Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 320
Filtrar
1.
Transl Psychiatry ; 14(1): 408, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39358336

RESUMEN

Dopamine (DA) neurons play a crucial role in the development and manifestation of depression, as well as in response to antidepressant treatments. While the function of the predominantly distributed DA neurons in the ventral tegmental area (VTA) is well established, the contribution of a small fraction of DA neurons in the dorsal raphe nucleus (DRN) during depression remains unclear. In this study, we found that chronic unpredictable stress (CUS) induces depression-related behaviors and decreases spontaneous firing rates, excitatory and inhibitory postsynaptic currents of DA neurons in the DRN associated with reduced excitatory synaptic transmission in male and female mice. The chemogenetic inhibition of DA neurons in the DRN produces depressive phenotypes. Conversely, their activation completely reversed the anhedonic and despair behaviors induced by CUS. Furthermore, we showed that a DRN dopaminergic projecting to the dorsal bed nucleus of the stria terminalis (dBNST) selectively controls depressive behaviors by influencing the neural activity and N-methyl-D-aspartate receptor (NMDAR) mediating EPSC of calcium/calmodulin-dependent protein kinase II+ (CaMKII+) target neurons by regulating dopamine neurotransmitter and dopamine receptor 2 (DR2) in the dBNST. Overall, these findings highlight the essential role of the DRNDA → dBNSTCaMKII+ neural circuit in bi-directionally mediating stress-induced depression-related behaviors. Our findings indicate that DRN DA neurons are a key component of the neural circuitry involved in regulating depression-related behaviors, making them a potential therapeutic target for depression.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Depresión , Neuronas Dopaminérgicas , Núcleo Dorsal del Rafe , Núcleos Septales , Animales , Neuronas Dopaminérgicas/metabolismo , Núcleo Dorsal del Rafe/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Ratones , Núcleos Septales/metabolismo , Núcleos Septales/fisiopatología , Masculino , Femenino , Depresión/metabolismo , Depresión/fisiopatología , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Ratones Endogámicos C57BL , Conducta Animal , Modelos Animales de Enfermedad , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Dopamina D2/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Transmisión Sináptica/fisiología
2.
Int J Mol Sci ; 25(15)2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39125652

RESUMEN

Methylphenidate (MPD) remains a cornerstone pharmacological intervention for managing ADHD, yet its increasing usage among ordinary youth and adults outside clinical contexts necessitates a thorough investigation into its developmental effects. This study seeks to simultaneously investigate the behavioral and neuronal changes within the dorsal raphe (DR) nucleus, a center of serotonergic neurons in the mammalian brain, before and after the administration of varying doses of acute and chronic MPD in freely behaving young and adult rats implanted with DR recording electrodes. Wireless neuronal and behavioral recording systems were used over 10 consecutive experimental days. Eight groups were examined: saline, 0.6, 2.5, and 10.0 mg/kg MPD for both young and adult rats. Six daily MPD injections were administered on experimental days 1 to 6, followed by a three-day washout period and MPD re-administration on experimental day 10 (ED10). The analysis of neuronal activity recorded from 504 DR neurons (DRNs) in young rats and 356 DRNs in adult rats reveals significant age-dependent differences in acute and chronic MPD responses. This study emphasizes the importance of aligning electrophysiological evaluations with behavioral outcomes following extended MPD exposure, elucidating the critical role of DRNs and serotonin signaling in modulating MPD responses and delineating age-specific variations in young versus adult rat models.


Asunto(s)
Conducta Animal , Núcleo Dorsal del Rafe , Metilfenidato , Serotonina , Animales , Metilfenidato/farmacología , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Ratas , Serotonina/metabolismo , Masculino , Conducta Animal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Edad
3.
Pharmacol Biochem Behav ; 244: 173849, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39142357

RESUMEN

Selective serotonin (5-HT) reuptake inhibitors (SSRIs) are commonly prescribed to women during pregnancy and breastfeeding despite posing a risk of adverse cognitive outcomes and affective disorders for the child. The consequences of SSRI-induced excess of 5-HT during development for the brain neuromodulatory 5-HT system remain largely unexplored. In this study, an SSRI - fluoxetine (FLX) - was administered to C57BL/6 J mouse dams during pregnancy and lactation to assess its effects on the offspring. We found that maternal FLX decreased field potentials, impaired long-term potentiation, facilitated long-term depression and tended to increase the density of 5-HTergic fibers in the medial prefrontal cortex (mPFC) of female but not male adolescent offspring. These effects were accompanied by deteriorated performance in the temporal order memory task and reduced sucrose preference with no change in marble burying behavior in FLX-exposed female offspring. We also found that maternal FLX reduced the axodendritic tree complexity of 5-HT dorsal raphe nucleus (DRN) neurons in female but not male offspring, with no changes in the excitability of DRN neurons of either sex. While no effects of maternal FLX on inhibitory postsynaptic currents (sIPSCs) in DRN neurons were found, we observed a significant influence of FLX exposure on kinetics of spontaneous excitatory postsynaptic currents (sEPSCs) in DRN neurons. Finally, we report that no changes in field potentials and synaptic plasticity were evident in the mPFC of the offspring after maternal exposure during pregnancy and lactation to a new antidepressant, vortioxetine. These findings show that in contrast to the mPFC, long-term consequences of maternal FLX exposure on the structure and function of DRN 5-HT neurons are mild and suggest a sex-dependent, distinct sensitivity of cortical and brainstem neurons to FLX exposure in early life. Vortioxetine appears to exert fewer side effects with regards to the mPFC when compared with FLX.


Asunto(s)
Núcleo Dorsal del Rafe , Fluoxetina , Ratones Endogámicos C57BL , Plasticidad Neuronal , Corteza Prefrontal , Efectos Tardíos de la Exposición Prenatal , Inhibidores Selectivos de la Recaptación de Serotonina , Transmisión Sináptica , Animales , Fluoxetina/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Femenino , Ratones , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Embarazo , Masculino , Plasticidad Neuronal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Neuronas/efectos de los fármacos , Serotonina/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos
4.
Neurosci Lett ; 839: 137933, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39128818

RESUMEN

The dorsal raphe nucleus (DRN) receives dopaminergic inputs from the ventral tegmental area (VTA). Also, the DRN contains a small population of cells that express dopamine (DRNDA neurons). However, the physiological role of dopamine (DA) in the DRN and its interaction with serotonergic (5-HT) neurons is poorly understood. Several works have reported moderate levels of D1, D2, and D3 DA receptors in the DRN. Furthermore, it was found that the activation of D2 receptors increased the firing of putative 5-HT neurons. Other studies have reported that D1 and D2 dopamine receptors can interact with glutamate NMDA receptors, modulating the excitability of different cell types. In the present work, we used immunocytochemical techniques to determine the kind of DA receptors in the DRN. Additionally, we performed electrophysiological experiments in brainstem slices to study the effect of DA agonists on NMDA-elicited currents recorded from identified 5-HT DRN neurons. We found that D2 and D3 but not D1 receptors are present in this nucleus. Also, we demonstrated that the activation of D2-like receptors increases NMDA-elicited currents in 5-HT neurons through a mechanism involving phospholipase C (PLC) and protein kinase C (PKC) enzymes. Possible physiological implications related to the sleep-wake cycle are discussed.


Asunto(s)
Núcleo Dorsal del Rafe , Receptores de Dopamina D2 , Receptores de N-Metil-D-Aspartato , Neuronas Serotoninérgicas , Animales , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Receptores de Dopamina D2/metabolismo , Neuronas Serotoninérgicas/metabolismo , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/fisiología , Masculino , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Dopamina D3/metabolismo , N-Metilaspartato/farmacología , N-Metilaspartato/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D1/agonistas , Agonistas de Dopamina/farmacología , Ratas , Fosfolipasas de Tipo C/metabolismo , Ratas Wistar
5.
Nat Commun ; 15(1): 7603, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39217143

RESUMEN

Resistance to stress is a key determinant for mammalian functioning. While many studies have revealed neural circuits and substrates responsible for initiating and mediating stress responses, little is known about how the brain resists to stress and prevents overreactions. Here, we identified a previously uncharacterized neuropeptide Y (NPY) neuronal population in the dorsal raphe nucleus and ventrolateral periaqueductal gray region (DRN/vlPAG) with anxiolytic effects in male mice. NPYDRN/vlPAG neurons are rapidly activated by various stressful stimuli. Inhibiting these neurons exacerbated hypophagic and anxiety responses during stress, while activation significantly ameliorates acute stress-induced hypophagia and anxiety levels and transmits positive valence. Furthermore, NPYDRN/vlPAG neurons exert differential but synergic anxiolytic effects via inhibitory projections to the paraventricular thalamic nucleus (PVT) and the lateral hypothalamic area (LH). Together, our findings reveal a feedforward inhibition neural mechanism underlying stress resistance and suggest NPYDRN/vlPAG neurons as a potential therapeutic target for stress-related disorders.


Asunto(s)
Neuronas , Neuropéptido Y , Estrés Psicológico , Animales , Masculino , Neuropéptido Y/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Ratones , Estrés Psicológico/fisiopatología , Ratones Endogámicos C57BL , Ansiedad/fisiopatología , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/fisiología , Sustancia Gris Periacueductal/fisiología , Tronco Encefálico/fisiología , Área Hipotalámica Lateral/fisiología , Estrés Fisiológico
6.
Eur J Neurosci ; 60(7): 5658-5670, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39189108

RESUMEN

Fluctuations in estradiol levels at each stage of life in women are considered one of the causes of mental diseases through their effects on the central nervous system. During menopause, a decrease in estradiol levels has been reported to affect the serotonin nervous system and induce depression-like and anxiety symptoms. However, the regulation of brain and behaviour during childhood and adolescence is poorly understood. Moreover, the role of oestrogen receptors α and ß in the regulation of the serotonergic nervous system has been reported, but little is known about the involvement of G protein-coupled receptor 30. Therefore, in this study, we used an ovariectomized childhood mouse model to analyse behaviour and investigate the effects on the serotonin nervous system. We showed that ovariectomy surgery at 4 weeks of age, which is the weaning period, induced a decrease in spontaneous locomotor activity during the active period and a preference for novel mice over familiar mice in the three-chamber social test at 10 weeks of age. In addition, the administration of G-1, a protein-coupled receptor 30 agonist, to ovariectomized mice suppressed spontaneous locomotor activity and the preference for novel mice. Furthermore, we demonstrated that childhood ovariectomy induces increased tryptophan hydroxylase gene expression in the raphe nucleus and increased serotonin release in the amygdaloid nucleus, and administration of G-1 ameliorated these effects. Our study suggests that G protein-coupled receptor 30-mediated regulation of serotonin synthesis is involved in changes in activity and social-cognitive behaviour due to decreased estradiol levels during childhood.


Asunto(s)
Ovariectomía , Receptores Acoplados a Proteínas G , Serotonina , Triptófano Hidroxilasa , Animales , Femenino , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Ratones , Serotonina/metabolismo , Triptófano Hidroxilasa/metabolismo , Triptófano Hidroxilasa/genética , Conducta Animal/fisiología , Receptores de Estrógenos/metabolismo , Estradiol/farmacología , Estradiol/metabolismo , Ratones Endogámicos C57BL , Conducta Social , Quinolinas/farmacología , Neuronas Serotoninérgicas/metabolismo , Neuronas Serotoninérgicas/fisiología , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Locomoción/fisiología , Locomoción/efectos de los fármacos , Actividad Motora/fisiología
7.
Neuropharmacology ; 261: 110132, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39208980

RESUMEN

Selective serotonin (5-HT) reuptake inhibitors (SSRIs) like fluoxetine remain a first-line treatment for major depression, but are effective in less than half of patients and can take 4-8 weeks to show results. In this study, we examined cF1ko mice with genetically induced upregulation of 5-HT1A autoreceptors that reduces 5-HT neuronal activity. These mice display anxiety- and depression-related behaviors that did not respond to chronic fluoxetine treatment. We examined treatment with NLX-101, a biased agonist that preferentially targets 5-HT1A heteroreceptors. By testing different doses of NLX-101, we found that a dose of 0.2 mg/kg was effective in reducing depression-related behavior in cF1ko mice without causing hypothermia, a 5-HT1A autoreceptor-mediated response. After 1 h, this dose activated dorsal raphe 5-HT neurons and cells in the medial prefrontal cortex (mPFC), increasing nuclear c-fos labelling in cF1ko mice. In cF1ko mice but not wild-type littermates, 0.2 mg/kg NLX-101 administered 1 h prior to each behavioral test for two weeks reduced depressive behavior in the forced swim test, but increased anxiety-related behaviors in the open field, elevated plus maze, and novelty suppressed feeding tests. During this treatment, NLX-101 induced widespread increases in the density of 5-HT axons, varicosities, and especially synaptic and triadic structures, particularly in depression-related brain regions including mPFC, hippocampal CA1 and CA2/3, amygdala and nucleus accumbens of cF1ko mice. Overall, NLX-101 was rapid and effective in reducing depressive behavior in SSRI-resistant mice, but also induced anxiety-related behaviors. The increase in serotonin innervation induced by intermittent NLX-101 may contribute to its behavioral actions.


Asunto(s)
Fluoxetina , Ratones Noqueados , Receptor de Serotonina 5-HT1A , Animales , Fluoxetina/farmacología , Ratones , Masculino , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT1A/genética , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Piridinas/farmacología , Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Serotonina/metabolismo , Neuronas Serotoninérgicas/efectos de los fármacos , Ratones Endogámicos C57BL , Relación Dosis-Respuesta a Droga , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Ansiedad/tratamiento farmacológico , Piperazinas/farmacología , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Piperidinas , Pirimidinas
8.
Eur J Pharmacol ; 981: 176918, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39159717

RESUMEN

Maladaptive reactive aggression is a core symptom of neuropsychiatric disorders such as schizophrenia. While uncontrolled aggression dampens societal safety, there is a limited understanding of the neural regulation involved in reactive aggression and its treatment. High levels of aggression have been linked to low serotonin (5-HT) levels. Additionally, post-weaning socially isolated (SI) mice exhibit outbursts of aggression following encountering acute stress, and hyperactivated ventral hippocampus (vHip) involves this stress-provoked escalated aggression. Here, we investigated the potential role of the raphe nucleus projecting to the vHip in modulating aggressive behavior. Chemogenetically activating the dorsal raphe nucleus (DRN) soma projecting the vHip or DRN nerve terminals in the vHip reduced reactive aggression. The reduction of attack behavior was abolished by the pretreatment of 5-HT1B receptor antagonist SB-224289. However, activating the median raphe nucleus (MRN)-to-vHip pathway ameliorated depression-like behavior but did not affect reactive aggression. DRN→vHip activation suppressed the vHip downstream area, the ventromedial hypothalamus (VMH), which is a core aggression area. Intra-vHip infusion of 5-HT1B receptor agonists (anpirtoline, CP-93129) suppressed reactive aggression and decreased c-Fos levels in the vHip neurons projecting to the VMH, suggesting an inhibition mechanism. Our findings indicate that activating the DRN projecting to the vHip is sufficient to inhibit reactive aggression in a 5-HT1B receptor-dependent manner. Thus, targeting 5-HT1B receptor could serve as a promising therapeutic approach to ameliorate symptoms of reactive aggression.


Asunto(s)
Agresión , Núcleo Dorsal del Rafe , Hipocampo , Receptor de Serotonina 5-HT1B , Animales , Agresión/efectos de los fármacos , Agresión/fisiología , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Receptor de Serotonina 5-HT1B/metabolismo , Masculino , Ratones , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-fos/metabolismo
9.
Commun Biol ; 7(1): 998, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39147805

RESUMEN

Affective disorders are frequently associated with disrupted circadian rhythms. The existence of rhythmic secretion of central serotonin (5-hydroxytryptamine, 5-HT) pattern has been reported; however, the functional mechanism underlying the circadian control of 5-HTergic mood regulation remains largely unknown. Here, we investigate the role of the circadian nuclear receptor REV-ERBα in regulating tryptophan hydroxylase 2 (Tph2), the rate-limiting enzyme of 5-HT synthesis. We demonstrate that the REV-ERBα expressed in dorsal raphe (DR) 5-HTergic neurons functionally competes with PET-1-a nuclear activator crucial for 5-HTergic neuron development. In mice, genetic ablation of DR 5-HTergic REV-ERBα increases Tph2 expression, leading to elevated DR 5-HT levels and reduced depression-like behaviors at dusk. Further, pharmacological manipulation of the mice DR REV-ERBα activity increases DR 5-HT levels and affects despair-related behaviors. Our findings provide valuable insights into the molecular and cellular link between the circadian rhythm and the mood-controlling DR 5-HTergic systems.


Asunto(s)
Ritmo Circadiano , Núcleo Dorsal del Rafe , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares , Serotonina , Triptófano Hidroxilasa , Animales , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Núcleo Dorsal del Rafe/metabolismo , Serotonina/metabolismo , Serotonina/biosíntesis , Triptófano Hidroxilasa/metabolismo , Triptófano Hidroxilasa/genética , Ratones , Masculino , Afecto/fisiología , Ratones Noqueados , Ratones Endogámicos C57BL , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Depresión/metabolismo
10.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3828-3836, 2024 Jul.
Artículo en Chino | MEDLINE | ID: mdl-39099356

RESUMEN

This study aims to further elucidate the efficacy targets of celastrol(CEL) intervention in central inflammation in mice with obesity-depression comorbiditiy, based on the differential mRNA expression in the amygdala(AMY) and dorsal raphe nucleus(DRN) after CEL intervention. C57BL/6J mice were randomly divided into a normal diet group(Chow), a obesity-depression comorbidity(COM) group, and low-, medium-, and high-dose CEL groups(CEL-L, CEL-M, CEL-H, 0.5, 1.0, 2.0 mg·kg~(-1)). The Chow group received a normal diet, while the COM group and CEL-L, CEL-M, CEL-H groups received a high-fat diet combined with chronic stress from wet bedding. After 10 weeks of feeding, the mice were orally administered CEL for three weeks. Subsequently, the AMY and DRN of mice in the Chow, COM, and CEL-H groups were subjected to transcriptome analysis, and the intersection of target differentially expressed genes in both nuclei was visualized using a Venn diagram. The intersected genes were then imported into STRING for protein-protein interaction(PPI) analysis, and Gene Ontology(GO) analysis was performed using DAVID to identify the core targets regulated by CEL in the AMY and DRN. Independent samples were subjected to quantitative real-time PCR(qPCR) to validate the intersection genes. The results revealed that the common genes regulated by CEL in the AMY and DRN included chemokine family genes Ccl2, Ccl5, Ccl7, Cxcl10, Cxcr6, and Hsp70 family genes Hspa1a, Hspa1b, as well as Myd88, Il2ra, Irf7, Slc17a8, Drd2, Parp9, and Nampt. GO analysis showed that the top 5 nodes Ccl2, Cxcl10, Myd88, Ccl5, and Irf7 were all involved in immune-inflammation regulation(P<0.01). The qPCR results from independent samples showed that in the AMY, compared with the results in the Chow group, chemokine family genes, Hsp70, Myd88, Il2ra, Irf7, Slc17a8, Parp9, and Nampt were significantly up-regulated in the COM group, with Drd2 showing a decreasing trend; these pathological changes were significantly improved in the CEL-H group compared to the COM group. In the DRN, compared with the results in the Chow group, chemokine family genes, Hsp70, Myd88, Il2ra, Irf7, Parp9, and Nampt were significantly down-regulated, while Slc17a8 was significantly up-regulated in the COM group; compared with those in the COM group, Cxcr6, Irf7, and Drd2 were significantly up-regulated, while Slc17a8 was significantly down-regulated in the CEL-H group. In both the AMY and DRN, the expression of Irf7 by CEL showed both inhibition and activation in a dose-dependent manner(R~2 were 0.709 8 and 0.917 2, respectively). These findings suggest that CEL can effectively improve neuroinflammation by regulating bidirectional expression of the same target proteins, thereby intervening in the immune activation of the AMY and immune suppression of the DRN in COM mice.


Asunto(s)
Amígdala del Cerebelo , Depresión , Núcleo Dorsal del Rafe , Ratones Endogámicos C57BL , Obesidad , Triterpenos Pentacíclicos , Triterpenos , Animales , Ratones , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Masculino , Depresión/tratamiento farmacológico , Depresión/genética , Depresión/metabolismo , Obesidad/genética , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Triterpenos/farmacología , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/genética , Humanos
11.
Behav Brain Res ; 472: 115147, 2024 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-39029628

RESUMEN

Early life adversity has been linked with a higher probability of developing behavioral impairments and environmental manipulation is a strategy that may reduce the negative effects of exposure to adversity in early life. Here, we focused on exploring the influence of environmental enrichment (EE) as a protective factor in the context of early life adversity. We hypothesized that 24 hours of maternal deprivation (MD), in the second week of life, could induce anxiety-like behavior alterations and that exposure to EE could induce resilience to these behaviors due to alterations in the serotonergic system. Male Wistar rats were exposed to MD, on postnatal days 11 and 13, and to EE, after weaning. In adulthood, we performed a series of behavioral tests for fear, anxiety, and locomotor activity. We also measured the levels of serotonin in the amygdala and dorsal raphe nucleus. Our results revealed that MD does not impact fear behavior or the levels of serotonin, while EE decreases locomotor activity in a novel environment and enhances exploration in the predator odor test. EE also decreases serotonin in the amygdala and increases its turnover rate levels. Our findings provide insights into the critical timeframe during which stress exposure impacts the development and confirm that exposure to EE has an independent and protective effect for anxiety-like behaviors later in life.


Asunto(s)
Experiencias Adversas de la Infancia , Emociones , Ambiente , Experiencias Adversas de la Infancia/psicología , Ansiedad/psicología , Privación Materna , Masculino , Femenino , Animales , Ratas , Actividad Motora , Conducta Exploratoria , Memoria , Núcleo Dorsal del Rafe/metabolismo , Amígdala del Cerebelo/metabolismo , Serotonina/metabolismo , Miedo/psicología
12.
Neuropharmacology ; 258: 110068, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-38996832

RESUMEN

Birth stress is a risk factor for psychiatric disorders and associated with exaggerated release of the stress hormone arginine vasopressin (AVP) into circulation and in the brain. In perinatal hippocampus, AVP activates GABAergic interneurons which leads to suppression of spontaneous network events and suggests a protective function of AVP on cortical networks during birth. However, the role of AVP in developing subcortical networks is not known. Here we tested the effect of AVP on the dorsal raphe nucleus (DRN) 5-hydroxytryptamine (5-HT, serotonin) system in male and female neonatal rats, since early 5-HT homeostasis is critical for the development of cortical brain regions and emotional behaviors. We show that AVP is strongly excitatory in neonatal DRN: it increases excitatory synaptic inputs of 5-HT neurons via V1A receptors in vitro and promotes their action potential firing through a combination of its effect on glutamatergic synaptic transmission and a direct effect on the excitability of these neurons. Furthermore, we identified two major firing patterns of neonatal 5-HT neurons in vivo, tonic regular firing and low frequency oscillations of regular spike trains and confirmed that these neurons are also activated by AVP in vivo. Finally, we show that the sparse vasopressinergic innervation in neonatal DRN originates exclusively from cell groups in medial amygdala and bed nucleus of stria terminalis. Hyperactivation of the neonatal 5-HT system by AVP during birth stress may impact its own functional development and affect the maturation of cortical target regions, which may increase the risk for psychiatric conditions later on.


Asunto(s)
Animales Recién Nacidos , Arginina Vasopresina , Núcleo Dorsal del Rafe , Neuronas Serotoninérgicas , Animales , Arginina Vasopresina/metabolismo , Arginina Vasopresina/farmacología , Femenino , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/fisiología , Masculino , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/fisiología , Ratas , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Ratas Sprague-Dawley , Serotonina/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Receptores de Vasopresinas/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
13.
Transl Neurodegener ; 13(1): 34, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39044270

RESUMEN

BACKGROUND: Depressive symptoms often occur in patients with Alzheimer's disease (AD) and exacerbate the pathogenesis of AD. However, the neural circuit mechanisms underlying the AD-associated depression remain unclear. The serotonergic system plays crucial roles in both AD and depression. METHODS: We used a combination of in vivo trans-synaptic circuit-dissecting anatomical approaches, chemogenetic manipulations, optogenetic manipulations, pharmacological methods, behavioral testing, and electrophysiological recording to investigate dorsal raphe nucleus serotonergic circuit in AD-associated depression in AD mouse model. RESULTS: We found that the activity of dorsal raphe nucleus serotonin neurons (DRN5-HT) and their projections to the dorsal hippocampal CA1 (dCA1) terminals (DRN5-HT-dCA1CaMKII) both decreased in brains of early 5×FAD mice. Chemogenetic or optogenetic activation of the DRN5-HT-dCA1CaMKII neural circuit attenuated the depressive symptoms and cognitive impairments in 5×FAD mice through serotonin receptor 1B (5-HT1BR) and 4 (5-HT4R). Pharmacological activation of 5-HT1BR or 5-HT4R attenuated the depressive symptoms and cognitive impairments in 5×FAD mice by regulating the DRN5-HT-dCA1CaMKII neural circuit to improve synaptic plasticity. CONCLUSIONS: These findings provide a new mechanistic connection between depression and AD and provide potential pharmaceutical prevention targets for AD.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Depresión , Modelos Animales de Enfermedad , Núcleo Dorsal del Rafe , Ratones Transgénicos , Neuronas Serotoninérgicas , Animales , Núcleo Dorsal del Rafe/metabolismo , Masculino , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/psicología , Disfunción Cognitiva/fisiopatología , Ratones , Neuronas Serotoninérgicas/metabolismo , Neuronas Serotoninérgicas/fisiología , Depresión/metabolismo , Depresión/genética , Depresión/psicología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Hipocampo/metabolismo , Serotonina/metabolismo , Optogenética , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología
14.
Nature ; 632(8023): 147-156, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39020173

RESUMEN

Changes in the amount of daylight (photoperiod) alter physiology and behaviour1,2. Adaptive responses to seasonal photoperiods are vital to all organisms-dysregulation associates with disease, including affective disorders3 and metabolic syndromes4. The circadian rhythm circuitry is implicated in such responses5,6, yet little is known about the precise cellular substrates that underlie phase synchronization to photoperiod change. Here we identify a brain circuit and system of axon branch-specific and reversible neurotransmitter deployment that are critical for behavioural and sleep adaptation to photoperiod. A type of neuron called mrEn1-Pet17 in the mouse brainstem median raphe nucleus segregates serotonin from VGLUT3 (also known as SLC17A8, a proxy for glutamate) to different axonal branches that innervate specific brain regions involved in circadian rhythm and sleep-wake timing8,9. This branch-specific neurotransmitter deployment did not distinguish between daylight and dark phase; however, it reorganized with change in photoperiod. Axonal boutons, but not cell soma, changed neurochemical phenotype upon a shift away from equinox light/dark conditions, and these changes were reversed upon return to equinox conditions. When we genetically disabled Vglut3 in mrEn1-Pet1 neurons, sleep-wake periods, voluntary activity and clock gene expression did not synchronize to the new photoperiod or were delayed. Combining intersectional rabies virus tracing and projection-specific neuronal silencing, we delineated a preoptic area-to-mrEn1Pet1 connection that was responsible for decoding the photoperiodic inputs, driving the neurotransmitter reorganization and promoting behavioural synchronization. Our results reveal a brain circuit and periodic, branch-specific neurotransmitter deployment that regulates organismal adaptation to photoperiod change.


Asunto(s)
Adaptación Fisiológica , Axones , Ritmo Circadiano , Neurotransmisores , Fotoperiodo , Animales , Femenino , Ratones , Adaptación Fisiológica/fisiología , Sistemas de Transporte de Aminoácidos Acídicos/deficiencia , Sistemas de Transporte de Aminoácidos Acídicos/genética , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Axones/metabolismo , Axones/fisiología , Ritmo Circadiano/fisiología , Proteínas CLOCK/genética , Oscuridad , Núcleo Dorsal del Rafe/citología , Núcleo Dorsal del Rafe/metabolismo , Vías Nerviosas/fisiología , Neurotransmisores/metabolismo , Área Preóptica/citología , Área Preóptica/metabolismo , Terminales Presinápticos/metabolismo , Terminales Presinápticos/fisiología , Virus de la Rabia , Serotonina/metabolismo , Sueño/fisiología , Vigilia/fisiología
15.
Neuropharmacology ; 258: 110055, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-38950692

RESUMEN

Sleep disturbances and persistent pain conditions are public health challenges worldwide. Although it is well-known that sleep deficit increases pain sensitivity, the underlying mechanisms remain elusive. We have recently demonstrated the involvement of nucleus accumbens (NAc) and anterior cingulate cortex (ACC) in the pronociceptive effect of sleep restriction. In this study, we found that sleep restriction increases c-Fos expression in NAc and ACC, suggesting hyperactivation of these regions during prolonged wakefulness in male Wistar rats. Blocking adenosine A2A receptors in the NAc or GABAA receptors in the ventral tegmental area (VTA), dorsal raphe nucleus (DRN), or locus coeruleus (LC) effectively mitigated the pronociceptive effect of sleep restriction. In contrast, the blockade of GABAA receptors in each of these nuclei only transiently reduced carrageenan-induced hyperalgesia. Pharmacological activation of dopamine D2, serotonin 5-HT1A and noradrenaline alpha-2 receptors within the ACC also prevented the pronociceptive effect of sleep restriction. While pharmacological inhibition of these same monoaminergic receptors in the ACC restored the pronociceptive effect which had been prevented by the GABAergic disinhibition of the of the VTA, DRN or LC. Overall, these findings suggest that the pronociceptive effect of sleep restriction relies on increased adenosinergic activity on NAc, heightened GABAergic activity in VTA, DRN, and LC, and reduced inhibitory monoaminergic activity on ACC. These findings advance our understanding of the interplay between sleep and pain, shedding light on potential NAc-brainstem-ACC mechanisms that could mediate increased pain sensitivity under conditions of sleep impairment.


Asunto(s)
Núcleo Accumbens , Ratas Wistar , Privación de Sueño , Área Tegmental Ventral , Animales , Masculino , Privación de Sueño/metabolismo , Privación de Sueño/fisiopatología , Ratas , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/efectos de los fármacos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efectos de los fármacos , Receptor de Adenosina A2A/metabolismo , Hiperalgesia/metabolismo , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Giro del Cíngulo/metabolismo , Giro del Cíngulo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Tronco Encefálico/metabolismo , Tronco Encefálico/efectos de los fármacos , Locus Coeruleus/metabolismo , Locus Coeruleus/efectos de los fármacos , Carragenina , Receptores de GABA-A/metabolismo , Receptores de Dopamina D2/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología
16.
Acta Physiol (Oxf) ; 240(9): e14198, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38958443

RESUMEN

AIM: Neural activity in the olfactory bulb (OB) can represent odor information during different brain and behavioral states. For example, the odor responses of mitral/tufted (M/T) cells in the OB change during learning of odor-discrimination tasks and, at the network level, beta power increases and the high gamma (HG) power decreases during odor presentation in such tasks. However, the neural mechanisms underlying these observations remain poorly understood. Here, we investigate whether serotonergic modulation from the dorsal raphe nucleus (DRN) to the OB is involved in shaping activity during the learning process in a go/no-go task in mice. METHODS: Fiber photometry was used to record the population activity of DRN serotonergic neurons during a go/no-go task. In vivo electrophysiology was used to record neural activity (single units and local field potentials) in the OB during the go/no-go task. Real-time place preference (RTPP) and intracranial light administration in a specific subarea (iClass) tests were used to assess the ability of mice to encoding reward information. RESULTS: Odor-evoked population activity in serotonergic neurons in the DRN was shaped during the learning process in a go/no-go task. In the OB, neural activity from oscillations to single cells showed complex, learning-associated changes and ability to encode information during an odor discrimination task. However, these properties were not observed after ablation of DRN serotonergic neurons. CONCLUSION: The activity of neural networks and single cells in the OB, and their ability to encode information about odor value, are shaped by serotonergic projections from the DRN.


Asunto(s)
Núcleo Dorsal del Rafe , Odorantes , Bulbo Olfatorio , Neuronas Serotoninérgicas , Animales , Bulbo Olfatorio/fisiología , Núcleo Dorsal del Rafe/fisiología , Núcleo Dorsal del Rafe/metabolismo , Ratones , Masculino , Neuronas Serotoninérgicas/fisiología , Ratones Endogámicos C57BL , Aprendizaje/fisiología , Serotonina/metabolismo , Olfato/fisiología
17.
Nat Commun ; 15(1): 5042, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871707

RESUMEN

Mood disorders are an enigmatic class of debilitating illnesses that affect millions of individuals worldwide. While chronic stress clearly increases incidence levels of mood disorders, including major depressive disorder (MDD), stress-mediated disruptions in brain function that precipitate these illnesses remain largely elusive. Serotonin-associated antidepressants (ADs) remain the first line of therapy for many with depressive symptoms, yet low remission rates and delays between treatment and symptomatic alleviation have prompted skepticism regarding direct roles for serotonin in the precipitation and treatment of affective disorders. Our group recently demonstrated that serotonin epigenetically modifies histone proteins (H3K4me3Q5ser) to regulate transcriptional permissiveness in brain. However, this non-canonical phenomenon has not yet been explored following stress and/or AD exposures. Here, we employed a combination of genome-wide and biochemical analyses in dorsal raphe nucleus (DRN) of male and female mice exposed to chronic social defeat stress, as well as in DRN of human MDD patients, to examine the impact of stress exposures/MDD diagnosis on H3K4me3Q5ser dynamics, as well as associations between the mark and depression-related gene expression. We additionally assessed stress-induced/MDD-associated regulation of H3K4me3Q5ser following AD exposures, and employed viral-mediated gene therapy in mice to reduce H3K4me3Q5ser levels in DRN and examine its impact on stress-associated gene expression and behavior. We found that H3K4me3Q5ser plays important roles in stress-mediated transcriptional plasticity. Chronically stressed mice displayed dysregulated H3K4me3Q5ser dynamics in DRN, with both AD- and viral-mediated disruption of these dynamics proving sufficient to attenuate stress-mediated gene expression and behavior. Corresponding patterns of H3K4me3Q5ser regulation were observed in MDD subjects on vs. off ADs at their time of death. These findings thus establish a neurotransmission-independent role for serotonin in stress-/AD-associated transcriptional and behavioral plasticity, observations of which may be of clinical relevance to human MDD and its treatment.


Asunto(s)
Antidepresivos , Trastorno Depresivo Mayor , Núcleo Dorsal del Rafe , Histonas , Estrés Psicológico , Animales , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Histonas/metabolismo , Masculino , Femenino , Estrés Psicológico/metabolismo , Humanos , Antidepresivos/farmacología , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/tratamiento farmacológico , Ratones , Serotonina/metabolismo , Ratones Endogámicos C57BL , Epigénesis Genética/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Derrota Social
18.
Int J Mol Sci ; 25(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892125

RESUMEN

A total of 3102 neurons were recorded before and following acute and chronic methylphenidate (MPD) administration. Acute MPD exposure elicits mainly increases in neuronal and behavioral activity in dose-response characteristics. The response to chronic MPD exposure, as compared to acute 0.6, 2.5, or 10.0 mg/kg MPD administration, elicits electrophysiological and behavioral sensitization in some animals and electrophysiological and behavioral tolerance in others when the neuronal recording evaluations were performed based on the animals' behavioral responses, or amount of locomotor activity, to chronic MPD exposure. The majority of neurons recorded from those expressing behavioral sensitization responded to chronic MPD with further increases in firing rate as compared to the initial MPD responses. The majority of neurons recorded from animals expressing behavioral tolerance responded to chronic MPD with decreases in their firing rate as compared to the initial MPD exposures. Each of the six brain areas studied-the ventral tegmental area, locus coeruleus, dorsal raphe, nucleus accumbens, prefrontal cortex, and caudate nucleus (VTA, LC, DR, NAc, PFC, and CN)-responds significantly (p < 0.001) differently to MPD, suggesting that each one of the above brain areas exhibits different roles in the response to MPD. Moreover, this study demonstrates that it is essential to evaluate neuronal activity responses to psychostimulants based on the animals' behavioral responses to acute and chronic effects of the drug from several brain areas simultaneously to obtain accurate information on each area's role in response to the drug.


Asunto(s)
Conducta Animal , Núcleo Caudado , Metilfenidato , Neuronas , Núcleo Accumbens , Corteza Prefrontal , Área Tegmental Ventral , Animales , Metilfenidato/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Ratas , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuronas/metabolismo , Núcleo Caudado/efectos de los fármacos , Núcleo Caudado/fisiología , Núcleo Caudado/metabolismo , Masculino , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/fisiología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiología , Conducta Animal/efectos de los fármacos , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/fisiología , Ratas Sprague-Dawley , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/fisiología , Núcleo Dorsal del Rafe/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología
19.
Biomed Pharmacother ; 176: 116937, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38870632

RESUMEN

The advent of general anesthesia (GA) has significant implications for clinical practice. However, the exact mechanisms underlying GA-induced transitions in consciousness remain elusive. Given some similarities between GA and sleep, the sleep-arousal neural nuclei and circuits involved in sleep-arousal, including the 5-HTergic system, could be implicated in GA. Herein, we utilized pharmacology, optogenetics, chemogenetics, fiber photometry, and retrograde tracing to demonstrate that both endogenous and exogenous activation of the 5-HTergic neural circuit between the dorsal raphe nucleus (DR) and basolateral amygdala (BLA) promotes arousal and facilitates recovery of consciousness from sevoflurane anesthesia. Notably, the 5-HT1A receptor within this pathway holds a pivotal role. Our findings will be conducive to substantially expanding our comprehension of the neural circuit mechanisms underlying sevoflurane anesthesia and provide a potential target for modulating consciousness, ultimately leading to a reduction in anesthetic dose requirements and side effects.


Asunto(s)
Anestésicos por Inhalación , Complejo Nuclear Basolateral , Estado de Conciencia , Núcleo Dorsal del Rafe , Sevoflurano , Sevoflurano/farmacología , Animales , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Estado de Conciencia/efectos de los fármacos , Anestésicos por Inhalación/farmacología , Complejo Nuclear Basolateral/efectos de los fármacos , Complejo Nuclear Basolateral/metabolismo , Complejo Nuclear Basolateral/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Serotonina/metabolismo , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Receptor de Serotonina 5-HT1A/metabolismo , Optogenética
20.
Int J Mol Sci ; 25(11)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38891901

RESUMEN

The diverse effects of serotonin on cognition may emerge from the modulation of large-scale brain networks that support distinct cognitive processes. Yet, the specific effect of serotoninergic modulation on the properties of these networks remains elusive. Here, we used a simultaneous PET-fMRI scanner combined with graph theory analyses to investigate the modulation of network properties by the Serotonin Transporter (SERT) availability measured in the dorsal raphe nucleus (DRN). We defined global efficiency as the average mean of efficiencies over all pairs of distinct nodes of specific brain networks, and determined whether SERT levels correlated with the global efficiency of each network. SERT availability in the DRN correlated negatively with the global efficiency of the executive control brain network, which is engaged in cognitive control and directed attention. No relationship was observed between SERT availability and the global efficiency of the default mode or the salience brain networks. These findings indicate a specific role of serotoninergic modulation in the executive control brain network via a change in its global efficiency.


Asunto(s)
Encéfalo , Función Ejecutiva , Imagen por Resonancia Magnética , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Humanos , Masculino , Función Ejecutiva/fisiología , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Adulto , Red Nerviosa/metabolismo , Red Nerviosa/fisiología , Femenino , Tomografía de Emisión de Positrones/métodos , Serotonina/metabolismo , Adulto Joven , Núcleo Dorsal del Rafe/metabolismo , Mapeo Encefálico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA