Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.595
Filtrar
1.
Transl Psychiatry ; 14(1): 426, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39379355

RESUMEN

Importin α4, which is encoded by the Kpna4 gene, is a well-characterized nuclear-cytoplasmic transport factor known to mediate transport of transcription factors including NF-κB. Here, we report that Kpna4 knock-out (KO) mice exhibit psychiatric disorder-related behavioral abnormalities such as anxiety-related behaviors, decreased social interaction, and sensorimotor gating deficits. Contrary to a previous study predicting attenuated NF-κB activity as a result of Kpna4 deficiency, we observed a significant increase in expression levels of NF-κB genes and proinflammatory cytokines such as TNFα, Il-1ß or Il-6 in the prefrontal cortex or basolateral amygdala of the KO mice. Moreover, examination of inflammatory responses in primary cells revealed that Kpna4 deficient cells have an increased inflammatory response, which was rescued by addition of not only full length, but also a nuclear transport-deficient truncation mutant of importin α4, suggesting contribution of its non-transport functions. Furthermore, RNAseq of sorted adult microglia and astrocytes and subsequent transcription factor analysis suggested increases in polycomb repressor complex 2 (PRC2) activity in Kpna4 KO cells. Taken together, importin α4 deficiency induces psychiatric disorder-related behavioral deficits in mice, along with an increased inflammatory response and possible alteration of PRC2 activity in glial cells.


Asunto(s)
Conducta Animal , Ratones Noqueados , Enfermedades Neuroinflamatorias , alfa Carioferinas , Animales , Ratones , Enfermedades Neuroinflamatorias/metabolismo , alfa Carioferinas/genética , alfa Carioferinas/metabolismo , Conducta Animal/fisiología , Masculino , Ansiedad/genética , Ansiedad/metabolismo , Corteza Prefrontal/metabolismo , Microglía/metabolismo , FN-kappa B/metabolismo , Citocinas/metabolismo , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Astrocitos/metabolismo , Complejo Nuclear Basolateral/metabolismo
2.
Transl Psychiatry ; 14(1): 418, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39368965

RESUMEN

The basolateral amygdala (BLA) is increasingly recognized as a key regulator of depression and anxiety-like behaviors. However, the specific contribution of individual BLA neurons to these behaviors remains poorly understood. Building on our previous study, which demonstrated increased activity in glutamatergic BLA neurons in response to aversive stimuli and that enhancing inhibition in the BLA can alleviate depressive-like behaviors, we investigated the role of individual BLA GABAergic neurons (BLAGABA) in depressive and anxiety-like phenotypes. To address this question, we employed a comprehensive array of techniques, including c-fos staining, fiber photometry recording, optogenetic and chemogenetic manipulation, and behavior analysis. Our findings indicate that BLAGABA neurons show decreased activity during tail suspension and after chronic social defeat stress (CSDS) during social interaction. High-frequency activation of BLAGABA neurons attenuated depressive and anxiety-like behaviors, while low-frequency activation had no effect. Fiber photometry recordings revealed increased activity in BLA GABAergic neurons expressing somatostatin (SST), parvalbumin (PV), and cholecystokinin (CCK) during footshock aversive stimuli. Moreover, we found increased activity in PV and SST neurons and decreased activity in CCK-GABA neurons in the BLA during tail suspension stress. However, after CSDS, BLAPV neurons displayed decreased activity, while SST and CCK neurons showed no changes during the social interaction test. Behavioral analysis demonstrated that chemogenetic inhibition of PV and CCK-GABA neurons induced depressive and anxiety-like behaviors. whereas SST neuron inhibition had no effect. Conversely, chemogenetic activation of BLAPV neurons alleviated depressive behaviors, and activation of BLACCK-GABA neurons alleviated at least partly both depressive and anxiety-like behaviors. This study provides compelling evidence that BLAPV neurons play a critical role in regulating depressive-like behaviors, and that BLACCK-GABA neurons are involved, at least in part, in modulating both depressive-like and anxiety-like behaviors in mice.


Asunto(s)
Ansiedad , Complejo Nuclear Basolateral , Conducta Animal , Colecistoquinina , Depresión , Neuronas GABAérgicas , Parvalbúminas , Animales , Neuronas GABAérgicas/metabolismo , Colecistoquinina/metabolismo , Parvalbúminas/metabolismo , Complejo Nuclear Basolateral/metabolismo , Ratones , Ansiedad/metabolismo , Ansiedad/fisiopatología , Depresión/metabolismo , Masculino , Optogenética , Ratones Endogámicos C57BL , Somatostatina/metabolismo , Estrés Psicológico/metabolismo , Derrota Social , Modelos Animales de Enfermedad , Suspensión Trasera , Interacción Social
3.
Commun Biol ; 7(1): 1199, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39341879

RESUMEN

Chronic stress is associated with anxiety and cognitive impairment. Repeated social defeat (RSD) in mice induces anxiety-like behavior driven by microglia and the recruitment of inflammatory monocytes to the brain. Nonetheless, it is unclear how microglia communicate with other cells to modulate the physiological and behavioral responses to stress. Using single-cell (sc)RNAseq, we identify novel, to the best of our knowledge, stress-associated microglia in the hippocampus defined by RNA profiles of cytokine/chemokine signaling, cellular stress, and phagocytosis. Microglia depletion with a CSF1R antagonist (PLX5622) attenuates the stress-associated profile of leukocytes, endothelia, and astrocytes. Furthermore, RSD-induced social withdrawal and cognitive impairment are microglia-dependent, but social avoidance is microglia-independent. Furthermore, single-nuclei (sn)RNAseq shows robust responses to RSD in hippocampal neurons that are both microglia-dependent and independent. Notably, stress-induced CREB, oxytocin, and glutamatergic signaling in neurons are microglia-dependent. Collectively, these stress-associated microglia influence transcriptional profiles in the hippocampus related to social and cognitive deficits.


Asunto(s)
Disfunción Cognitiva , Hipocampo , Ratones Endogámicos C57BL , Microglía , Derrota Social , Animales , Microglía/metabolismo , Ratones , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/genética , Disfunción Cognitiva/etiología , Masculino , Hipocampo/metabolismo , Estrés Psicológico/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Transcriptoma , Conducta Social , Ansiedad/metabolismo , Compuestos Orgánicos
4.
Front Cell Infect Microbiol ; 14: 1421791, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39301289

RESUMEN

The role of the gut microbiota in the gut-brain axis has attracted attention in recent years. Some gut microbiota produces γ-aminobutyric acid (GABA), a major inhibitory neurotransmitter in mammals, in vitro, but the correlation between gut microbiota composition and intestinal GABA concentration, as well as the action of intestinal GABA in vivo, are poorly understood. Herein, we found that the intestinal GABA concentration was increased in mice by the intervention of the gut microbiota with neomycin or Bifidobacterium bifidum TMC3115 (TMC3115). Administration of TMC3115 reduced anxiety without affecting serum levels of serotonin, corticosterone, or GABA. We further found that intestinal epithelial cells expressed GABA receptor subunits and mediated mitogen-activated protein kinase signaling upon GABA stimulation. In addition, administration of TMC3115 induced mitogen-activated protein kinase signaling in colonic epithelial cells but not in small intestinal epithelial cells in mice. These results indicate that GABA produced by the gut microbiota, mainly in the colon, may affect host behavioral characteristics via GABA receptors expressed in intestinal epithelial cells without being transferred to the blood. This study suggests a novel mechanism by which intestinal GABA exerts physiological effects, even in the presence of the blood-brain barrier.


Asunto(s)
Ansiedad , Células Epiteliales , Microbioma Gastrointestinal , Ácido gamma-Aminobutírico , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Ácido gamma-Aminobutírico/metabolismo , Ansiedad/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Neomicina/farmacología , Ratones Endogámicos C57BL , Receptores de GABA/metabolismo , Bifidobacterium/metabolismo , Probióticos/farmacología , Humanos
5.
Theranostics ; 14(14): 5682-5697, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39310110

RESUMEN

Maternal immune activation (MIA) is reported to increase the risk of psychiatric disorders in the offspring. However, the underlying mechanism remains unclear. Methods: We constructed a MIA mouse model by intraperitoneal injection of LPS into pregnant mice and evaluated the behaviors and gene expression profiles in the brains of the female and male offspring, respectively. Results: We found that the MIA female offspring exhibited increased anxiety and a large number of differentially expressed genes (DEGs) in the brain, which were enriched with candidate gene sets of psychiatric disorders and immune functions. In contrast, the MIA male offspring exhibited no significant abnormal behaviors and only a small number of DEGs that were not enriched with disease genes and immune functions. Therefore, we further pursued the downstream study on the molecular mechanism underlying the increased anxiety in the female offspring. We identified the lncRNA AU020206-IRFs-STAT1-cytokine axis by integrating lncRNA-protein interaction data and TF-promoter interaction data, and verified the axis in vitro and in vivo. Conclusion: This study illustrates that MIA upregulates the AU020206-IRFs-STAT1 axis in controlling the brain immunity linked to abnormal behaviors, providing a basis for understanding the role of MIA in psychiatric disorders.


Asunto(s)
Encéfalo , Citocinas , Modelos Animales de Enfermedad , Factor de Transcripción STAT1 , Animales , Femenino , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Ratones , Encéfalo/metabolismo , Encéfalo/inmunología , Embarazo , Citocinas/metabolismo , Masculino , Regulación hacia Arriba , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Lipopolisacáridos , Efectos Tardíos de la Exposición Prenatal/inmunología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ansiedad/inmunología , Ansiedad/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal
6.
Dev Psychobiol ; 66(7): e22548, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39268564

RESUMEN

Interpersonal closeness has important health benefits; however, recent work suggests that in certain contexts, closeness can come at a cost. In the current study, curvilinear relations between mother-child closeness and health (e.g., depressive and anxiety symptoms and hair cortisol concentrations [HCC]) were tested. Our sample consisted of 117 mother (Mage = 36.86) and child (Mage = 73.07 months, 50.86% male) dyads. A quadratic relationship between maternal perceived closeness with their child and self-reported depressive and anxiety symptoms, along with overall hair cortisol output, was hypothesized. Path analysis suggested that the quadratic term was predictive of maternal cortisol (ß = 0.28, p = 0.001) and depression (ß = 0.23, p = 0.014), such that both high and low perceived closeness predict greater maternal depressive symptoms and HCCs as compared to moderate levels of closeness. Results are discussed in terms of parenting support and burnout.


Asunto(s)
Ansiedad , Depresión , Cabello , Hidrocortisona , Relaciones Madre-Hijo , Madres , Humanos , Cabello/química , Femenino , Hidrocortisona/metabolismo , Masculino , Adulto , Ansiedad/metabolismo , Ansiedad/fisiopatología , Depresión/metabolismo , Niño , Preescolar
7.
Behav Pharmacol ; 35(7): 386-398, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39230562

RESUMEN

Increasing evidence indicates that prenatal cocaine exposure may result in many developmental and long-lasting neurological and behavioral effects. The behaviors of female animals are strongly associated with the estrous cycle. Estrogen receptors and oxytocin are important neuroendocrine factors that regulate social behavior and are of special relevance to females. However, whether prenatal cocaine exposure induces estrous cycle changes in offspring and whether neurobehavioral changes in estrus and diestrus offspring differ remains unclear. On gestational day 12, mice were administered cocaine once daily for seven consecutive days, then the estrous cycle was examined in adult female offspring, as well as locomotion, anxiety level, and social behaviors, and the expression of estrogen receptor alpha-immunoreactive and oxytocin-immunoreactive neurons were compared between estrus and diestrus offspring. Prenatal cocaine exposure resulted in the shortening of proestrus and estrus in the offspring. During estrus and diestrus, prenatally cocaine-exposed offspring showed increased anxiety levels and changed partial social behaviors; their motility showed no significant differences in estrus, but declined in diestrus. Prenatal cocaine exposure reduced estrogen receptor alpha-immunoreactive expression in the medial preoptic area, ventromedial hypothalamic nucleus, and arcuate nucleus and oxytocin-immunoreactive expression in the paraventricular nucleus in estrus and diestrus offspring. These results suggest that prenatal cocaine exposure induces changes in the offspring's estrous cycle and expression of estrogen receptor alpha and oxytocin in a brain region-specific manner and that prenatal cocaine exposure and the estrous cycle interactively change motility and partial social behavior. Estrogen receptor alpha and oxytocin signaling are likely to play important concerted roles in mediating the effects of prenatal cocaine exposure on the offspring.


Asunto(s)
Cocaína , Diestro , Receptor alfa de Estrógeno , Ciclo Estral , Estro , Oxitocina , Efectos Tardíos de la Exposición Prenatal , Conducta Social , Animales , Oxitocina/metabolismo , Femenino , Efectos Tardíos de la Exposición Prenatal/metabolismo , Embarazo , Receptor alfa de Estrógeno/metabolismo , Cocaína/farmacología , Ratones , Ciclo Estral/efectos de los fármacos , Estro/efectos de los fármacos , Diestro/efectos de los fármacos , Diestro/metabolismo , Conducta Animal/efectos de los fármacos , Ansiedad/metabolismo , Masculino , Neuronas/metabolismo , Neuronas/efectos de los fármacos
8.
Behav Pharmacol ; 35(7): 408-417, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39230572

RESUMEN

Omeprazole, a drug of choice for the management of gastric hyperacidity, influences serotonergic neurotransmission in brain regions and its long-term use is known to cause stress-related behavioral deficits including anxiety. Aim of the current study was to explore the effects of omeprazole treatment on immobilization-induced anxiety in rats, specifically on the role of serotonin (5-HT). In view of the role of serotonin-1A (5-HT1A) autoreceptor in the availability of 5-HT in brain regions, mRNA expression of this autoreceptor was performed in raphe nuclei. Similarly, because of the role of hippocampal 5-HT neurotransmission in anxiety-like disorders, expression of the 5-HT1A heteroreceptors was determined in this region. We found that the treatment with omeprazole reduces anxiety-like behavior in rats, increases the expression of 5-HT1A autoreceptor in the raphe and decreases the hippocampal expression of 5-HT1A heteroreceptor. This suggests a role of 5-HT1A receptor types in omeprazole-induced behavioral changes. It also indicates a potential role of omeprazole in the management of serotonergic disorders.


Asunto(s)
Ansiedad , Modelos Animales de Enfermedad , Hipocampo , Omeprazol , Receptor de Serotonina 5-HT1A , Estrés Psicológico , Animales , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Omeprazol/farmacología , Masculino , Ratas , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Estrés Psicológico/metabolismo , Estrés Psicológico/tratamiento farmacológico , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Ratas Wistar , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Serotonina/metabolismo , Núcleos del Rafe/metabolismo , Núcleos del Rafe/efectos de los fármacos , ARN Mensajero/metabolismo , Restricción Física , Inmovilización
9.
Gut Microbes ; 16(1): 2401939, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39259834

RESUMEN

Early life stress alters gut microbiota and increases the risk of neuropsychiatric disorders, including social deficits and anxiety, in the host. However, the role of gut commensal bacteria in early life stress-induced neurobehavioral abnormalities remains unclear. Using the maternally separated (MS) mice, our research has unveiled a novel aspect of this complex relationship. We discovered that the reduced levels of amino acid transporters in the intestine of MS mice led to low glutamine (Gln) levels in the blood and synaptic dysfunction in the medial prefrontal cortex (mPFC). Abnormally low blood Gln levels limit the brain's availability of Gln, which is required for presynaptic glutamate (Glu) and γ-aminobutyric acid (GABA) replenishment. Furthermore, MS resulted in gut microbiota dysbiosis characterized by a reduction in the relative abundance of Lactobacillus reuteri (L. reuteri). Notably, supplementation with L. reuteri ameliorates neurobehavioral abnormalities in MS mice by increasing intestinal amino acid transport and restoring synaptic transmission in the mPFC. In conclusion, our findings on the role of L. reuteri in regulating intestinal amino acid transport and buffering early life stress-induced behavioral abnormalities provide a novel insight into the microbiota-gut-brain signaling basis for emotional behaviors.


Asunto(s)
Ansiedad , Microbioma Gastrointestinal , Estrés Psicológico , Animales , Microbioma Gastrointestinal/fisiología , Ratones , Ansiedad/microbiología , Ansiedad/metabolismo , Estrés Psicológico/microbiología , Estrés Psicológico/metabolismo , Aminoácidos/metabolismo , Masculino , Ratones Endogámicos C57BL , Sistemas de Transporte de Aminoácidos/metabolismo , Corteza Prefrontal/metabolismo , Conducta Animal , Disbiosis/microbiología , Privación Materna , Glutamina/metabolismo , Eje Cerebro-Intestino/fisiología , Transmisión Sináptica , Femenino , Ácido Glutámico/metabolismo
10.
Transl Psychiatry ; 14(1): 392, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39341799

RESUMEN

Early social isolation (SI) leads to various abnormalities in emotion and behavior during adulthood. However, the negative impact of SI on offspring remains unclear. This study has discovered that paternal early SI causes social memory deficits and anxiety-like behavior in F1 young adult mice, with alterations of myelin and synapses in the medial prefrontal cortex (mPFC). The 2-week SI in the F1 progeny exacerbates social memory impairment and hypomyelination in the mPFC. Furthermore, the down-regulation of miR-124, a key inhibitor of myelinogenesis, or over-expression of its target gene Nr4a1 in the mPFC of the F1 mice improves social interaction ability and enhances oligodendrocyte maturation and myelin formation. Mechanistically, elevated levels of miR-124 in the sperm of paternal SI mice are transmitted epigenetically to offspring, altering the expression levels of miR-124/Nr4a1/glucocorticoid receptors in mPFC oligodendrocytes. This, in turn, impedes the establishment of myelinogenesis-dependent social behavior. This study unveils a novel mechanism through which miR-124 mediates the intergenerational effects of early isolation stress, ultimately impairing the establishment of social behavior and neurodevelopment.


Asunto(s)
MicroARNs , Corteza Prefrontal , Conducta Social , Aislamiento Social , Animales , MicroARNs/genética , MicroARNs/metabolismo , Ratones , Masculino , Corteza Prefrontal/metabolismo , Oligodendroglía/metabolismo , Ratones Endogámicos C57BL , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Conducta Animal/fisiología , Vaina de Mielina/metabolismo , Vaina de Mielina/genética , Ansiedad/genética , Ansiedad/metabolismo , Femenino
11.
Neurochem Res ; 49(11): 3143-3155, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39235577

RESUMEN

Prolactin (PRL) assumes a pivotal role during the postpartum phase, particularly within the hippocampus-a region densely populated with receptors for stress hormones, where stress significantly inhibits adult hippocampal neurogenesis (AHN). The reduction in neurogenesis is implicated in the pathogenesis of anxiety and depression. Mothers are at an increased risk of developing depression when exposed to chronic stress. Therefore, it is imperative to investigate the potential role of PRL in depression-like behaviors stemming from prolonged postpartum stress, and to explore any underlying mechanisms. Despite pup separation (PS) being a natural postpartum care practice, the impact of various PS methods on lactating dams remains uncertain. Lactating C57BL/6J mice, from postpartum day (PPD) 1 to PPD 21, underwent no PS (NPS), brief PS (15 min per day, PS15), or long PS (180 min per day, PS180), followed by 21 days of chronic restraint stress (CRS). Behavioral tests were conducted, and measurements included serum PRL concentration, PRL-R expression, and AHN in the hippocampus. Dams with CRS exhibited cognitive decline, depressive- and anxiety-like behaviors, and reduced PRL secretion, correlating with lower levels of AHN. PS15 dams displayed lower levels of depressive- and anxiety-like behaviors and cognitive decline compared to NPS and PS180 dams. Significantly, PS15 dams exhibited higher levels of AHN, PRL-R expression in the hippocampus, and serum PRL concentration. This study collectively reveals reduced serum PRL and AHN in dams with cognitive decline and depressive- and anxiety-like behaviors after CRS. Brief PS confers resistance to behavioral deficits after CRS, increasing serum PRL concentration and reversing AHN decrease in dams.


Asunto(s)
Hipocampo , Lactancia , Ratones Endogámicos C57BL , Neurogénesis , Prolactina , Estrés Psicológico , Animales , Prolactina/sangre , Prolactina/metabolismo , Femenino , Hipocampo/metabolismo , Neurogénesis/fisiología , Estrés Psicológico/metabolismo , Lactancia/metabolismo , Ratones , Periodo Posparto , Ansiedad/metabolismo , Privación Materna , Depresión/metabolismo , Receptores de Prolactina/metabolismo
12.
J Affect Disord ; 366: 262-272, 2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39209273

RESUMEN

BACKGROUND: Social isolation not only increases the risk of mortality in later life but also causes depressive symptoms, cognitive and physical disabilities. Although RNA m6A modifications are suggested to play key roles in brain development, neuronal signaling and neurological disorders, both the roles of m6A and the enzymes that regulate RNA m6A modification in social isolation induced abnormal behavior is unknown. The present study aims to explore the possible epitranscriptomic role of RNA m6A modifications and its enzymes in social isolation induced impaired behavior. METHODS: 3-4 weeks mice experiencing 8 weeks social isolation stress (SI) were used in the present study. We quantified m6A levels in brain regions related to mood and cognitive behavior. And the expression of hippocampal m6A enzymes was also determined. The role of hippocampal m6A and its enzymes in SI induced abnormal behavior was further verified by the virus tool. RESULTS: SI led to not only depressive and anxiety-like behaviors but also cognitive impairment, with corresponding decreases in hippocampal m6A and METTL14. Hippocampal over-expression METTL14 with lentivirus not only rescued these behaviors but also enhanced the hippocampal m6A level. Hippocampal over-expression METTL14 resulted in increased synaptic related genes. CONCLUSIONS: We provide the first evidence that post-weaning social isolation reduces hippocampal m6A level and causes altered expression of m6A enzyme in mice. Importantly, hippocampal METTL14 over-expression alleviated the SI-induced depression/anxiety-like and impaired cognitive behaviors and enhanced m6A level and synaptic related genes expression.


Asunto(s)
Hipocampo , Metiltransferasas , Aislamiento Social , Animales , Hipocampo/metabolismo , Masculino , Ratones , Metiltransferasas/genética , Metiltransferasas/metabolismo , Depresión/genética , Depresión/metabolismo , Conducta Animal/fisiología , Ansiedad/genética , Ansiedad/metabolismo , Estrés Psicológico/metabolismo , Estrés Psicológico/genética , Ratones Endogámicos C57BL , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Epigénesis Genética , Metilación de ARN
13.
Neurobiol Dis ; 200: 106633, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39117119

RESUMEN

The triggers of status epilepticus (SE) in non-epileptic patients can vary widely, from idiopathic causes to exposure to chemoconvulsants. Regardless of its etiology, prolonged SE can cause significant brain damage, commonly resulting in the development of epilepsy, which is often accompanied by increased anxiety. GABAA receptor (GABAAR)-mediated inhibition has a central role among the mechanisms underlying brain damage and the ensuing epilepsy and anxiety. During SE, calcium influx primarily via ionotropic glutamate receptors activates signaling cascades which trigger a rapid internalization of synaptic GABAARs; this weakens inhibition, exacerbating seizures and excitotoxicity. GABAergic interneurons are more susceptible to excitotoxic death than principal neurons. During the latent period of epileptogenesis, the aberrant reorganization in synaptic interactions that follow interneuronal loss in injured brain regions, leads to the formation of hyperexcitable, seizurogenic neuronal circuits, along with disturbances in brain oscillatory rhythms. Reduction in the spontaneous, rhythmic "bursts" of IPSCs in basolateral amygdala neurons is likely to play a central role in anxiogenesis. Protecting interneurons during SE is key to preventing both epilepsy and anxiety. Antiglutamatergic treatments, including antagonism of calcium-permeable AMPA receptors, can be expected to control seizures and reduce excitotoxicity not only by directly suppressing hyperexcitation, but also by counteracting the internalization of synaptic GABAARs. Benzodiazepines, as delayed treatment of SE, have low efficacy due to the reduction and dispersion of their targets (the synaptic GABAARs), but also because themselves contribute to further reduction of available GABAARs at the synapse; furthermore, benzodiazepines may be completely ineffective in the immature brain.


Asunto(s)
Ansiedad , Receptores de GABA-A , Estado Epiléptico , Estado Epiléptico/metabolismo , Receptores de GABA-A/metabolismo , Animales , Humanos , Ansiedad/metabolismo , Inhibición Neural/fisiología
14.
Psychoneuroendocrinology ; 169: 107168, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39146876

RESUMEN

Poor maternal diet and psychosocial stress represent two environmental factors that can significantly impact maternal health during pregnancy. While various mouse models have been developed to study the relationship between maternal and offspring health and behaviour, few incorporate multiple sources of stress that mirror the complexity of human experiences. Maternal high-fat diet (HF) models in rodents are well-established, whereas use of psychosocial stress interventions in female mice are still emerging. The social instability stress (SIS) paradigm, serves as a chronic and unpredictable form of social stress. To evaluate the combined effects of a poor maternal diet and intermittent social stress on maternal health and behaviour, we developed a novel maternal stress model using adult female C57Bl/6 mice. We observed that all HF+ mice demonstrated rapid weight gain, elevated fasting blood glucose levels and impaired glucose tolerance independent of the presence (+) or absence (-) of SIS. Behavioural testing output revealed anxiety-like behaviours remained similar across all groups prior to pregnancy. However, integrated anxiety z-scores revealed a mixed anxious profile amongst HF+/SIS+ females prior to pregnancy. HF+/SIS+ females also did not show reduced plasma ACTH and corticosterone levels that were observed in our other HF+ and HF- stress groups after SIS exposure. Further, HF+/SIS+ females demonstrated significant postpartum maternal neglect, resulting in fewer numbers of live offspring. These findings suggest that prolonged maternal HF diet consumption, coupled with previous exposure to SIS, places a significant burden on the maternal stress response system, resulting in reduced parental investment and negative postpartum behaviour towards offspring.


Asunto(s)
Ansiedad , Dieta Alta en Grasa , Conducta Materna , Ratones Endogámicos C57BL , Estrés Psicológico , Femenino , Animales , Dieta Alta en Grasa/efectos adversos , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Embarazo , Ratones , Conducta Materna/fisiología , Conducta Materna/psicología , Ansiedad/metabolismo , Ansiedad/psicología , Corticosterona/sangre , Efectos Tardíos de la Exposición Prenatal/metabolismo , Conducta Animal/fisiología , Adaptación Psicológica/fisiología , Glucemia/metabolismo , Hormona Adrenocorticotrópica/sangre , Aumento de Peso/fisiología
15.
Brain Res ; 1844: 149196, 2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39181223

RESUMEN

This work was done to investigate the ameliorating impact of 4-methylumbilliferon (4-MU) on spatial learning and memory dysfunction and restraint stress (STR)-induced anxiety-like behaviors in male Wistar rats and the underlying mechanisms. Thirty-two animals were assigned into 4 cohorts: control, 4-MU, STR, and STR+4-MU. Animals were exposed to STR for 4 h per day for 14 consecutive days or kept in normal conditions (healthy animals without exposure to stress). 4-MU (25 mg/kg) was intraperitoneally administered once daily to STR rats before restraint stress for 14 consecutive days. The behavioral tests were performed through Morris water maze tests and elevated-plus maze to examine learning/memory function, and anxiety levels, respectively. The levels of the antioxidant defense biomarkers (GPX, SOD) and MDA as an oxidant molecule in the brain tissues were measured using commercial ELISA kits. Neuronal loss or density of neurons was evaluated using Nissl staining. STR exposure could cause significant alterations in the levels of the antioxidant defense biomarkers (MDA, GPX, and SOD) in the prefrontal cortex and hippocampus, induce anxiety, and impair spatial learning and memory function. Treatment with 4-MU markedly reduced anxiety levels and improved spatial learning and memory dysfunction via restoring the antioxidant defense biomarkers to normal values and reducing MDA levels. Moreover, more intact cells with normal morphologies were detected in STR-induced animals treated with 4-MU. 4-MU could attenuate the STR-induced anxiety-like behaviors and spatial learning and memory dysfunction by reducing oxidative damage and neuronal loss in the prefrontal cortex and hippocampus region. Taken together, our findings provide new insights regarding the potential therapeutic effects of 4-MU against neurobehavioral disorders induced by STR.


Asunto(s)
Ansiedad , Muerte Celular , Trastornos de la Memoria , Neuronas , Estrés Oxidativo , Ratas Wistar , Animales , Estrés Oxidativo/efectos de los fármacos , Masculino , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Muerte Celular/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/complicaciones , Aprendizaje por Laberinto/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Antioxidantes/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo
16.
Cell Metab ; 36(9): 2104-2117.e4, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39163863

RESUMEN

Lactate is a critical metabolite during the body's adaption to exercise training, which effectively relieves anxiety-like disorders. The biological mechanism of lactate in the exercise-mediated anxiolytic effect has, however, not been comprehensively investigated. Here, we report that exercise-induced lactate markedly potentiates the lactylation of multiple synaptic proteins, among which synaptosome-associated protein 91 (SNAP91) is the critical molecule for synaptic functions. Both anatomical evidence and in vivo recording data showed that the lactylation of SNAP91 confers resilience against chronic restraint stress (CRS) via potentiating synaptic structural formation and neuronal activity in the medial prefrontal cortex (mPFC). More interestingly, exercise-potentiated lactylation of SNAP91 is necessary for the prevention of anxiety-like behaviors in CRS mice. These results collectively suggest a previously unrecognized non-histone lactylation in the brain for modulating mental functions and provide evidence for the brain's metabolic adaption during exercise paradigms.


Asunto(s)
Ratones Endogámicos C57BL , Condicionamiento Físico Animal , Estrés Psicológico , Animales , Ratones , Masculino , Estrés Psicológico/metabolismo , Corteza Prefrontal/metabolismo , Ansiedad/metabolismo , Sinapsis/metabolismo , Ácido Láctico/metabolismo
17.
Int J Mol Sci ; 25(16)2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39201372

RESUMEN

Accumulating evidence suggests an involvement of sphingolipids, vital components of cell membranes and regulators of cellular processes, in the pathophysiology of both Parkinson's disease and major depressive disorder, indicating a potential common pathway in these neuropsychiatric conditions. Based on this interaction of sphingolipids and synuclein proteins, we explored the gene expression patterns of α-, ß-, and γ-synuclein in a knockout mouse model deficient for acid sphingomyelinase (ASM), an enzyme catalyzing the hydrolysis of sphingomyelin to ceramide, and studied associations with behavioral parameters. Normalized Snca, Sncb, and Sncg gene expression was determined by quantitative PCR in twelve brain regions of sex-mixed homozygous (ASM-/-, n = 7) and heterozygous (ASM+/-, n = 7) ASM-deficient mice, along with wild-type controls (ASM+/+, n = 5). The expression of all three synuclein genes was brain region-specific but independent of ASM genotype, with ß-synuclein showing overall higher levels and the least variation. Moreover, we discovered correlations of gene expression levels between brain regions and depression- and anxiety-like behavior and locomotor activity, such as a positive association between Snca mRNA levels and locomotion. Our results suggest that the analysis of synuclein genes could be valuable in identifying biomarkers and comprehending the common pathological mechanisms underlying various neuropsychiatric disorders.


Asunto(s)
Ansiedad , Encéfalo , Depresión , Modelos Animales de Enfermedad , Locomoción , Ratones Noqueados , Esfingomielina Fosfodiesterasa , Animales , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Ratones , Encéfalo/metabolismo , Depresión/genética , Depresión/metabolismo , Ansiedad/genética , Ansiedad/metabolismo , Locomoción/genética , Masculino , Sinucleínas/metabolismo , Sinucleínas/genética , Conducta Animal , Femenino , Genotipo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Ratones Endogámicos C57BL
18.
Neurochem Res ; 49(10): 2957-2971, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39088164

RESUMEN

Depression and anxiety disorders are prevalent stress-related neuropsychiatric disorders and involve multiple molecular changes and dysfunctions across various brain regions. However, the specific and shared pathophysiological mechanisms occurring in these regions remain unclear. Previous research used a rat model of chronic mild stress (CMS) to segregate and identify depression-susceptible, anxiety-susceptible, and insusceptible groups; then the proteomes of six distinct brain regions (the hippocampus, prefrontal cortex, hypothalamus, pituitary, olfactory bulb, and striatum) were separately and quantitatively analyzed. To gain a comprehensive and systematic understanding of the molecular abnormalities, this study aimed to investigate and compare differential proteomics data from the six regions. Differentially expressed proteins (DEPs) were identified in between specific regions and across all regions and subjected to a series of bioinformatics analyses. Regional comparisons showed that stress-induced proteomic changes and corresponding gene ontology and pathway enrichments were largely distinct, attributable to differences in cell populations, protein compositions, and brain functions of these areas. Additionally, a notable degree of overlap in the significantly enriched terms was identified, potentially suggesting strong connections in the enrichment across different regions. Furthermore, intra-regional and inter-regional protein-protein interaction networks and drug-target-DEP networks were constructed. Integrated analysis of the three association networks in the six regions, along with the DisGeNET database, identified ten DEPs as potential targets for anti-depression/anxiety drugs. Collectively, these findings revealed commonalities and differences across different brain regions at the protein level induced by CMS, and identified several novel protein targets for the development of new therapeutics for depression and anxiety.


Asunto(s)
Ansiolíticos , Encéfalo , Proteoma , Ratas Sprague-Dawley , Estrés Psicológico , Animales , Estrés Psicológico/metabolismo , Estrés Psicológico/tratamiento farmacológico , Proteoma/metabolismo , Masculino , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Ansiolíticos/farmacología , Ansiolíticos/uso terapéutico , Ansiedad/metabolismo , Ansiedad/tratamiento farmacológico , Depresión/metabolismo , Depresión/tratamiento farmacológico , Mapas de Interacción de Proteínas , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Ratas , Proteómica
19.
Neuroscience ; 557: 67-80, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39127344

RESUMEN

Puberty is a sensitive developmental period during which stressors can cause lasting brain and behavioural deficits. While the acute effects of pubertal lipopolysaccharide (LPS) and antimicrobial (AMNS) treatments are known, their enduring impacts on neurodegeneration-related mechanisms and behaviours remain unclear. This study examined these effects in male and female mice. At five weeks old, mice received 200ul of either broad-spectrum antimicrobials or water through oral gavage twice daily for seven days. At six weeks of age, they received an intraperitoneal injection of either saline or LPS. Four weeks later, adult mice underwent neurodegeneration-related behavioural tests, including the rotarod, forepaw stride length, reversed grid hang, open field, and buried pellet tests. Two days after the final test, brain and ileal samples were collected. Results showed that female mice treated with both AMNS and LPS exhibited deficits in neuromuscular strength, while males treated with LPS alone showed increased anxiety-like behaviours. Males treated with AMNS alone had decreased sigma-1 receptor (S1R) expression in the cornu ammonis 1 (CA1) and dentate gyrus (DG), while females treated with both AMNS and LPS had decreased S1R expression. Additionally, males treated with either LPS or AMNS had lower glial-derived neurotrophic factor receptor alpha-1 (GFRA1) expression in the primary motor cortex (M1) than females. Mice treated with LPS alone had decreased GFRA1 expression in the DG and decreased S1R expression in the secondary motor cortex (M2). These findings suggest that pubertal AMNS and LPS treatments may lead to enduring changes in biomarkers and behaviours related to neurodegeneration.


Asunto(s)
Lipopolisacáridos , Animales , Femenino , Masculino , Lipopolisacáridos/farmacología , Ratones , Maduración Sexual/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ansiedad/inducido químicamente , Ansiedad/metabolismo
20.
Neurosci Lett ; 841: 137944, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39154843

RESUMEN

Depression and anxiety are prominent symptoms of withdrawal syndrome, often caused by the abuse of addictive drugs like morphine. N-palmitoylethanolamide (PEA), a biologically active lipid, is utilized as an anti-inflammatory and analgesic medication. Recent studies have highlighted PEA's role in mitigating cognitive decline and easing depression resulting from chronic pain. However, it remains unknown whether PEA can influence negative emotions triggered by morphine withdrawal. This study seeks to explore the impact of PEA on such emotions and investigate the underlying mechanisms. Mice subjected to morphine treatment underwent a 10-day withdrawal period, followed by assessments of the effect of PEA on anxiety- and depression-like behaviors using various tests. Enzyme-linked immunosorbent assay was conducted to measure levels of monoamine neurotransmitters in specific brain regions. The findings indicate that PEA mitigated anxiety and depression symptoms and reduced 5-hydroxytryptamine, noradrenaline, and dopamine levels in the hippocampus and prefrontal cortex. In summary, PEA demonstrates a significant positive effect on negative emotions associated with morphine withdrawal, accompanied with the reduction in levels of monoamine neurotransmitters in key brain regions. These insights could be valuable for managing negative emotions arising from morphine withdrawal.


Asunto(s)
Amidas , Ansiedad , Depresión , Etanolaminas , Morfina , Ácidos Palmíticos , Síndrome de Abstinencia a Sustancias , Animales , Síndrome de Abstinencia a Sustancias/psicología , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Etanolaminas/farmacología , Ácidos Palmíticos/farmacología , Ratones , Masculino , Morfina/farmacología , Depresión/metabolismo , Depresión/tratamiento farmacológico , Depresión/psicología , Depresión/etiología , Amidas/farmacología , Ansiedad/tratamiento farmacológico , Ansiedad/psicología , Ansiedad/metabolismo , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Emociones/efectos de los fármacos , Serotonina/metabolismo , Dependencia de Morfina/metabolismo , Dependencia de Morfina/psicología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Norepinefrina/metabolismo , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA