotine exposure which produced the selective increase of latVTA neuronal activity. Acute lesion of the LHb, similarly to chronic nicotine treatment, reversed the pattern of DA cell activation induced by acute nicotine increasing latVTA but not mVTA neuronal activity. Our evidence indicates that LHb plays an important role in mediating the effects of acute and chronic nicotine within the VTA by activating distinct subregional responses of DA neurons. The LHb/VTA modulation might be part of the neural substrate of nicotine aversive properties. By silencing the LHb chronic nicotine could shift the balance of motivational states toward the reward.


Asunto(s)
Dopamina/fisiología , Electroencefalografía/métodos , Habénula/efectos de los fármacos , Habénula/fisiopatología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiopatología , Nicotina/efectos adversos , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/fisiopatología , Animales , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/fisiología , Relación Dosis-Respuesta a Droga , Masculino , Nicotina/farmacología , Ratas Sprague-Dawley , Recompensa
8.
Cell Rep ; 37(5): 109936, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34731609

RESUMEN

Depression symptoms are often found in patients suffering from chronic pain, a phenomenon that is yet to be understood mechanistically. Here, we systematically investigate the cellular mechanisms and circuits underlying the chronic-pain-induced depression behavior. We show that the development of chronic pain is accompanied by depressive-like behaviors in a mouse model of trigeminal neuralgia. In parallel, we observe increased activity of the dopaminergic (DA) neuron in the midbrain ventral tegmental area (VTA), and inhibition of this elevated VTA DA neuron activity reverses the behavioral manifestations of depression. Further studies establish a pathway of glutamatergic projections from the spinal trigeminal subnucleus caudalis (Sp5C) to the lateral parabrachial nucleus (LPBN) and then to the VTA. These glutamatergic projections form a direct circuit that controls the development of the depression-like behavior under the state of the chronic neuropathic pain.


Asunto(s)
Conducta Animal , Dolor Crónico/fisiopatología , Depresión/fisiopatología , Núcleos Parabraquiales/fisiopatología , Neuralgia del Trigémino/fisiopatología , Área Tegmental Ventral/fisiopatología , Potenciales de Acción , Animales , Dolor Crónico/metabolismo , Dolor Crónico/psicología , Depresión/metabolismo , Depresión/psicología , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Neuronas Dopaminérgicas/metabolismo , Femenino , Ácido Glutámico/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Núcleos Parabraquiales/metabolismo , Núcleo Caudal del Trigémino/metabolismo , Núcleo Caudal del Trigémino/fisiopatología , Neuralgia del Trigémino/metabolismo , Neuralgia del Trigémino/psicología , Área Tegmental Ventral/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
9.
Mol Psychiatry ; 26(11): 6170-6186, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34642456

RESUMEN

Plasticity of neurons in the ventral tegmental area (VTA) is critical for establishment of drug dependence. However, the remodeling of the circuits mediating the transition between positive and negative effect remains unclear. Here, we used neuronal activity-dependent labeling technique to characterize and temporarily control the VTA neuronal ensembles recruited by the initial morphine exposure (morphine-positive ensembles, Mor-Ens). Mor-Ens preferentially projected to NAc, and induced dopamine-dependent positive reinforcement. Electrophysiology and rabies viral tracing revealed the preferential connections between the VTA-projective corticotrophin-releasing hormone (CRH) neurons of central amygdala (CRHCeA→VTA) and Mor-Ens, which was enhanced after escalating morphine exposure and mediated the negative effect during opiate withdrawal. Pharmacologic intervention or CRISPR-mediated repression of CRHR1 in Mor-Ens weakened the inhibitory CRHCeA→VTA inputs, and alleviated the negative effect during opiate withdrawal. These data suggest that neurons encoding opioid reward experience are inhibited by enhanced CRHCeA→VTA inputs induced by chronic morphine exposure, leading to negative effect during opiate withdrawal, and provide new insight into the pathological changes in VTA plasticity after drug abuse and mechanism of opiate dependence.


Asunto(s)
Morfina/efectos adversos , Plasticidad Neuronal , Síndrome de Abstinencia a Sustancias/fisiopatología , Área Tegmental Ventral , Amígdala del Cerebelo/citología , Analgésicos Opioides/efectos adversos , Animales , Hormona Liberadora de Corticotropina/metabolismo , Área Tegmental Ventral/citología , Área Tegmental Ventral/fisiopatología
10.
Biochem Biophys Res Commun ; 579: 22-28, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34583191

RESUMEN

Orexin has been implicated in comorbid diseases of depression, making it a promising target for anti-depression treatment. Although orexin neurons exhibit abnormal activity in depression, the neurocircuit mechanism of orexin remains unclear. As one of the important downstream factors of orexin neurons, the ventral tegmental area (VTA) is considered crucial to the mechanism of depression. However, the role of VTA orexinergic afferents in depression remains unclear. In this study, we applied a combination of opto/chemogenetic and neuropharmacology methods to investigate whether the VTA orexinergic afferents participate in the pathogenesis of depression in a chronic unpredictable mild stress (CUMS) mouse model. We found that c-Fos expression in these VTA-projecting orexin neurons specifically decreased in CUMS-treated mice. Optogenetic and chemogenetic activation of orexin terminals in the VTA significantly reversed depressive behavior. Microinjection of orexin-A, but not orexin-B, into the VTA significantly improved depressive-like behavior. Our study provided direct evidence that the VTA orexinergic afferents participate in the mechanism of depression, and the orexin-1 receptor plays a major role.


Asunto(s)
Depresión/metabolismo , Orexinas/farmacología , Área Tegmental Ventral/fisiopatología , Animales , Conducta Animal , Encéfalo/metabolismo , Conducta de Elección , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Optogenética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Estrés Fisiológico , Azúcares , Área Tegmental Ventral/efectos de los fármacos
11.
Neuropharmacology ; 198: 108763, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34433088

RESUMEN

Between 2005 and 2009, several research groups identified a strikingly dense inhibitory input to midbrain dopamine neurons arising from a previously uncharted region posterior to the ventral tegmental area (VTA). This region is now denoted as either the rostromedial tegmental nucleus (RMTg) or the "tail of the VTA" (tVTA), and is recognized to express distinct genetic markers, encode negative "prediction errors" (inverse to dopamine neurons), and play critical roles in behavioral inhibition and punishment learning. RMTg neurons are also influenced by many categories of abused drugs, and may drive some aversive responses to such drugs, particularly cocaine and alcohol. However, despite much progress, many important questions remain about RMTg molecular/genetic properties, diversity of projection targets, and applications to addiction, depression, and other neuropsychiatric disorders. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.


Asunto(s)
Conducta Animal/fisiología , Conducta/fisiología , Dopamina/fisiología , Área Tegmental Ventral/fisiología , Animales , Neuronas Dopaminérgicas/fisiología , Humanos , Trastornos Relacionados con Sustancias/fisiopatología , Trastornos Relacionados con Sustancias/psicología , Tegmento Mesencefálico/efectos de los fármacos , Área Tegmental Ventral/fisiopatología
12.
Nat Neurosci ; 24(10): 1402-1413, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34373644

RESUMEN

Pain decreases the activity of many ventral tegmental area (VTA) dopamine (DA) neurons, yet the underlying neural circuitry connecting nociception and the DA system is not understood. Here we show that a subpopulation of lateral parabrachial (LPB) neurons is critical for relaying nociceptive signals from the spinal cord to the substantia nigra pars reticulata (SNR). SNR-projecting LPB neurons are activated by noxious stimuli and silencing them blocks pain responses in two different models of pain. LPB-targeted and nociception-recipient SNR neurons regulate VTA DA activity directly through feed-forward inhibition and indirectly by inhibiting a distinct subpopulation of VTA-projecting LPB neurons thereby reducing excitatory drive onto VTA DA neurons. Correspondingly, ablation of SNR-projecting LPB neurons is sufficient to reduce pain-mediated inhibition of DA release in vivo. The identification of a neural circuit conveying nociceptive input to DA neurons is critical to our understanding of how pain influences learning and behavior.


Asunto(s)
Neuronas Dopaminérgicas , Mesencéfalo/fisiopatología , Vías Nerviosas/fisiopatología , Dolor/fisiopatología , Núcleos Parabraquiales/fisiopatología , Médula Espinal/fisiopatología , Animales , Conducta Animal , Mapeo Encefálico , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas , Nocicepción , Optogenética , Dolor/psicología , Manejo del Dolor , Sustancia Negra/fisiopatología , Área Tegmental Ventral/fisiopatología
13.
Nat Neurosci ; 24(10): 1414-1428, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34385700

RESUMEN

The long-range GABAergic input from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) is relatively understudied, and therefore its role in reward processing has remained unknown. In the present study, we show, in both male and female mice, that long-range GABAergic projections from the VTA to the ventral NAc shell, but not to the dorsal NAc shell or NAc core, are engaged in reward and reinforcement behavior. We show that this GABAergic projection exclusively synapses on to cholinergic interneurons (CINs) in the ventral NAc shell, thereby serving a specialized function in modulating reinforced reward behavior through the inhibition of ventral NAc shell CINs. These findings highlight the diversity in the structural and functional topography of VTA GABAergic projections, and their neuromodulatory interactions across the dorsoventral gradient of the NAc shell. They also further our understanding of neuronal circuits that are directly implicated in neuropsychiatric conditions such as depression and addiction.


Asunto(s)
Neuronas Colinérgicas/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Refuerzo en Psicología , Área Tegmental Ventral/fisiopatología , Ácido gamma-Aminobutírico/fisiología , Animales , Mapeo Encefálico , Condicionamiento Operante/efectos de los fármacos , Fenómenos Electrofisiológicos , Femenino , Interneuronas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Recompensa , Autoestimulación
14.
Mol Neurobiol ; 58(11): 5635-5648, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34382160

RESUMEN

Numerous human clinical studies have suggested that decreased locomotor activity is a common symptom of major depressive disorder (MDD), as well as other psychiatric diseases. In MDD, the midbrain ventral tegmental area (VTA) dopamine (DA) neurons are closely related to regulate the information processing of reward, motivation, cognition, and aversion. However, the neural circuit mechanism that underlie the relationship between VTA-DA neurons and MDD-related motor impairments, especially hypolocomotion, is still largely unknown. Herein, we investigate how the VTA-DA neurons contribute to the hypolocomotion performance in chronic social defeat stress (CSDS), a mouse model of depression-relevant neurobehavioral states. The results show that CSDS could affect the spontaneous locomotor activity of mice, but not the grip strength and forced locomotor ability. Chemogenetic activation of VTA-DA neurons alleviated CSDS-induced hypolocomotion. Subsequently, quantitative whole-brain mapping revealed decreased projections from VTA-DA neurons to substantia nigra pars reticulata (SNr) after CSDS treatment. Optogenetic activation of dopaminergic projection from VTA to SNr with the stimulation of phasic firing, but not tonic firing, could significantly increase the locomotor activity of mice. Moreover, chemogenetic activation of VTA-SNr dopaminergic circuit in CSDS mice could also rescued the decline of locomotor activity. Taken together, our data suggest that the VTA-SNr dopaminergic projection mediates CSDS-induced hypolocomotion, which provides a theoretical basis and potential therapeutic target for MDD.


Asunto(s)
Dopamina/fisiología , Neuronas Dopaminérgicas/fisiología , Locomoción , Vías Nerviosas/fisiopatología , Porción Reticular de la Sustancia Negra/fisiopatología , Derrota Social , Estrés Psicológico/fisiopatología , Área Tegmental Ventral/fisiopatología , Animales , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Enfermedad Crónica , Clozapina/análogos & derivados , Clozapina/farmacología , Trastorno Depresivo Mayor/fisiopatología , Modelos Animales de Enfermedad , Genes Reporteros , Vectores Genéticos/administración & dosificación , Fuerza de la Mano , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/efectos de los fármacos , Optogenética , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Proteínas Recombinantes/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante , Estrés Psicológico/etiología , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
15.
Sci Rep ; 11(1): 15322, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321562

RESUMEN

DNA methylation and gene expression can be altered by early life stress (ELS) and/or ethanol consumption. The present study aimed to investigate whether DNA methylation of the Vesicular Glutamate Transporters (Vglut)1-3 is related to previously observed Vglut1-3 transcriptional differences in the ventral tegmental area (VTA), nucleus accumbens (Acb), dorsal striatum (dStr) and medial prefrontal cortex (mPFC) of adult rats exposed to ELS, modelled by maternal separation, and voluntary ethanol consumption. Targeted next-generation bisulfite sequencing was performed to identify the methylation levels on 61 5'-cytosine-phosphate-guanosine-3' sites (CpGs) in potential regulatory regions of Vglut1, 53 for Vglut2, and 51 for Vglut3. In the VTA, ELS in ethanol-drinking rats was associated with Vglut1-2 CpG-specific hypomethylation, whereas bidirectional Vglut2 methylation differences at single CpGs were associated with ELS alone. Exposure to both ELS and ethanol, in the Acb, was associated with lower promoter and higher intronic Vglut3 methylation; and in the dStr, with higher and lower methylation in 26% and 43% of the analyzed Vglut1 CpGs, respectively. In the mPFC, lower Vglut2 methylation was observed upon exposure to ELS or ethanol. The present findings suggest Vglut1-3 CpG-specific methylation signatures of ELS and ethanol drinking, underlying previously reported Vglut1-3 transcriptional differences in the mesocorticolimbic brain.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Ansiedad de Separación/genética , Epigénesis Genética , Proteína 1 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/genética , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/fisiopatología , Animales , Ansiedad de Separación/metabolismo , Ansiedad de Separación/fisiopatología , Mapeo Encefálico , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , Islas de CpG , Metilación de ADN/efectos de los fármacos , Etanol/farmacología , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiopatología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Ratas , Ratas Wistar , Transducción de Señal , Estrés Fisiológico/genética , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/fisiopatología , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo
16.
Cell Rep ; 34(12): 108874, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33761364

RESUMEN

Exposure to prolonged stress in critical developmental periods induces heightened vulnerability to psychiatric disorders, which may have sex-specific consequences. Here we investigate the neuronal circuits mediating behavioral changes in mice after chronic adolescent social isolation stress. Escalated aggression is exhibited in stressed males, while social withdrawal is shown in stressed females. In vivo multichannel recordings of free-moving animals indicate that pyramidal neurons in prefrontal cortex (PFC) from stressed males exhibit the significantly decreased spike activity during aggressive attacks, while PFC pyramidal neurons from stressed females show a blunted increase of discharge rates during sociability tests. Chemogenetic and electrophysiological evidence shows that PFC hypofunctioning and BLA principal neuron hyperactivity contribute to the elevated aggression in stressed males, while PFC hypofunctioning and VTA dopamine neuron hypoactivity contribute to the diminished sociability in stressed females. These results establish a framework for understanding the circuit and physiological mechanisms underlying sex-specific divergent effects of stress.


Asunto(s)
Red Nerviosa/fisiopatología , Caracteres Sexuales , Aislamiento Social/psicología , Estrés Psicológico/fisiopatología , Agresión , Amígdala del Cerebelo/fisiopatología , Animales , Enfermedad Crónica , Neuronas Dopaminérgicas/patología , Femenino , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Corteza Prefrontal/fisiopatología , Células Piramidales/patología , Área Tegmental Ventral/fisiopatología
17.
Brain Res Bull ; 171: 1-9, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33675933

RESUMEN

Nociception alterations are frequent non-motor symptoms of the prodromal phase of Parkinson's disease (PD). The period for the onset of symptoms and the pathophysiological mechanisms underlying these alterations remain unclear. We investigated the course of nociception alterations in a progressive model of parkinsonism induced by reserpine (RES) in rats. Male Wistar rats (6-7 months) received 5 or 10 subcutaneous injections of RES (0.1 mg/kg) or vehicle daily for 20 days. Motor evaluation and nociceptive assessment were performed throughout the treatment. At the end of the treatment rats were euthanized, the brains removed and processed for immunohistochemical analysis (TH and c-Fos). The RES-treated rats exhibited an increased nociceptive response to mechanical and chemical stimulation in the electronic von Frey and formalin tests, respectively. Moreover, these alterations preceded the motor impairment observed in the catalepsy test. In addition, the RES treatment reduced the TH-immunoreactivity in the ventral tegmental area (VTA) and increased the c-Fos expression in the ventral-lateral periaqueductal gray (vlPAG), rostral ventral medulla (RVM) and dorsal raphe nucleus (DRN) after noxious stimuli induced by formalin. Taken together, our results reinforce that nociceptive changes are one of the early signs of PD and monoamine depletion in basal ganglia can be involved in the abnormal processing of nociceptive information in PD.


Asunto(s)
Núcleo Dorsal del Rafe/metabolismo , Actividad Motora/fisiología , Nocicepción/fisiología , Enfermedad de Parkinson Secundaria/fisiopatología , Sustancia Gris Periacueductal/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Modelos Animales de Enfermedad , Núcleo Dorsal del Rafe/fisiopatología , Masculino , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/metabolismo , Sustancia Gris Periacueductal/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Reserpina , Tirosina 3-Monooxigenasa/metabolismo , Área Tegmental Ventral/fisiopatología
18.
Artículo en Inglés | MEDLINE | ID: mdl-33631251

RESUMEN

Anhedonia is one of the core symptoms of major depressive disorder (MDD), which is often inadequately treated by traditional antidepressants. The modern framework of anhedonia extends the definition from impaired consummatory pleasure or interest in rewards to a broad spectrum of deficits that impact functions such as reward anticipation, approach motivation, effort expenditure, reward valuation, expectation, and reward-cue association learning. Substantial preclinical and clinical research has explored the neural basis of reward deficits in the context of depression, and has implicated mesocorticolimbic reward circuitry comprising the nucleus accumbens, ventral pallidum, ventral tegmental area, amygdala, hippocampus, anterior cingulate, insula, orbitofrontal cortex, and other prefrontal cortex regions. Dopamine modulates several reward facets including anticipation, motivation, effort, and learning. As well, serotonin, norepinephrine, opioids, glutamate, Gamma aminobutyric acid (GABA), and acetylcholine are also involved in anhedonia, and medications targeting these systems may also potentially normalize reward processing in depression. Unfortunately, whereas reward anticipation and reward outcome are extensively explored by both preclinical and clinical studies, translational gaps remain in reward motivation, effort, valuation, and learning, where clinical neuroimaging studies are in the early stages. This review aims to synthesize the neurobiological mechanisms underlying anhedonia in MDD uncovered by preclinical and clinical research. The translational difficulties in studying the neural basis of reward are also discussed.


Asunto(s)
Anhedonia , Depresión/fisiopatología , Motivación , Neurobiología , Neurotransmisores/metabolismo , Recompensa , Dopamina/metabolismo , Humanos , Red Nerviosa , Núcleo Accumbens/fisiopatología , Corteza Prefrontal/fisiopatología , Investigación Biomédica Traslacional , Área Tegmental Ventral/fisiopatología
19.
Mol Neurobiol ; 58(5): 2423-2434, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33428093

RESUMEN

Post-traumatic stress disorder (PTSD) is a debilitating psychiatric condition characterized by intrusive recollections of the traumatic event, avoidance behaviors, hyper-arousal to event-related cues, cognitive disruption, and mood dysregulation. Accumulating preclinical and clinical evidence implicates dysfunction of the ventral tegmental area (VTA) dopaminergic system in PTSD pathogenesis. This article reviews recent advances in our knowledge of the relationship between dopaminergic dyshomeostasis and PTSD, including the contributions of specific dopaminergic gene variants to disease susceptibility, alterations in VTA dopamine neuron activity, dysregulation of dopaminergic transmission, and potential pharmacological and psychological interventions for PTSD targeting the dopaminergic system. An in-depth understanding of PTSD etiology is crucial for the development of innovative risk assessment, diagnostic, and treatment strategies following traumatic events.


Asunto(s)
Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Trastornos por Estrés Postraumático/fisiopatología , Área Tegmental Ventral/fisiopatología , Animales , Homeostasis/fisiología , Humanos , Trastornos por Estrés Postraumático/metabolismo , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Área Tegmental Ventral/metabolismo
20.
J Neurochem ; 157(5): 1572-1584, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33486769

RESUMEN

The actions of dopamine are essential to relapse to drug seeking but we still lack a precise understanding of how dopamine achieves these effects. Here we review recent advances from animal models in understanding how dopamine controls relapse to drug seeking. These advances have been enabled by important developments in understanding the basic neurochemical, molecular, anatomical, physiological and functional properties of the major dopamine pathways in the mammalian brain. The literature shows that although different forms of relapse to seeking different drugs of abuse each depend on dopamine, there are distinct dopamine mechanisms for relapse. Different circuit-level mechanisms, different populations of dopamine neurons and different activity profiles within these dopamine neurons, are important for driving different forms of relapse. This diversity highlights the need to better understand when, where and how dopamine contributes to relapse behaviours.


Asunto(s)
Dopamina/fisiología , Comportamiento de Búsqueda de Drogas/fisiología , Trastornos Relacionados con Sustancias/fisiopatología , Animales , Dopamina/metabolismo , Humanos , Núcleo Accumbens/fisiopatología , Recurrencia , Recompensa , Trastornos Relacionados con Sustancias/metabolismo , Área Tegmental Ventral/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...