Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.180
Filtrar
1.
Neurobiol Dis ; 200: 106642, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39173845

RESUMEN

Adverse experiences during infancy and adolescence have an important and enduring effect on the brain and are predisposing factors for mental disorders, particularly major depression. This impact is particularly notable in regions with protracted development, such as the prefrontal cortex. The inhibitory neurons of this cortical region are altered by peripubertal stress (PPS), particularly in female mice. In this study we have explored whether the inhibitory circuits of the thalamus are impacted by PPS in male and female mice. This diencephalic structure, as the prefrontal cortex, also completes its development during postnatal life and is affected by adverse experiences. The long-term changes induced by PPS were exclusively found in adult female mice. We have found that PPS increases depressive-like behavior and induces changes in parvalbumin-expressing (PV+) cells of the thalamic reticular nucleus (TRN). We observed reductions in the volume of the TRN, together with those of parameters related to structures/molecules that regulate the plasticity and connectivity of PV+ cells: perineuronal nets, matricellular structures surrounding PV+ neurons, and the polysialylated form of the neural cell adhesion molecule (PSA-NCAM). The expression of the GluN1, but not of GluN2C, NMDA receptor subunit was augmented in the TRN after PPS. An increase in the fluorescence intensity of PV+ puncta was also observed in the synaptic output of TRN neurons in the lateral posterior thalamic nucleus. These results demonstrate that the inhibitory circuits of the thalamus, as those of the prefrontal cortex, are vulnerable to the effects of aversive experiences during early life, particularly in females. This vulnerability is probably related to the protracted development of the TRN and might contribute to the development of psychiatric disorders.


Asunto(s)
Estrés Psicológico , Animales , Femenino , Masculino , Ratones , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Núcleos Talámicos/metabolismo , Ratones Endogámicos C57BL , Parvalbúminas/metabolismo , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
2.
Neurobiol Dis ; 200: 106610, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39032799

RESUMEN

Chronic stress is a major precursor to various neuropsychiatric disorders and is linked with increased inflammation in the brain. However, the bidirectional association between inflammation and chronic stress has yet to be fully understood. Astrocytes are one of the key inflammatory regulators in the brain, and the morphological change in reactive astrocytes serves as an important indicator of inflammation. In this study, we evaluated the sex-specific astrocyte response to chronic stress or systemic inflammation in key brain regions associated with mood disorders. We conducted the unpredictable chronic mild stress (UCMS) paradigm to model chronic stress, or lipopolysaccharide (LPS) injection to model systemic inflammation. To evaluate stress-induced morphological changes in astrocyte complexity, we measured GFAP fluorescent intensity for astrocyte expression, branch bifurcation by quantifying branch points and terminal points, branch arborization by conducting Sholl analysis, and calculated the ramification index. Our analysis indicated that chronic stress-induced morphological changes in astrocytes in all brain regions investigated. The effects of chronic stress were region and sex specific. Notably, females had greater stress or inflammation-induced astrocyte activation in the hypothalamus (HYPO), CA1, CA3, and amygdala (AMY) than males. These findings indicate that chronic stress induces astrocyte activation that may drive sex and region-specific effects in females, potentially contributing to sex-dependent mechanisms of disease.


Asunto(s)
Astrocitos , Encéfalo , Caracteres Sexuales , Estrés Psicológico , Astrocitos/patología , Astrocitos/metabolismo , Animales , Masculino , Estrés Psicológico/patología , Estrés Psicológico/metabolismo , Femenino , Encéfalo/patología , Encéfalo/metabolismo , Lipopolisacáridos/farmacología , Inflamación/patología , Inflamación/metabolismo , Ratones Endogámicos C57BL
3.
Mol Brain ; 17(1): 31, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38831333

RESUMEN

Chronic psychological stress is a critical factor for neurological complications like anxiety disorders, dementia, and depression. Our previous results show that chronic restraint stress causes cognitive deficits and mood dysregulation by inducing autophagic death of adult hippocampal neural stem cells (NSCs). However, it is unknown whether other models of psychological stress also induce autophagic death of adult hippocampal NSCs. Here, we show that chronic unpredictable stress (CUS) for 10 days impaired memory function and increased anxiety in mice. Immunohistochemical staining with SOX2 and KI67 revealed a significant reduction in the number of NSCs in the hippocampus following exposure to CUS. However, these deficits were prevented by NSC-specific, inducible conditional deletion of Atg7. These findings suggest that autophagic death of adult hippocampal NSCs is a critical pathogenic mechanism underlying stress-induced brain disorders.


Asunto(s)
Hipocampo , Células-Madre Neurales , Estrés Psicológico , Animales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Hipocampo/patología , Estrés Psicológico/patología , Ratones Endogámicos C57BL , Autofagia/fisiología , Enfermedad Crónica , Proteína 7 Relacionada con la Autofagia/metabolismo , Proteína 7 Relacionada con la Autofagia/genética , Ansiedad/patología , Ansiedad/fisiopatología , Masculino , Células Madre Adultas/patología , Muerte Celular Autofágica , Memoria/fisiología , Ratones
4.
Acta Neuropathol Commun ; 12(1): 100, 2024 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-38886854

RESUMEN

A link between chronic stress and Parkinson's disease (PD) pathogenesis is emerging. Ample evidence demonstrates that the presynaptic neuronal protein alpha-synuclein (asyn) is closely tied to PD pathogenesis. However, it is not known whether stress system dysfunction is present in PD, if asyn is involved, and if, together, they contribute to neurodegeneration. To address these questions, we assess stress axis function in transgenic rats overexpressing full-length wildtype human asyn (asyn BAC rats) and perform multi-level stress and PD phenotyping following chronic corticosterone administration. Stress signaling, namely corticotropin-releasing factor, glucocorticoid and mineralocorticoid receptor gene expression, is also examined in post-mortem PD patient brains. Overexpression of human wildtype asyn leads to HPA axis dysregulation in rats, while chronic corticosterone administration significantly aggravates nigrostriatal degeneration, serine129 phosphorylated asyn (pS129) expression and neuroinflammation, leading to phenoconversion from a prodromal to an overt motor PD phenotype. Interestingly, chronic corticosterone in asyn BAC rats induces a robust, twofold increase in pS129 expression in the hypothalamus, the master regulator of the stress response, while the hippocampus, both a regulator and a target of the stress response, also demonstrates elevated pS129 asyn levels and altered markers of stress signalling. Finally, defective hippocampal stress signalling is mirrored in human PD brains and correlates with asyn expression levels. Taken together, our results link brain stress system dysregulation with asyn and provide evidence that elevated circulating glucocorticoids can contribute to asyn-induced neurodegeneration, ultimately triggering phenoconversion from prodromal to overt PD.


Asunto(s)
Corticosterona , Enfermedad de Parkinson , Ratas Transgénicas , Estrés Psicológico , alfa-Sinucleína , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Animales , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Humanos , Ratas , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Masculino , Corticosterona/sangre , Encéfalo/metabolismo , Encéfalo/patología , Sistema Hipotálamo-Hipofisario/metabolismo , Femenino , Sistema Hipófiso-Suprarrenal/metabolismo
5.
Glia ; 72(9): 1646-1662, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38801194

RESUMEN

The adult brain retains a high repopulation capacity of astrocytes after deletion, and both mature astrocytes in the neocortex and neural stem cells in neurogenic regions possess the potential to generate astrocytes. However, the origin and the repopulation dynamics of the repopulating astrocytes after deletion remain largely unclear. The number of astrocytes is reduced in the medial prefrontal cortex (mPFC) of patients with depression, and selective elimination of mPFC astrocytes is sufficient to induce depression-like behaviors in rodents. However, whether astrocyte repopulation capacity is impaired in depression is unknown. In this study, we used different transgenic mouse lines to genetically label different cell types and demonstrated that in the mPFC of normal adult mice of both sexes, mature astrocytes were a major source of the repopulating astrocytes after acute deletion induced by an astrocyte-specific toxin, L-alpha-aminoadipic acid (L-AAA), and astrocyte regeneration was accomplished within two weeks accompanied by reversal of depression-like behaviors. Furthermore, re-ablation of mPFC astrocytes post repopulation led to reappearance of depression-like behaviors. In adult male mice subjected to 14-day chronic restraint stress, a well-validated mouse model of depression, the number of mPFC astrocytes was reduced; however, the ability of mPFC astrocytes to repopulate after L-AAA-induced deletion was largely unaltered. Our study highlights a potentially beneficial role for repopulating astrocytes in depression and provides novel therapeutic insights into enhancing local mature astrocyte generation in depression.


Asunto(s)
Astrocitos , Depresión , Ratones Transgénicos , Corteza Prefrontal , Animales , Astrocitos/metabolismo , Corteza Prefrontal/metabolismo , Masculino , Depresión/genética , Depresión/patología , Femenino , Ratones Endogámicos C57BL , Ratones , Modelos Animales de Enfermedad , Restricción Física , Ácido 2-Aminoadípico , Estrés Psicológico/patología , Estrés Psicológico/metabolismo
6.
Brain Res Bull ; 213: 110981, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38777132

RESUMEN

INTRODUCTION: The medial prefrontal cortex (mPFC) forms output pathways through projection neurons, inversely receiving adjacent and long-range inputs from other brain regions. However, how afferent neurons of mPFC are affected by chronic stress needs to be clarified. In this study, the effects of chronic restraint stress (CRS) on the distribution density of mPFC dendrites/dendritic spines and the projections from the cortex and subcortical brain regions to the mPFC were investigated. METHODS: In the present study, C57BL/6 J transgenic (Thy1-YFP-H) mice were subjected to CRS to establish an animal model of depression. The infralimbic (IL) of mPFC was selected as the injection site of retrograde AAV using stereotactic technique. The effects of CRS on dendrites/dendritic spines and afferent neurons of the mPFC IL were investigaed by quantitatively assessing the distribution density of green fluorescent (YFP) positive dendrites/dendritic spines and red fluorescent (retrograde AAV recombinant protein) positive neurons, respectively. RESULTS: The results revealed that retrograde tracing virus labeled neurons were widely distributed in ipsilateral and contralateral cingulate cortex (Cg1), second cingulate cortex (Cg2), prelimbic cortex (PrL), infralimbic cortex, medial orbital cortex (MO), and dorsal peduncular cortex (DP). The effects of CRS on the distribution density of mPFC red fluorescence positive neurons exhibited regional differences, ranging from rostral to caudal or from top to bottom. Simultaneously, CRS resulted a decrease in the distribution density of basal, proximal and distal dendrites, as well as an increase in the loss of dendritic spines of the distal dendrites in the IL of mPFC. Furthermore, varying degrees of red retrograde tracing virus fluorescence signals were observed in other cortices, amygdala, hippocampus, septum/basal forebrain, hypothalamus, thalamus, mesencephalon, and brainstem in both ipsilateral and contralateral brain. CRS significantly reduced the distribution density of red fluorescence positive neurons in other cortices, hippocampus, septum/basal forebrain, hypothalamus, and thalamus. Conversely, CRS significantly increased the distribution density of red fluorescence positive neurons in amygdala. CONCLUSION: Our results suggest a possible mechanism that CRS leads to disturbances in synaptic plasticity by affecting multiple inputs to the mPFC, which is characterized by a decrease in the distribution density of dendrites/dendritic spines in the IL of mPFC and a reduction in input neurons of multiple cortices to the IL of mPFC as well as an increase in input neurons of amygdala to the IL of mPFC, ultimately causing depression-like behaviors.


Asunto(s)
Depresión , Ratones Endogámicos C57BL , Ratones Transgénicos , Corteza Prefrontal , Restricción Física , Estrés Psicológico , Animales , Corteza Prefrontal/patología , Corteza Prefrontal/metabolismo , Estrés Psicológico/patología , Estrés Psicológico/metabolismo , Ratones , Depresión/patología , Masculino , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Vías Aferentes , Dendritas/patología , Dendritas/metabolismo , Neuronas Aferentes/patología , Neuronas Aferentes/metabolismo , Encéfalo/patología , Encéfalo/metabolismo
7.
Int J Dev Neurosci ; 84(4): 314-327, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38584149

RESUMEN

Autism spectrum disorder (ASD) is a fast-growing neurodevelopmental disorder throughout the world. Experiencing early life stresses (ELS) like maternal separation (MS) is associated with autistic-like behaviors. It has been proposed that disturbance in the gut-brain axis-mediated psychiatric disorders following MS. The role of disruption in the integrity of gut-brain barrier in ASD remains unclear. Addressing this knowledge gap, in this study we aimed to investigate role of the gut-brain barrier integrity in mediating autistic-like behaviors in mouse models of MS stress. To do this, mice neonates are separated daily from their mothers from postnatal day (PND) 2 to PND 14 for 3 hours. During PND58-60, behavioral tests related to autistic-like behaviors including three-chamber sociability, shuttle box, and resident-intruder tests were performed. Then, prefrontal cortex (PFC), hippocampus, and colon samples were dissected out for histopathological and molecular evaluations. Results showed that MS is associated with impaired sociability and social preference indexes, aggressive behaviors, and impaired passive avoidance memory. The gene expression of CLDN1 decreased in the colon, and the gene expression of CLDN5, CLDN12, and MMP9 increased in the PFC of the MS mice. MS is associated with decrease in the diameter of CA1 and CA3 areas of the hippocampus. In addition, MS led to histopathological changes in the colon. We concluded that, probably, disturbance in the gut-brain barrier integrities mediated the autistic-like behavior in MS stress in mice.


Asunto(s)
Modelos Animales de Enfermedad , Privación Materna , Estrés Psicológico , Animales , Ratones , Estrés Psicológico/patología , Eje Cerebro-Intestino/fisiología , Femenino , Conducta Animal/fisiología , Masculino , Hipocampo/patología , Hipocampo/metabolismo , Corteza Prefrontal/patología , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Trastorno del Espectro Autista/fisiopatología , Conducta Social , Trastorno Autístico/patología , Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Barrera Hematoencefálica/patología , Animales Recién Nacidos , Colon/patología
8.
Glia ; 72(1): 111-132, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37675659

RESUMEN

Chronic environmental stress and traumatic social experiences induce maladaptive behavioral changes and is a risk factor for major depressive disorder (MDD) and various anxiety-related psychiatric disorders. Clinical studies and animal models of chronic stress have reported that symptom severity is correlated with innate immune responses and upregulation of neuroinflammatory cytokine signaling in brain areas implicated in mood regulation (mPFC; medial Prefrontal Cortex). Despite increasing evidence implicating impairments of neuroplasticity and synaptic signaling deficits into the pathophysiology of stress-related mental disorders, how microglia may modulate neuronal homeostasis in response to chronic stress has not been defined. Here, using the repeated social defeat stress (RSDS) mouse model we demonstrate that microglial-induced inflammatory responses are regulating neuronal plasticity associated with psychosocial stress. Specifically, we show that chronic stress induces a rapid activation and proliferation of microglia as well as macrophage infiltration in the mPFC, and these processes are spatially related to neuronal activation. Moreover, we report a significant association of microglial inflammatory responses with susceptibility or resilience to chronic stress. In addition, we find that exposure to chronic stress exacerbates phagocytosis of synaptic elements and deficits in neuronal plasticity. Importantly, by utilizing two different CSF1R inhibitors (the brain penetrant PLX5622 and the non-penetrant PLX73086) we highlight a crucial role for microglia (and secondarily macrophages) in catalyzing the pathological manifestations linked to psychosocial stress in the mPFC and the resulting behavioral deficits usually associated with depression.


Asunto(s)
Trastorno Depresivo Mayor , Microglía , Ratones , Animales , Humanos , Microglía/patología , Macrófagos , Neuronas , Estrés Psicológico/complicaciones , Estrés Psicológico/patología
9.
Neurosci Lett ; 812: 137403, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37473795

RESUMEN

In males, chronic stress enhances dendritic complexity in the amygdala, a region important in emotion regulation. An amygdalar subregion, the basolateral amygdala (BLA), is influenced by the hippocampus and prefrontal cortex to coordinate emotional learning and memory. This study quantified changes in dendritic complexity of BLA stellate neurons ten days after an unpredictable chronic stressor ended in both male and female rats. In addition, dendritic complexity of hippocampal neurons in male rats was assessed at a similar timepoint. Following Golgi processing, stressed male and female rats showed enhanced BLA dendritic complexity; increased arborization occurred near the soma in males and distally in females. As the brain was sampled ten days after chronic stress ended, BLA dendritic hypertrophy persisted in both sexes after the stressor had ended. For the hippocampus, CA3 dendritic complexity was similar for control and stressed males when assessed eight days after stress ended, suggesting that any stress-induced changes had resolved. These results show persistent enhancement of BLA dendritic arborization in both sexes following chronic stress, reveal sex differences in how BLA hypertrophy manifests, and suggest a putative neurobiological substrate by which chronic stress may create a vulnerable phenotype for emotional dysfunction.


Asunto(s)
Amígdala del Cerebelo , Espinas Dendríticas , Hipocampo , Hipertrofia , Neuronas , Estrés Psicológico , Animales , Femenino , Masculino , Ratas , Amígdala del Cerebelo/patología , Complejo Nuclear Basolateral/patología , Enfermedad Crónica , Espinas Dendríticas/patología , Hipocampo/citología , Hipertrofia/patología , Neuronas/patología , Estrés Psicológico/patología , Factores de Tiempo , Caracteres Sexuales , Restricción Física
10.
Cell Death Dis ; 14(4): 278, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-37076499

RESUMEN

Neuronal apoptosis has been well-recognized as a critical mediator in the pathogenesis of depressive disorders. Tissue kallikrein-related peptidase 8 (KLK8), a trypsin-like serine protease, has been implicated in the pathogenesis of several psychiatric disorders. The present study aimed to explore the potential function of KLK8 in hippocampal neuronal cell apoptosis associated with depressive disorders in rodent models of chronic unpredictable mild stress (CUMS)-induced depression. It was found that depression-like behavior in CUMS-induced mice was associated with hippocampal KLK8 upregulation. Transgenic overexpression of KLK8 exacerbated, whereas KLK8 deficiency attenuated CUMS-induced depression-like behaviors and hippocampal neuronal apoptosis. In HT22 murine hippocampal neuronal cells and primary hippocampal neurons, adenovirus-mediated overexpression of KLK8 (Ad-KLK8) was sufficient to induce neuron apoptosis. Mechanistically, it was identified that the neural cell adhesion molecule 1 (NCAM1) may associate with KLK8 in hippocampal neurons as KLK8 proteolytically cleaved the NCAM1 extracellular domain. Immunofluorescent staining exhibited decreased NCAM1 in hippocampal sections obtained from mice or rats exposed to CUMS. Transgenic overexpression of KLK8 exacerbated, whereas KLK8 deficiency largely prevented CUMS-induced loss of NCAM1 in the hippocampus. Both adenovirus-mediated overexpression of NCAM1 and NCAM1 mimetic peptide rescued KLK8-overexpressed neuron cells from apoptosis. Collectively, this study identified a new pro-apoptotic mechanism in the hippocampus during the pathogenesis of CUMS-induced depression via the upregulation of KLK8, and raised the possibility of KLK8 as a potential therapeutic target for depression.


Asunto(s)
Antígeno CD56 , Depresión , Hipocampo , Calicreínas , Animales , Ratones , Ratas , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Ratones Noqueados , Ratas Transgénicas , Hipocampo/metabolismo , Hipocampo/patología , Regulación hacia Arriba , Depresión/metabolismo , Depresión/patología , Neuronas/patología , Apoptosis , Biomimética , Calicreínas/metabolismo , Antígeno CD56/metabolismo
11.
Braz J Med Biol Res ; 56: e12547, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995873

RESUMEN

The main goal of this study was to determine whether oxidative imbalance mediated by AT1 receptor (AT1R) is responsible for deleterious endothelial responses to mental stress (MS) in overweight/obese class I men. Fifteen overweight/obese men (27±7 years old; 29.8±2.6 kg/m2) participated in three randomized experimental sessions with oral administration of the AT1R blocker olmesartan (40 mg; AT1R blockade) or ascorbic acid (AA; 3g) infusion or placebo [both intravenously (0.9% NaCl) and orally]. After two hours, endothelial function was determined by flow-mediated dilation (FMD) before (baseline), 30 min (30MS), and 60 min (60MS) after a five-minute acute MS session (Stroop Color Word Test). Blood was collected before (baseline), during MS, and 60 min after MS for redox homeostasis profiling: lipid peroxidation (TBARS; thiobarbituric acid reactive species), protein carbonylation, and catalase activity by colorimetry and superoxide dismutase (SOD) activity by an ELISA kit. At the placebo session, FMD significantly decreased 30MS (P=0.05). When compared to baseline, TBARS (P<0.02), protein carbonylation (P<0.01), catalase (P<0.01), and SOD (P<0.01) increased during the placebo session. During AT1R blockade, FMD increased 30 min after MS (P=0.01 vs baseline; P<0.01 vs placebo), while AA infusion increased FMD only 60 min after MS. No differences were observed during MS with the AT1R blockade and AA regarding TBARS, protein carbonylation, catalase, and SOD. AT1R-mediated redox imbalances played an important role in endothelial dysfunction to mental stress.


Asunto(s)
Obesidad , Estrés Psicológico , Humanos , Estrés Psicológico/patología , Células Endoteliales/patología , Estrés Oxidativo , Masculino , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/patología , Receptor de Angiotensina Tipo 1/metabolismo
12.
J Adolesc ; 95(4): 740-750, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36751143

RESUMEN

INTRODUCTION: Exposure to childhood trauma is found to increase internalizing and externalizing behavior problems in adolescents, however, the potential mechanism of this link remains underexplored. This study investigated the associations between childhood trauma and internalizing and externalizing behavior problems among adolescents, and tested the mediating role of executive function and the moderating role of life events stress in this relationship. METHODS: Questionnaire data were collected from 952 junior students in Northwest China. Participants ranged in age from 11 to 15 years old (M = 12.88 years, SD = 0.72; 53% females). SPSS 26.0 was used to analyze the relationship between variables and examine the mediation model and the moderated mediation model. RESULTS: Childhood trauma was positively associated with internalizing and externalizing behavior problems among adolescents. In addition, executive function partially mediated the relations between childhood trauma and internalizing and externalizing behavior problems. Life events stress was observed to moderate the relations between childhood trauma and executive function, as well as executive function and internalizing and externalizing behavior problems, but the effect sizes were relatively small. CONCLUSIONS: These findings underscore the role of executive function and life events stress in the association between childhood trauma and behavioral problems among adolescents.


Asunto(s)
Experiencias Adversas de la Infancia , Función Ejecutiva , Problema de Conducta , Estrés Psicológico , Adolescente , Niño , Femenino , Humanos , Masculino , Experiencias Adversas de la Infancia/psicología , Problema de Conducta/psicología , Estrés Psicológico/complicaciones , Estrés Psicológico/patología , Encuestas y Cuestionarios , China
13.
eNeuro ; 10(3)2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36808099

RESUMEN

Women are twice as likely as men to experience emotional dysregulation after stress, resulting in substantially higher psychopathology for equivalent lifetime stress exposure, yet the mechanisms underlying this vulnerability remain unknown. Studies suggest changes in medial prefrontal cortex (mPFC) activity as a potential contributor. Whether maladaptive changes in inhibitory interneurons participate in this process, and whether adaptations in response to stress differ between men and women, producing sex-specific changes in emotional behaviors and mPFC activity, remained undetermined. This study examined whether unpredictable chronic mild stress (UCMS) in mice differentially alters behavior and mPFC parvalbumin (PV) interneuron activity by sex, and whether the activity of these neurons drives sex-specific behavioral changes. Four weeks of UCMS increased anxiety-like and depressive-like behaviors associated with FosB activation in mPFC PV neurons, particularly in females. After 8 weeks of UCMS, both sexes displayed these behavioral and neural changes. Chemogenetic activation of PV neurons in UCMS-exposed and nonstressed males induced significant changes in anxiety-like behaviors. Importantly, patch-clamp electrophysiology demonstrated altered excitability and basic neural properties on the same timeline as the emergence of behavioral effects: changes in females after 4 weeks and in males after 8 weeks of UCMS. These findings show, for the first time, that sex-specific changes in the excitability of prefrontal PV neurons parallel the emergence of anxiety-like behavior, revealing a potential novel mechanism underlying the enhanced vulnerability of females to stress-induced psychopathology and supporting further investigation of this neuronal population to identify new therapeutic targets for stress disorders.


Asunto(s)
Ansiedad , Parvalbúminas , Masculino , Ratones , Femenino , Animales , Parvalbúminas/metabolismo , Ansiedad/patología , Neuronas/metabolismo , Trastornos de Ansiedad , Emociones , Interneuronas/fisiología , Corteza Prefrontal/metabolismo , Estrés Psicológico/patología
14.
Pharmacol Biochem Behav ; 223: 173513, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36610590

RESUMEN

Binge patterns of alcohol use, prevalent among adolescents, are associated with a higher probability of developing alcohol use disorders (AUD) and other psychiatric disorders, like anxiety and depression. Additionally, adverse life events strongly predict AUD and other psychiatric disorders. As such, the combined fields of stress and AUD have been well established, and animal models indicate that both binge-like alcohol exposure and stress exposure elevate anxiety-like behaviors. However, few have investigated the interaction of adolescent intermittent ethanol (AIE) and adult stressors. We hypothesized that AIE would increase vulnerability to restraint-induced stress (RS), manifested as increased anxiety-like behavior. After AIE exposure, in adulthood, animals were tested on forced swim (FST) and saccharin preference (SP) and then exposed to either RS (90 min/5 days) or home-cage control. Twenty-four hours after the last RS session, animals began testing on the elevated plus maze (EPM), and were re-tested on FST and SP. A separate group of animals were sacrificed in adulthood after AIE and RS, and brains were harvested for immunoblot analysis of dorsal and ventral hippocampus. Consistent with previous reports, AIE had no significant effect on closed arm time in the EPM (anxiety-like behavior). However, in male rats the interaction of AIE and adult RS increased time spent in the closed arms. No effect was observed among female animals. AIE and RS-specific alterations were found in glial and synaptic markers (GLT-1, FMRP and PSD-95) in male animals. These findings indicate AIE has sex-specific effects on both SP and the interaction of AIE and adult RS, which induces a propensity toward anxiety-like behavior in males. Also, AIE produces persistent hippocampal deficits that may interact with adult RS to cause increased anxiety-like behaviors. Understanding the mechanisms behind this AIE-induced increase in stress vulnerability may provide insight into treatment and prevention strategies for alcohol use disorders.


Asunto(s)
Ansiedad , Consumo Excesivo de Bebidas Alcohólicas , Etanol , Animales , Femenino , Masculino , Ratas , Consumo de Bebidas Alcohólicas/efectos adversos , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/patología , Consumo de Bebidas Alcohólicas/psicología , Alcoholismo , Ansiedad/etiología , Ansiedad/metabolismo , Ansiedad/patología , Ansiedad/psicología , Trastornos de Ansiedad/etiología , Trastornos de Ansiedad/metabolismo , Trastornos de Ansiedad/patología , Trastornos de Ansiedad/psicología , Consumo Excesivo de Bebidas Alcohólicas/complicaciones , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Consumo Excesivo de Bebidas Alcohólicas/patología , Consumo Excesivo de Bebidas Alcohólicas/psicología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Etanol/efectos adversos , Etanol/farmacología , Factores Sexuales , Estrés Psicológico/complicaciones , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Estrés Psicológico/psicología
15.
Horm Res Paediatr ; 96(1): 8-16, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35272295

RESUMEN

BACKGROUND: Human organisms have to cope with a large number of external or internal stressful stimuli that threaten - or are perceived as threatening - their internal dynamic balance or homeostasis. To face these disturbing forces, or stressors, organisms have developed a complex neuroendocrine system, the stress system, which consists of the hypothalamic-pituitary-adrenal axis and the locus caeruleus/norepinephrine-autonomic nervous system. SUMMARY: Upon exposure to stressors beyond a certain threshold, the activation of the stress system leads to a series of physiological and behavioral adaptations that help achieve homeostasis and increase the chances of survival. When, however, the stress response to stressors is inadequate, excessive, or prolonged, the resultant maladaptation may lead to the development of several stress-related pathologic conditions. Adverse environmental events, especially during critical periods of life, such as prenatal life, childhood, and puberty/adolescence, in combination with the underlying genetic background, may leave deep, long-term epigenetic imprints in the human expressed genome. KEY MESSAGES: In this review, we describe the components of the stress system and its functional interactions with other homeostatic systems of the organism; we present the hormonal regulators of the stress response, and we discuss the development of stress-related pathologies.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Humanos , Niño , Glucocorticoides , Norepinefrina , Homeostasis , Estrés Fisiológico , Estrés Psicológico/patología
16.
Nature ; 613(7945): 696-703, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36450985

RESUMEN

In humans, traumatic social experiences can contribute to psychiatric disorders1. It is suggested that social trauma impairs brain reward function such that social behaviour is no longer rewarding, leading to severe social avoidance2,3. In rodents, the chronic social defeat stress (CSDS) model has been used to understand the neurobiology underlying stress susceptibility versus resilience following social trauma, yet little is known regarding its impact on social reward4,5. Here we show that, following CSDS, a subset of male and female mice, termed susceptible (SUS), avoid social interaction with non-aggressive, same-sex juvenile C57BL/6J mice and do not develop context-dependent social reward following encounters with them. Non-social stressors have no effect on social reward in either sex. Next, using whole-brain Fos mapping, in vivo Ca2+ imaging and whole-cell recordings, we identified a population of stress/threat-responsive lateral septum neurotensin (NTLS) neurons that are activated by juvenile social interactions only in SUS mice, but not in resilient or unstressed control mice. Optogenetic or chemogenetic manipulation of NTLS neurons and their downstream connections modulates social interaction and social reward. Together, these data suggest that previously rewarding social targets are possibly perceived as social threats in SUS mice, resulting from hyperactive NTLS neurons that occlude social reward processing.


Asunto(s)
Vías Nerviosas , Trauma Psicológico , Recompensa , Núcleos Septales , Conducta Social , Estrés Psicológico , Animales , Femenino , Masculino , Ratones , Encéfalo/patología , Encéfalo/fisiopatología , Calcio/análisis , Calcio/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neurotensina/metabolismo , Optogenética , Trauma Psicológico/patología , Trauma Psicológico/fisiopatología , Núcleos Septales/patología , Núcleos Septales/fisiopatología , Estrés Psicológico/patología , Estrés Psicológico/fisiopatología
17.
Stress ; 26(1): 1-14, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36520154

RESUMEN

Bombesin receptor-activated protein (BRAP) and its homologous protein in mice, which is encoded by bc004004 gene, were expressed abundantly in brain tissues with unknown functions. We treated bc004004-/- mice with chronic unpredictable mild stress (CUMS) to test whether those mice were more vulnerable to stress-related disorders. The results of forced swimming test, sucrose preference test, and open field test showed that after being treated with CUMS for 28 days or 35 days both bc004004-/- and bc004004+/+ mice exhibited behavioural changes and there was no significant difference between bc004004+/+ and bc004004-/-. However, behavioural changes were observed only in bc004004-/- mice after being exposed to CUMS for 21 days, but not in bc004004+/+ after 21-day CUMS exposure, indicating that lack of BRAP homologous protein may cause vulnerability to stress-related disorders in mice. In addition, bc004004-/- mice showed a reduction in recognition memory as revealed by novel object recognition test. Since memory changes and stress related behavioural changes are all closely related to the hippocampus function we further analyzed the changes of dendrites and synapses of hippocampal neurons as well as expression levels of some proteins closely related to synaptic function. bc004004-/- mice exhibited decreased dendritic lengths and increased amount of immature spines, as well as altered expression pattern of synaptic related proteins including GluN2A, synaptophysin and BDNF in the hippocampus. Those findings suggest that BRAP homologous protein may have a protective effect on the behavioural response to stress via regulating dendritic spine formation and synaptic plasticity in the hippocampus.


Asunto(s)
Bombesina , Espinas Dendríticas , Hipocampo , Plasticidad Neuronal , Receptores de Bombesina , Estrés Psicológico , Animales , Ratones , Bombesina/genética , Bombesina/metabolismo , Enfermedad Crónica , Espinas Dendríticas/genética , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Depresión/genética , Depresión/metabolismo , Depresión/patología , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Receptores de Bombesina/genética , Receptores de Bombesina/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Estrés Psicológico/patología
18.
J Neuroinflammation ; 19(1): 240, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36183107

RESUMEN

BACKGROUND: Major depressive disorder (MDD) is a prevalent and devastating psychiatric illness. Unfortunately, the current therapeutic practice, generally depending on the serotonergic system for drug treatment is unsatisfactory and shows intractable side effects. Multiple evidence suggests that dopamine (DA) and dopaminergic signals associated with neuroinflammation are highly involved in the pathophysiology of depression as well as in the mechanism of antidepressant drugs, which is still in the early stage of study and well worthy of investigation. METHODS: We established two chronic stress models, including chronic unpredictable mild stress (CUMS), and chronic social defeat stress (CSDS), to complementarily recapitulate depression-like behaviors. Then, hippocampal tissues were used to detect inflammation-related molecules and signaling pathways. Pathological changes in depressive mouse hippocampal astrocytes were examined by RNA sequencing. After confirming the dopamine receptor 2 (Drd2)/ß-arrestin2 signaling changes in the depressive mice brain, we then established the depressive mouse model using the ß-arrestin2 knockout mice or administrating the ß-arrestin2-biased Drd2 agonist to investigate the roles. Label-free mass spectrometry was used to identify the ß-arrestin2-binding proteins as the underlying mechanisms. We modeled neuroinflammation with interleukin-6 (IL-6) and corticosterone treatment and characterized astrocytes using multiple methods including cell viability assay, flow cytometry, and confocal immunofluorescence. RESULTS: Drd2-biased ß-arrestin2 pathway is significantly changed in the progression of depression, and genetic deletion of ß-arrestin2 aggravates neuroinflammation and depressive-like phenotypes. Mechanistically, astrocytic ß-arrestin2 retains STAT3 in the cytoplasm by structural combination with STAT3, therefore, inhibiting the JAK-STAT3 pathway-mediated inflammatory activation. Furtherly, pharmacological activation of Drd2/ß-arrestin2 pathway by UNC9995 abolishes the inflammation-induced loss of astrocytes and ameliorates depressive-like behaviors in mouse model for depression. CONCLUSIONS: Drd2/ß-arrestin2 pathway is a potential therapeutic target for depression and ß-arrestin2-biased Drd2 agonist UNC9995 is identified as a potential anti-depressant strategy for preventing astrocytic dysfunctions and relieving neuropathological manifestations in mouse model for depression, which provides insights for the therapy of depression.


Asunto(s)
Astrocitos , Trastorno Depresivo Mayor , Animales , Astrocitos/metabolismo , Corticosterona/metabolismo , Depresión/tratamiento farmacológico , Depresión/etiología , Trastorno Depresivo Mayor/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Agonistas de Dopamina/farmacología , Agonistas de Dopamina/uso terapéutico , Hipocampo/metabolismo , Inflamación/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Noqueados , Receptores de Dopamina D2/metabolismo , Estrés Psicológico/complicaciones , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/patología , beta-Arrestina 1/metabolismo , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
19.
J Comp Neurol ; 530(17): 3056-3071, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35972906

RESUMEN

Depression is one of the most common mental illnesses and seriously affects all aspects of life. Running exercise has been suggested to prevent or alleviate the occurrence and development of depression; however, the underlying mechanisms of these effects remain unclear. Independent studies have indicated that astrocytes play essential roles and that the medial prefrontal cortex (mPFC) is an important brain region involved in the pathology underlying depression. However, it is unknown whether running exercise achieves antidepressant effects by affecting the number of astrocytes and glutamate transport function in the mPFC. Here, animal models of depression were established using chronic unpredictable stress (CUS), and depression-like behavior was assessed by the sucrose preference test. After successfully establishing the depression model, experimental animals performed running exercise. Glial fibrillary acidic protein-positive (GFAP+ ) cell number in the mPFC was precisely quantified using immunohistochemical and stereological methods, and the densities of bromodeoxyuridine-positive (BrdU+ ) and BrdU+ /GFAP+ cells in the mPFC were measured using a semiquantitative immunofluorescence assay. Changes in glutamate transporter gene expression in mPFC astrocytes were detected by mRNA sequencing and qRT-PCR. We found that running exercise reversed CUS-induced decreases in sucrose preference, increased astrocyte number and the density of newborn astrocytes, and reversed decreases in gene expression levels of GFAP, S100b, and the glutamate transporters GLT-1 and GLAST in the mPFC of CUS animals. These results suggested that changes in astrocyte number and glutamate transporter function may be potential meditators of the effects of running exercise in the treatment of depression.


Asunto(s)
Astrocitos , Carrera , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Sistema de Transporte de Aminoácidos X-AG/farmacología , Animales , Antidepresivos/metabolismo , Antidepresivos/farmacología , Astrocitos/metabolismo , Bromodesoxiuridina/metabolismo , Depresión/patología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Ácido Glutámico/metabolismo , Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Estrés Psicológico/patología , Sacarosa
20.
J Neuroinflammation ; 19(1): 166, 2022 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-35761401

RESUMEN

BACKGROUND: Little is known about how the obesogenic environment influences emotional states associated with glial responses and neuronal function. Here, we investigated glial reactivation and neuronal electrophysiological properties in emotion-related brain regions of high-fat diet (HFD) and ob/ob mice under chronic stress. METHODS: The glial reactivation and neuronal activities in emotion-related brain regions were analyzed among normal diet mice (ND), HFD mice, wild-type mice, and ob/ob mice. To further activate or inhibit astrocytes in medial prefrontal cortex (mPFC), we injected astrocytes specific Gq-AAV or Gi-AAV into mPFC and ongoing treated mice with CNO. RESULTS: The results showed that obesogenic factors per se had no significant effect on neuronal activities in emotion-related brain regions, or on behavioral performance. However, exposure to a chronic stressor profoundly reduced the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) and spontaneous excitatory postsynaptic currents (sEPSCs) in the mPFC; depressive-like behaviors were seen, accompanied by significant upregulation of astrocyte reactivation. We identified resilient and susceptible mice among chronic social defeat stress-exposed HFD mice. As expected, astrocyte reactivity was upregulated, while neuronal activity was depressed, in the mPFC of susceptible compared to resilient mice. Furthermore, activating astrocytes resulted in similar levels of neuronal activity and depressive-like behaviors between resilient and susceptible mice. Additionally, inhibiting astrocyte reactivation in the mPFC of HFD mice upregulated neuronal activities and inhibited depressive-like behaviors. CONCLUSIONS: These observations indicate that obesogenic factors increase the risk of depression, and improve our understanding of the pathological relationship between obesity and depression.


Asunto(s)
Astrocitos , Corteza Prefrontal , Animales , Astrocitos/patología , Depresión/etiología , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , Obesidad/patología , Corteza Prefrontal/patología , Estrés Psicológico/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA