Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Neurochem Int ; 152: 105224, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34798194

RESUMEN

CRH system integrates responses to stress challenges, whereas antipsychotics may impinge on this process. Effect of haloperidol (HAL) and aripiprazole (ARI) on chronic mild stress (CMS) induced neurobehavioral and CRH/CRHR1 system changes was studied in functionally interconnected rat brain areas including prefrontal cortex (PFC), bed nucleus of the stria terminalis (BNST), hypothalamic paraventricular nucleus (PVN), hippocampus (HIP), and amygdala (AMY). Animals were exposed to CMS for 3-weeks and since the 7th day of CMS injected with vehicle (VEH), HAL (1 mg/kg) or ARI (10 mg/kg) for 4-weeks. Expression levels of CRH, CRHR1, and c-fos genes and anxiety-like and anhedonia behavioural patterns were evaluated. CMS in VEH animals suppressed CRH gene expression in the PFC and BNST, c-fos expression in all areas, except HIP, and increased CRHR1 gene expression in the HIP. Antipsychotics decreased CRH gene expression in all areas, except HIP and by CMS elevated CRHR1 expression in the HIP (ARI also in AMY). CMS and antipsychotics decreased the sucrose preference. Aripiprazole prevented CRH expression decrease in the BNST and sucrose preference induced by CMS. Haloperidol increased time spent in the EPM open arms. These data indicate that HAL and ARI selectively influenced behavioural parameters and CRH/CRHR1 gene expression levels in CMS animals.


Asunto(s)
Aripiprazol/farmacología , Conducta Animal/efectos de los fármacos , Hormona Liberadora de Corticotropina/efectos de los fármacos , Haloperidol/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Antipsicóticos/farmacología , Ansiedad/inducido químicamente , Ansiedad/tratamiento farmacológico , Hormona Liberadora de Corticotropina/metabolismo , Hormona Liberadora de Corticotropina/farmacología , Haloperidol/metabolismo , Masculino , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo
2.
Neurogastroenterol Motil ; 33(3): e14076, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33373484

RESUMEN

BACKGROUND: Psychological stress is a risk factor for irritable bowel syndrome, a functional gastrointestinal pain disorder featuring abnormal brain-gut connectivity. The guanylate cyclase-C (GC-C) agonist linaclotide has been shown to relieve abdominal pain in IBS-C and exhibits antinociceptive effects in rodent models of post-inflammatory visceral hypersensitivity. However, the role GC-C signaling plays in psychological stress-induced visceral hypersensitivity is unknown. Here, we test the hypothesis that GC-C agonism reverses stress-induced colonic hypersensitivity via inhibition of nociceptive afferent signaling resulting in normalization of stress-altered corticotropin-releasing factor (CRF) expression in brain regions involved in pain perception and modulation. METHODS: Adult female rats were exposed to water avoidance stress or sham stress for 10 days, and the effects of linaclotide on stress-induced changes in colonic sensitivity, corticolimbic phospho-extracellular signal-regulated kinase (pERK), and CRF expression were measured using a combination of behavioral assessments, immunohistochemistry, and qRT-PCR. KEY RESULTS: Stressed rats exhibited colonic hypersensitivity and elevated corticolimbic pERK on day 11, which was inhibited by linaclotide. qRT-PCR analysis revealed dysregulated CRF expression in the medial prefrontal cortex, paraventricular nucleus of the hypothalamus, and central nucleus of the amygdala on day 28. Dysregulated CRF expression was not affected by linaclotide treatment. CONCLUSIONS AND INFERENCES: Our results demonstrate that exposure to repeated stress induces chronic colonic hypersensitivity in conjunction with altered corticolimbic activation and CRF expression. GC-C agonism attenuated stress-induced colonic hypersensitivity and ERK phosphorylation, but had no effect on CRF expression, suggesting the analgesic effects of linaclotide occur independent of stress-driven CRF gene expression in corticolimbic circuitry.


Asunto(s)
Encéfalo/efectos de los fármacos , Colon/efectos de los fármacos , Hormona Liberadora de Corticotropina/genética , Agonistas de la Guanilato Ciclasa C/farmacología , Nocicepción/efectos de los fármacos , Péptidos/farmacología , Receptores de Enterotoxina/metabolismo , Estrés Psicológico/metabolismo , Animales , Encéfalo/metabolismo , Núcleo Amigdalino Central/efectos de los fármacos , Núcleo Amigdalino Central/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Colon/metabolismo , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema Límbico/efectos de los fármacos , Sistema Límbico/metabolismo , Umbral del Dolor , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Estrés Psicológico/fisiopatología
3.
Sci Rep ; 10(1): 16050, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32994491

RESUMEN

Independent studies have observed that a paternal history of stress or trauma is associated with his children having a greater likelihood of developing psychopathologies such as anxiety disorders. This father-to-child effect is reproduced in several mouse models of stress, which have been crucial in developing a greater understanding of intergenerational epigenetic inheritance. We previously reported that treatment of C57Bl/6J male breeders with low-dose corticosterone (CORT) for 28 days prior to mating yielded increased anxiety-related behaviours in their male F1 offspring. The present study aimed to determine whether subchronic 7-day CORT treatment of male mice just prior to mating would be sufficient to induce intergenerational modifications of anxiety-related behaviours in offspring. We report that subchronic CORT treatment of male breeders reduced their week-on-week body weight gain and altered NR3C1 and CRH gene expression in the hypothalamus. There were no effects on sperm count and glucocorticoid receptor protein levels within the epididymal tissue of male breeders. Regarding the F1 offspring, screening for anxiety-related behaviours using the elevated-plus maze, light-dark box, and novelty-suppressed feeding test revealed no differences between the offspring of CORT-treated breeders compared to controls. Thus, it is crucial that future studies take into consideration the duration of exposure when assessing the intergenerational impacts of paternal health.


Asunto(s)
Ansiedad/etiología , Ansiedad/metabolismo , Herencia Paterna/genética , Animales , Trastornos de Ansiedad/etiología , Trastornos de Ansiedad/genética , Conducta Animal/efectos de los fármacos , Corticosterona/metabolismo , Corticosterona/farmacología , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/genética , Epigénesis Genética/efectos de los fármacos , Padre , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/genética , Estrés Psicológico/metabolismo
4.
Aging (Albany NY) ; 12(3): 2101-2122, 2020 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-32007953

RESUMEN

As the elderly population grows, chronic metabolic dysfunction including obesity and diabetes are becoming increasingly common comorbidities. Hypothalamic inflammation through CNS resident microglia serves as a common pathway between developing obesity and developing systemic aging pathologies. Despite understanding aging as a life-long process involving interactions between individuals and their environment, limited studies address the dynamics of environment interactions with aging or aging therapeutics. We previously demonstrated environmental enrichment (EE) is an effective model for studying improved metabolic health and overall healthspan in mice, which acts through a brain-fat axis. Here we investigated the CSF1R inhibitor PLX5622 (PLX), which depletes microglia, and its effects on metabolic decline in aging in interaction with EE. PLX in combination with EE substantially improved metabolic outcomes in middle-aged female mice over PLX or EE alone. Chronic PLX treatment depleted 75% of microglia from the hypothalamus and reduced markers of inflammation without affecting brain-derived neurotrophic factor levels induced by EE. Adipose tissue remodeling and adipose tissue macrophage modulation were observed in response to CSF1R inhibition, which may contribute to the combined benefits seen in EE with PLX. Our study suggests benefits exist from combined drug and lifestyle interventions in aged animals.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Envejecimiento/metabolismo , Vivienda para Animales , Microglía/efectos de los fármacos , Compuestos Orgánicos/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Medio Social , Tejido Adiposo/metabolismo , Animales , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Prueba de Tolerancia a la Glucosa , Hormona Liberadora de Gonadotropina/efectos de los fármacos , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inflamación/genética , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Neuropéptido Y/efectos de los fármacos , Neuropéptido Y/genética , Proopiomelanocortina/efectos de los fármacos , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transcriptoma/efectos de los fármacos , Pérdida de Peso
5.
Pediatr Int ; 61(11): 1114-1119, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31281996

RESUMEN

BACKGROUND: Late-onset circulatory collapse (LCC) is the transient development of refractory hypotension and oliguria after the early neonatal period, which may cause periventricular leukomalacia (PVL). The aim of this study was to evaluate the endogenous cortisol response to corticotrophin-releasing hormone (CRH) and determine whether it is effective for elucidating the pathology and selecting treatment in LCC. METHODS: This retrospective study examined infants admitted to the neonatal intensive care unit. Included were preterm (gestational age <34 weeks) infants who underwent CRH stimulation test and were treated for LCC with no obvious cause. Hydrocortisone (HC; 3.3-10 mg/kg) was given by bolus injection to the LCC infants. At 2 h after treatment, infants without a 20% rise in blood pressure (systolic or mean) from before treatment were defined as non-responsive to HC, and given catecholamine and/or vasopressin. RESULTS: Sixteen infants (median gestational age, 24 weeks 3 days; birthweight, 638 g) were eligible. Six of the infants had a good response to the CRH stimulation test. HC was effective in only three CRH good-response cases, and catecholamine and/or vasopressin was needed in the three other cases. HC was effective, however, for all CRH non-response cases. CONCLUSIONS: Although HC is the first-choice treatment for LCC, the CRH stimulation test facilitates prompt treatment of LCC, which may prevent PVL. The present findings help elucidate the pathology and aid in the selection of treatment for infants with LCC.


Asunto(s)
Hormona Liberadora de Corticotropina/efectos de los fármacos , Hidrocortisona/administración & dosificación , Recien Nacido Prematuro , Choque/tratamiento farmacológico , Presión Sanguínea/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Edad Gestacional , Glucocorticoides/administración & dosificación , Humanos , Recién Nacido , Unidades de Cuidado Intensivo Neonatal , Masculino , Pronóstico , Estudios Retrospectivos , Choque/diagnóstico , Choque/metabolismo
6.
Brain Behav Immun ; 77: 127-140, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30597198

RESUMEN

Posttraumatic stress disorder (PTSD) is a trauma and stressor-related disorder that is characterized by dysregulation of glucocorticoid signaling, chronic low-grade inflammation, and impairment in the ability to extinguish learned fear. Corticotropin-releasing hormone (Crh) is a stress- and immune-responsive neuropeptide secreted from the paraventricular nucleus of the hypothalamus (PVN) to stimulate the hypothalamic-pituitary-adrenal (HPA) axis; however, extra-hypothalamic sources of Crh from the central nucleus of the amygdala (CeA) and bed nucleus of the stria terminalis (BNST) govern specific fear- and anxiety-related defensive behavioral responses. We previously reported that preimmunization with a heat-killed preparation of the immunoregulatory environmental bacterium Mycobacterium vaccae NCTC 11659 enhances fear extinction in a fear-potentiated startle (FPS) paradigm. In this follow-up study, we utilized an in situ hybridization histochemistry technique to investigate Crh, Crhr1, and Crhr2 mRNA expression in the CeA, BNST, and PVN of the same rats from the original study [Fox et al., 2017, Brain, Behavior, and Immunity, 66: 70-84]. Here, we demonstrate that preimmunization with M. vaccae NCTC 11659 decreases Crh mRNA expression in the CeA and BNST of rats exposed to the FPS paradigm, and, further, that Crh mRNA expression in these regions is correlated with fear behavior during extinction training. These data are consistent with the hypothesis that M. vaccae promotes stress-resilience by attenuating Crh production in fear- and anxiety-related circuits. These data suggest that immunization with M. vaccae may be an effective strategy for prevention of fear- and anxiety-related disorders.


Asunto(s)
Hormona Liberadora de Corticotropina/efectos de los fármacos , Miedo/efectos de los fármacos , Mycobacteriaceae/inmunología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/fisiopatología , Ansiedad/terapia , Encéfalo/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Miedo/fisiología , Estudios de Seguimiento , Expresión Génica/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Inmunización/métodos , Masculino , Neuropéptidos/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Núcleos Septales
7.
Neurosci Lett ; 685: 83-89, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30125644

RESUMEN

Early life stressors, including general anesthesia, can have adverse effects on adult neural and behavioral outcomes, such as disruptions in inhibitory signaling, stress responsivity and increased risk of psychiatric disorders. Here we used a rat model to determine the effects of combined exposure to etomidate (ET) neonatal anesthesia and maternal separation on adult amygdala expression of genes for corticotropin-releasing hormone (Crh) and the chloride co-transporters Nkcc1 and Kcc2, as well as ethanol intake. Male and female Sprague-Dawley rats were subjected to 2 h of ET anesthesia on postnatal days (P) 4, 5, or 6 followed by maternal separation for 3 h on P10 (ET + SEP). During the P91-P120 period rats had daily 2 h access to three 0.05% saccharin solutions containing 0%, 5%, or 10% ethanol, followed by gene expression analyses. The ET + SEP group had increased Crh mRNA levels and Nkcc1/Kcc2 mRNA ratios in the amygdala, with greater increases in Nkcc1/Kcc2 mRNA ratios in males. A moderate increase in 5% ethanol intake was evident in the ET + SEP males, but not females, after calculation of the ratio of alcohol intake between the last week and first week of exposure. In contrast, control males tended to decrease alcohol consumption during the same period. A brief exposure to ET combined with a subsequent episode of stress early in life induced significant alterations in expression of amygdala Crh, Nkcc1 and Kcc2 with greater changes in the Cl- transporter expression in males. The possibility of increased alcohol intake in the exposed males requires further confirmation using different alcohol intake paradigms.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Etanol , Etomidato/farmacología , Consumo de Bebidas Alcohólicas/efectos adversos , Amígdala del Cerebelo/metabolismo , Anestesia/métodos , Animales , Animales Recién Nacidos , Hormona Liberadora de Corticotropina/efectos de los fármacos , Etanol/efectos adversos , Ratas Sprague-Dawley , Simportadores/efectos de los fármacos , Simportadores/metabolismo
8.
Cell Mol Neurobiol ; 38(3): 627-633, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28608000

RESUMEN

The anorexigenic molecule nesfatin-1 has recently been taken as a potential mood regulator, but the potential mechanisms remain unknown. Results of our previous study have demonstrated that nesfatin-1 could induce anxiety- and depression-like behaviors in rats, accompanied by the hyperactivity of the hypothalamic-pituitary-adrenal axis and the imbalanced mRNA expression of synaptic vesicle proteins. To explore the potential neurobiological mechanism underlying the effect of nesfatin-1 on the synaptic plasticity, the human neuroblastoma SH-SY5Y cells were cultured and treated with different concentrations of nesfatin-1 in the present study. The mRNA and protein expressions of corticotropin-releasing hormone (CRH) were measured via real-time fluorescent quantitative PCR and western blot, respectively. The protein expressions of extracellular signal-regulated kinase 1/2 (ERK1/2), phosphorylated-ERK1/2 (p-ERK1/2), and synapsin I were detected via western blot. The results confirmed that nesfatin-1 (10-9~10-7 mol/L) could up-regulate the expression of CRH. Moreover, nesfatin-1 (10-9~10-7 mol/L) could also increase the protein expressions of p-ERK1/2 and synapsin I, and these effects could be blocked by CP376395, a selective antagonist of CRH type 1 receptor (CRHR1). Furthermore, the increased expression of synapsin I induced by nesfatin-1 could also be reversed by PD98059, a specific inhibitor of the p-ERK. These results indicated that CRHR1 might mediate the effect of nesfatin-1 on the expressions of synapsin I via ERK1/2 signaling pathway.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas del Tejido Nervioso/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Sinapsinas/efectos de los fármacos , Aminopiridinas/farmacología , Línea Celular , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Flavonoides/farmacología , Humanos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nucleobindinas , Fosforilación , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Sinapsinas/metabolismo , Regulación hacia Arriba/efectos de los fármacos
9.
Anesthesiology ; 127(2): 347-354, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28542000

RESUMEN

BACKGROUND: We compared the effects of etomidate and ketamine on the hypothalamic-pituitary-adrenal axis during sepsis. METHODS: Mice (n = 5/group) were injected intraperitoneally with lipopolysaccharide (10 mg/kg) and 6 h later randomized to receive ketamine (100 mg/kg), etomidate (30 mg/kg), or saline. At two time points (12 and 48 h), messenger RNA levels of hypothalamic corticotropin-releasing hormone, pituitary proopiomelanocortin, and four adrenal enzymes (P450 side-chain cleavage, 3ß-hydroxysteroid deshydrogenase, 21-hydroxylase, and 11ß-hydroxylase) were measured by in situ hybridization (results are presented as optical density), and plasma levels of corticosterone and adrenocorticotropin hormones were measured by enzyme-linked immunosorbent assay (mean ± SD). RESULTS: At 12 h, lipopolysaccharide induced an overexpression of corticotropin-releasing hormone (32 ± 5 vs. 18 ± 6, P < 0.01), proopiomelanocortin (21 ± 3 vs. 8 ± 0.9, P < 0.0001), P450 side-chain cleavage (32 ± 4 vs. 23 ± 10, P < 0.05), 21-hydroxylase (17 ± 5 vs. 12 ± 2, P < 0.05), and 11ß-hydroxylase (11 ± 4 vs. 6 ± 0.5, P = 0.001), and an elevation of corticosterone (642 ± 165 vs. 98.3 ± 63 ng/ml, P < 0.0001). Etomidate and ketamine reduced P450 side-chain cleavage (19 ± 7 and 19 ± 3 vs. 32 ± 4, P < 0.01), 21-hydroxylase (8 ± 0.8 and 8 ± 1 vs. 17 ± 5, P < 0.001), 11ß-hydroxylase (4 ± 0.5 and 7 ± 1 vs. 11 ± 4, P < 0.001 and P < 0.05), and corticosterone (413 ± 189 and 260 ± 161 vs. 642 ± 165 ng/ml, P < 0.05 and P < 0.01). Ketamine also inhibited adrenocorticotropin hormone production (2.5 ± 3.6 vs. 36 ± 15 pg/ml, P < 0.05). At 48 h, all four adrenal enzymes were down-regulated by lipopolysaccharide administration with corticosterone levels similar to the control group. Ketamine and etomidate did not modify corticosterone plasma levels. CONCLUSIONS: Our endotoxemic model induces an initial activation of the hypothalamic-pituitary-adrenal axis, followed by a secondary inhibition of adrenal steroidogenesis processes. Ketamine and etomidate inhibit the enzyme expression and activity of the adrenal gland at the early stage.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Endotoxemia , Etomidato/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Ketamina/farmacología , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Analgésicos/farmacología , Animales , Corticosterona/sangre , Hormona Liberadora de Corticotropina/sangre , Hormona Liberadora de Corticotropina/efectos de los fármacos , Modelos Animales de Enfermedad , Etomidato/sangre , Hipnóticos y Sedantes/farmacología , Sistema Hipotálamo-Hipofisario/fisiopatología , Ketamina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Hipófiso-Suprarrenal/fisiopatología , Proopiomelanocortina/sangre , Proopiomelanocortina/efectos de los fármacos , Esteroide 21-Hidroxilasa/sangre , Esteroide 21-Hidroxilasa/efectos de los fármacos
10.
Brain Res ; 1663: 114-122, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28315311

RESUMEN

The aim of our study was to determine the role of corticotropin-releasing factor (CRF), the urocortins (Ucn1, Ucn2 and Ucn3) and their receptors (CRF1 and CRF2) in the sociability of mice. Male CFLP mice were administered intracerebroventricularly (icv) with CRF and urocortins alone or in combination with antalarmin (specific CRF1 antagonist) and astressin2B (specific CRF2 antagonist) and then investigated in a Crawley social interaction test arena, that consists of three chambers. An unknown male in a cage was put in the first chamber and an empty cage was put in the opposite chamber. The tested male was habituated with the middle chamber for 5min and then allowed to explore the remaining chambers for 5min, during which the number of entries and the time of interaction were measured. Intracerebroventricular administration of CRF decreased significantly the number of entries and the time of interaction with the unknown male and these effects were blocked by antalarmin, but not astressin2B. In contrast, central administration of Ucn1 increased significantly the number of entries into the chamber of the unknown male, without changing the time of interaction and this effect was blocked by astressin2B, but not antalarmin. Central administration of Ucn2 and Ucn3 didn't influence remarkably the number of entries, but it reduced the time of interaction between the male mice. Our study suggests that CRF and Ucn1 may play important, but different roles in sociability, and that Ucn2 and Ucn3, playing similar roles, must be also involved in social interactions.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Hormona Liberadora de Corticotropina/farmacología , Urocortinas/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Hormona Liberadora de Corticotropina/efectos de los fármacos , Masculino , Ratones , Fragmentos de Péptidos , Péptidos Cíclicos , Pirimidinas , Pirroles , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Conducta Social , Urocortinas/farmacología
11.
Psychoneuroendocrinology ; 71: 1-11, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27235634

RESUMEN

Amphetamine (AMPH)-induced appetite suppression is associated with changes in hypothalamic reactive oxygen species (ROS), antioxidants, neuropeptides, and plasma glucocorticoid. This study explored whether ROS and glucocorticoid response element (GRE), which is the promoter site of corticotropin-releasing hormone (CRH) gene, participated in neuropeptides-mediated appetite control. Rats were treated daily with AMPH for four days, and changes in food intake, plasma glucocorticoid and expression levels of hypothalamic neuropeptide Y (NPY), proopiomelanocortin (POMC), superoxide dismutase (SOD), CRH, and glucocorticoid receptor (GR) were examined and compared. Results showed that food intake decreased and NPY gene down-regulated, while POMC, SOD, and CRH gene up-regulated during AMPH treatment. GR and GRE-DNA bindings were disrupted on Day 1 and Day 2 when glucocorticoid levels were still high. Pretreatment with GR inhibitor or ROS scavenger modulated mRNA levels in NPY, POMC, SOD and CRH in AMPH-treated rats. We suggest that disruptions of negative GRE (nGRE) on Day 1 and Day 2 are associated with an increase in oxidative stress during the regulation of NPY/POMC-mediated appetite control in AMPH-treated rats. These results advance the understanding of molecular mechanism in regulating AMPH-mediated appetite suppression.


Asunto(s)
Anfetamina/farmacología , Glucocorticoides/metabolismo , Anfetamina/efectos adversos , Animales , Depresores del Apetito/farmacología , Química Encefálica/efectos de los fármacos , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/sangre , Glucocorticoides/fisiología , Hipotálamo/metabolismo , Masculino , Neuropéptido Y/metabolismo , Estrés Oxidativo/fisiología , Proopiomelanocortina/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/efectos de los fármacos , Elementos de Respuesta , Superóxido Dismutasa/metabolismo
12.
Neuropsychobiology ; 73(3): 184-90, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27221315

RESUMEN

Vasopressin and CRH have complementary roles in the secretion of ACTH following different stress modalities. The concomitant use of V1b and CRF1 receptor antagonists completely inhibits ACTH secretion in response to different stress modalities. The combination of the CRF1 antagonist SSR125543 with the V1b antagonist SSR149415 effectively suppressed plasma ACTH 1.30 h after injection in rats stressed by ether vapor inhalation for 1 min, restraint stress for 1 h or forced swimming for 5 min. The duration of the effect was also studied. The CRF1 antagonist effectively suppressed ACTH secretion in restraint stress, while the V1b antagonist was effective against ether inhalation. Both antagonists were necessary to block the forced swimming stress response. SSR125543 induced a prolonged effect and can be used in a model of prolonged HPA axis blockade.


Asunto(s)
Hormona Adrenocorticotrópica/efectos de los fármacos , Antagonistas de los Receptores de Hormonas Antidiuréticas/farmacología , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hidrocarburos Halogenados/farmacología , Indoles/farmacología , Pirrolidinas/farmacología , Estrés Psicológico/metabolismo , Tiazinas/farmacología , Vasopresinas/efectos de los fármacos , Administración por Inhalación , Hormona Adrenocorticotrópica/metabolismo , Anestésicos por Inhalación/farmacología , Animales , Hormona Liberadora de Corticotropina/metabolismo , Éter/farmacología , Sistema Hipotálamo-Hipofisario , Masculino , Modelos Animales , Sistema Hipófiso-Suprarrenal , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Vasopresinas/metabolismo , Restricción Física , Natación , Vasopresinas/metabolismo
13.
Neuroreport ; 27(8): 580-6, 2016 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-27022819

RESUMEN

Nicotine is known to modulate the activity of the hypothalamic-pituitary-adrenal axis by stimulating corticotropin-releasing hormone (CRH) release from the hypothalamic paraventricular nucleus (PVN). However, the mechanism by which nicotine affects the hypothalamic-pituitary-adrenal axis by modulating PVN CRH neuronal activity is currently unclear. Here, we examined the effects of nicotine on PVN CRH-mRNA-expressing neurons in vitro in rats by whole-cell patch-clamp recordings, biocytin staining, and single-cell reverse transcription-multiplex PCR techniques. Of the 146 PVN putative parvocellular neurons, 17.1% (25/146) coexpressed GAPDH mRNA and CRH mRNA. Under current-clamp recording conditions, application of nicotine (1 µM) induced excitation in 92% (23/25) PVN CRH-mRNA-expressing neurons, which showed a significant increase in the spike firing rate accompanied by a depolarization of the membrane potential. Nicotine induced an increase in the spike firing rate of PVN CRH-mRNA-expressing neurons in a concentration-dependent manner. The half-effective concentration (EC50) of nicotine for increasing the spike firing rate of PVN CRH-mRNA-expressing neurons was 1.6 µM. Extracellular application of ionotropic glutamate receptor antagonist kynurenic acid (1 mM) abolished the nicotine-induced excitation of PVN CRH-mRNA-expressing neurons. Moreover, application of nicotine induced a significant increase in the spontaneous excitatory postsynaptic currents frequency, but without significantly altering the spontaneous excitatory postsynaptic currents amplitude of the CRH-mRNA-expressing neurons. Biocytin staining confirmed that the nicotine-sensitive CRH-mRNA-expressing neurons were located in the PVN parvocellular division. These results indicated that extracellular administration of nicotine indirectly excited PVN CRH-mRNA-expressing neurons, suggesting that nicotine modulated PVN CRH secretion by enhancement of both the presynaptic action potential drive and quantal glutamate release.


Asunto(s)
Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Neuronas/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Animales , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Lisina/análogos & derivados , Masculino , Reacción en Cadena de la Polimerasa Multiplex , Neuronas/metabolismo , Nicotina/administración & dosificación , Nicotina/farmacocinética , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/farmacocinética , Técnicas de Placa-Clamp , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Coloración y Etiquetado
14.
Psychoneuroendocrinology ; 65: 1-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26685709

RESUMEN

Predominantly emotional stressors activate a wide range of brain areas, as revealed by the expression of immediate early genes, such as c-fos. Chlorella vulgaris (CV) is considered a biological response modifier, as demonstrated by its protective activities against infections, tumors and stress. We evaluated the effect of acute pretreatment with CV on the peripheral and central responses to forced swimming stress in adult male rats. Pretreatment with CV produced a significant reduction of stress-related hypothalamic-pituitary-adrenal activation, demonstrated by decreased corticotrophin releasing factor gene expression in the hypothalamic paraventricular nucleus (PVN) and lower ACTH response. Hyperglycemia induced by the stressor was similarly reduced. This attenuated neuroendocrine response to stress occurred in parallel with a diminished c-fos expression in most evaluated areas, including the PVN. The data presented in this study reinforce the usefulness of CV to diminish the impact of stressors, by reducing the HPA response. Although our results suggest a central effect of CV, further studies are necessary to understand the precise mechanisms underpinning this effect.


Asunto(s)
Encéfalo/fisiología , Chlorella vulgaris , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/fisiología , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Estrés Fisiológico/fisiología , Hormona Adrenocorticotrópica/efectos de los fármacos , Hormona Adrenocorticotrópica/metabolismo , Animales , Encéfalo/metabolismo , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Genes fos , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/metabolismo , Hibridación in Situ , Masculino , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Natación
15.
Psychoneuroendocrinology ; 63: 94-101, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26433325

RESUMEN

There have been numerous studies into the interaction between stress and addictive drugs, yet few have specifically addressed how the organism responds to stress when under the influence of psychostimulants. Thus, we studied the effects of different acute stressors (immobilization, interleukin-1ß and forced swimming) in young adult male rats simultaneously exposed to amphetamine (AMPH, 4 mg/kg SC), evaluating classic biological markers. AMPH administration itself augmented the plasma hypothalamic-pituitary-adrenal (HPA) hormones, adrenocorticotropin (ACTH) and corticosterone, without affecting plasma glucose levels. By contrast, this drug dampened the peripheral HPA axis, as well as the response of glucose to the three stressors. We also found that AMPH administration completely blocked the forced swim-induced expression of the corticotropin-releasing hormone (hnCRH) and it partially reduced c-fos expression in the paraventricular nucleus of the hypothalamus (PVN). Indeed, this negative synergy in the forced swim test could even be observed with a lower dose of AMPH (1mg/kg, SC), a dose that is usually received in self-administration experiments. In conclusion, when rats that receive AMPH are subjected to stress, a negative synergy occurs that dampens the prototypic peripheral physiological response to stress and activation of the PVN.


Asunto(s)
Hormona Adrenocorticotrópica/efectos de los fármacos , Anfetamina/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Hormona Liberadora de Corticotropina/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Interleucina-1beta/farmacología , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Estrés Psicológico/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Sinergismo Farmacológico , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Restricción Física , Natación
16.
Psychoneuroendocrinology ; 63: 50-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26415118

RESUMEN

Oxytocin (Oxt) is released in various hypothalamic and extrahypothalamic brain areas in response to anxiogenic stimuli to regulate aspects of emotionality and stress coping. We examined the anxiolytic action of Oxt in the hypothalamic paraventricular nucleus (PVN) while appraising if Oxt recruits GABA neurons to inhibit the behavioral, hormonal, and neuronal response to stress in female prairie voles (Microtus ochrogaster). Voles received an injection of Oxt in the PVN either before or after an elevated platform stress to determine a time-course for the effects of Oxt on the hormonal stress response. Subsequently, we evaluated if ante-stress injections of Oxt affected anxiety-like behaviors as well as neuronal activity in the PVN, using real-time in-vivo retrodialysis and immunohistochemistry with c-Fos expression as a biomarker of neural activity. In addition, we exposed voles to Oxt and a GABAA receptor antagonist, concurrently, to evaluate the impact of pharmacological blockade of GABAA receptors on the anxiolytic effects of Oxt. Elevated platform stress amplified anxiety-like behaviors and hypothalamic-pituitary-adrenal (HPA) axis activity-catalyzing corticotrophin-releasing hormone (CRH) neuronal activity and augmenting corticosterone release in circulation. Ante-stress Oxt injections in the PVN blocked these stress effects while promoting PVN GABA activity and release. Post-stress Oxt treatments were ineffective. The anxiolytic effects of Oxt were hindered by concurrent pharmacological blockade of GABAA receptors. Together, our data demonstrate ante-stress treatments of Oxt in the PVN inhibit stress activation of the HPA axis through recruitment of GABAergic neurons, providing insights to the local circuitry and potential therapeutically-relevant mechanisms.


Asunto(s)
Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Agonistas de Receptores de GABA-A/farmacología , Oxitócicos/farmacología , Oxitocina/farmacología , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de GABA-A/metabolismo , Animales , Arvicolinae , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Antagonistas de Receptores de GABA-A/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Inmunohistoquímica , Microdiálisis , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo
17.
Brain Res Bull ; 123: 81-93, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26655889

RESUMEN

One of the most damaging aspects of drug addiction is the degree to which natural rewards (family, friends, employment) are devalued in favor of seeking, obtaining and taking drugs. We have utilized an animal model of reward devaluation and heroin self-administration to explore the role of the coricotropin releasing factor (CRF) pathway. Given access to a saccharin cue followed by the opportunity to self-administer heroin, animals will parse into distinct phenotypes that suppress their saccharin intake (in favor of escalating heroin self-administration) or vice versa. We find that large saccharin suppressors (large heroin takers) demonstrate increased mRNA expression for elements of the CRF signaling pathway (CRF, CRF receptors and CRF binding protein) within the hippocampus, medial prefrontal cortex and the ventral tegmental area. Moreover, there were no gene expression changes of these components in the nucleus accumbens. Use of bisulfite conversion sequencing suggests that changes in CRF binding protein and CRF receptor gene expression may be mediated by differential promoter methylation.


Asunto(s)
Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Dependencia de Heroína/psicología , Área Tegmental Ventral/efectos de los fármacos , Animales , Hormona Liberadora de Corticotropina/farmacología , Señales (Psicología) , Heroína/metabolismo , Heroína/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Corteza Prefrontal/efectos de los fármacos , Ratas , Recompensa , Sacarina/administración & dosificación , Autoadministración , Área Tegmental Ventral/metabolismo
18.
Alcohol ; 49(3): 179-84, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25716197

RESUMEN

The central amygdala is a critical brain region for many aspects of alcohol dependence. Much of the work examining the mechanisms by which the central amygdala mediates the development of alcohol dependence has focused on the interaction of acute and chronic ethanol with central amygdala corticotropin releasing factor signaling. This work has led to a great deal of success in furthering the general understanding of central amygdala neurocircuitry and its role in alcohol dependence. Much of this work has primarily focused on the hypothesis that ethanol utilizes endogenous corticotropin releasing factor signaling to upregulate inhibitory GABAergic transmission in the central amygdala. Work that is more recent suggests that corticotropin releasing factor also plays an important role in mediating anxiety-like behaviors via the enhancement of central amygdala glutamatergic transmission, implying that ethanol/corticotropin releasing factor interactions may modulate excitatory neurotransmission in this brain region. In addition, a number of studies utilizing optogenetic strategies or transgenic mouse lines have begun to examine specific central amygdala neurocircuit dynamics and neuronal subpopulations to better understand overall central amygdala neurocircuitry and the role of neuronal subtypes in mediating anxiety-like behaviors. This review will provide a brief update on this literature and describe some potential future directions that may be important for the development of better treatments for alcohol addiction.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Hormona Liberadora de Corticotropina/efectos de los fármacos , Etanol/farmacología , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Hormona Liberadora de Corticotropina/metabolismo , Ácido Glutámico/efectos de los fármacos , Ácido Glutámico/metabolismo , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
19.
J Neurochem ; 131(6): 825-35, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25087915

RESUMEN

Melanocortin receptor four (MC4R) is implicated in regulation of stress-related functions. We previously demonstrated that intranasal infusion of MC4R antagonist HS014, shortly before single prolonged stress (SPS) animal model of post-traumatic stress disorder, lessened the development of anxiety- and depression-like behavior depending on the dose. Here, we evaluated effects of HS014 on SPS-elicited changes in hypothalamic-pituitary-adrenal axis and expression of several genes of interest in mediobasal hypothalamus, hippocampus, and locus coeruleus. Rats were given intranasal infusion of HS014 (3.5 ng or 100 µg) and 30 min later subjected to SPS stressors. Short-term responses of HS014 rats in comparison with vehicle-treated, evident 30 min following SPS stressors, included smaller rise in plasma corticosterone (100 µg HS014), absence of induction of corticotrophin-releasing hormone mRNA in mediobasal hypothalamus and of mRNA for tyrosine hydroxylase and dopamine-ß hydroxylase in locus coeruleus. Long-term responses found 7 days after SPS stressors, included lower induction corticotrophin-releasing hormone mRNA levels in the mediobasal hypothalamus without effect on mRNAs for the glucocorticoid receptor (GR) and FK506-binding protein 51 (FKBP5), a component of GR co-chaperone complex; and no induction of GR protein in ventral hippocampus. Thus, antagonism of MC4R prior to SPS attenuates development of several abnormalities in gene expression in regions implicated in post-traumatic stress disorder. Blockade of brain melanocortine receptor 4 (MC4R) with intranasal infusion of the MC4R antagonist HS014 to rats prior to single prolonged stress (SPS) leads to faster termination of stress responses (30 min later) and prevents or attenuates SPS-triggered abnormal gene expression related to post-traumatic stress disorder (7 days later). Targeting of brain MC4R is a promising strategy to protect HPA axis, LC-NE (locus coeruleus-norepinephrine) systems and hippocampus from overstimulation.


Asunto(s)
Hipocampo/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Péptidos Cíclicos/farmacología , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Receptor de Melanocortina Tipo 4/efectos de los fármacos , Administración Intranasal , Animales , Ansiedad/metabolismo , Corticosterona/sangre , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Péptidos Cíclicos/administración & dosificación , Sistema Hipófiso-Suprarrenal/metabolismo , Ratas Sprague-Dawley , Receptores de Glucocorticoides/efectos de los fármacos , Estrés Psicológico/metabolismo
20.
Addict Biol ; 19(4): 606-11, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23362976

RESUMEN

Corticotrophin-releasing factor (CRF) is a mediator of stress responses and a key modulator of ethanol-mediated behaviors. We report here that the CRF receptor 1 (CRF-R1) antagonist, CP-376395 reduces 20% ethanol consumption in animals trained to consume ethanol on an intermittent, but not a continuous, schedule. Furthermore, using [(35) S]GTPγS binding assays, we demonstrate that CRF-mediated G-protein signaling in the hypothalamus of the intermittent drinkers is decreased when compared to controls suggesting that the effects of CP-376395 are mediated by extrahypothalamic mechanisms. The present study provides further support for the use of CRF-R1 antagonists for the treatment of alcohol use disorders and suggests that ethanol consumption dysregulates CRF function in the hypothalamus.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Aminopiridinas/farmacología , Depresores del Sistema Nervioso Central/farmacología , Hormona Liberadora de Corticotropina/efectos de los fármacos , Etanol/farmacología , Hipotálamo/fisiopatología , Análisis de Varianza , Animales , Depresores del Sistema Nervioso Central/administración & dosificación , Etanol/administración & dosificación , Hipotálamo/efectos de los fármacos , Masculino , Modelos Animales , Ratas , Ratas Long-Evans
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...