Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(9)2021 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-34068684

RESUMO

Results over the last decades have provided evidence suggesting that HPA axis dysfunction is a major risk factor predisposing to the development of psychopathological behaviour. This susceptibility can be programmed during developmental windows of marked neuroplasticity, allowing early-life adversity to convey vulnerability to mental illness later in life. Besides genetic predisposition, also environmental factors play a pivotal role in this process, through embodiment of the mother's emotions, or via nutrients and hormones transferred through the placenta and the maternal milk. The aim of the current translational study was to mimic a severe stress condition by exposing female CD-1 mouse dams to abnormal levels of corticosterone (80 µg/mL) in the drinking water either during the last week of pregnancy (PreCORT) or the first one of lactation (PostCORT), compared to an Animal Facility Rearing (AFR) control group. When tested as adults, male mice from PostCORT offspring and somewhat less the PreCORT mice exhibited a markedly increased corticosterone response to acute restraint stress, compared to perinatal AFR controls. Aberrant persistence of adolescence-typical increased interest towards novel social stimuli and somewhat deficient emotional contagion also characterised profiles in both perinatal-CORT groups. Intranasal oxytocin (0 or 20.0 µg/kg) generally managed to reduce the stress response and restore a regular behavioural phenotype. Alterations in density of glucocorticoid and mineralocorticoid receptors, oxytocin and µ- and κ-opioid receptors were found. Changes differed as a function of brain areas and the specific age window of perinatal aberrant stimulation of the HPA axis. Present results provided experimental evidence in a translational mouse model that precocious adversity represents a risk factor predisposing to the development of psychopathological behaviour.


Assuntos
Corticosterona/genética , Ocitocina/genética , Receptores Opioides mu/genética , Estresse Psicológico/genética , Animais , Emoções/fisiologia , Feminino , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Camundongos , Sistema Hipófise-Suprarrenal/metabolismo , Angústia Psicológica , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética , Estresse Psicológico/imunologia
2.
Int J Mol Sci ; 22(9)2021 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-34068707

RESUMO

BDNF plays a pivotal role in neuroplasticity events, vulnerability and resilience to stress-related disorders, being decreased in depressive patients and increased after antidepressant treatment. BDNF was found to be reduced in patients carrying the human polymorphism in the serotonin transporter promoter region (5-HTTLPR). The serotonin knockout rat (SERT-/-) is one of the animal models used to investigate the underlying molecular mechanisms of depression in humans. They present decreased BDNF levels, and anxiety- and depression-like behavior. To investigate whether upregulating BDNF would ameliorate the phenotype of SERT-/- rats, we overexpressed BDNF locally into the ventral hippocampus and submitted the animals to behavioral testing. The results showed that BDNF overexpression in the vHIP of SERT-/- rats promoted higher sucrose preference and sucrose intake; on the first day of the sucrose consumption test it decreased immobility time in the forced swim test and increased the time spent in the center of a novel environment. Furthermore, BDNF overexpression altered social behavior in SERT-/- rats, which presented increased passive contact with test partner and decreased solitary behavior. Finally, it promoted decrease in plasma corticosterone levels 60 min after restraint stress. In conclusion, modulation of BDNF IV levels in the vHIP of SERT-/- rats led to a positive behavioral outcome placing BDNF upregulation in the vHIP as a potential target to new therapeutic approaches to improve depressive symptoms.


Assuntos
Transtornos de Ansiedade/tratamento farmacológico , Fator Neurotrófico Derivado do Encéfalo/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Estresse Psicológico/tratamento farmacológico , Animais , Ansiolíticos/farmacologia , Antidepressivos/farmacologia , Transtornos de Ansiedade/genética , Transtornos de Ansiedade/patologia , Corticosterona/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Plasticidade Neuronal/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ratos , Serotonina/genética , Estresse Psicológico/genética , Estresse Psicológico/patologia
3.
Artigo em Inglês | MEDLINE | ID: mdl-32810572

RESUMO

Chronic stress is the leading risk factor of a broad range of severe psychopathologies. Nonetheless, the molecular mechanisms triggering these pathological processes are not well understood. In our study, we investigated the effects of 15-day social defeat stress (SDS) on the genome-wide landscape of trimethylation at the 4th lysine residue of histone H3 (H3K4me3) and on the transcriptome in the prefrontal cortex of mice that were reared normally (group SDS) or subjected to maternal separation early in life (group MS+SDS). The mice with the history of stress early in life showed increased susceptibility to SDS in adulthood and demonstrated long-lasting genome-wide alterations in gene expression and splicing as well as in the H3K4me3 epigenetic landscape in the prefrontal cortex. Thus, the high-throughput techniques applied here allowed us to simultaneously detect, for the first time, genome-wide epigenetic and transcriptional changes in the murine prefrontal cortex that are associated with both chronic SDS and increased susceptibility to this stressor.


Assuntos
Epigênese Genética/fisiologia , Histonas/metabolismo , Privação Materna , Córtex Pré-Frontal/metabolismo , Derrota Social , Estresse Psicológico/metabolismo , Fatores Etários , Processamento Alternativo/fisiologia , Animais , Corticosterona/genética , Corticosterona/metabolismo , Feminino , Expressão Gênica , Histonas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Psicológico/genética , Estresse Psicológico/psicologia
4.
PLoS One ; 15(9): e0239231, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32997686

RESUMO

It is controversially discussed whether immune-deficient mice experience severity in the absence of infection. Because a comprehensive analysis of the well-being of immune-deficient mice under specific pathogen free conditions is missing, we used a multi-parametric test analyzing, corticosterone, weight, nest building and facial expression over a period of 9 month to determine the well-being of two immune-deficient mouse lines (recombination activating gene 2- and interferon gamma receptor-deficient mice). We do not find evidence for severity when comparing immune-deficient mice to their heterozygous immune-competent littermates. Our data challenge the assumption that immune-deficiency per se regardless of housing conditions causes severity. Based on our study we propose to use objective non-invasive parameters determined by laboratory animal science for decisions concerning severity of immune-deficient mice.


Assuntos
Corticosterona/genética , Proteínas de Ligação a DNA/genética , Interferon gama/genética , Camundongos SCID/genética , Animais , Linfócitos B/imunologia , Corticosterona/imunologia , Humanos , Infecções/genética , Infecções/imunologia , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos SCID/imunologia , Dor/genética , Dor/patologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Transdução de Sinais/genética , Linfócitos T/imunologia , Testosterona/genética , Receptor de Interferon gama
5.
J Neuroendocrinol ; 32(5): e12852, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32364267

RESUMO

Glucocorticoids (CORT) are well-known as important regulators of behaviour and cognition at basal levels and under stress. However, the precise mechanisms governing CORT action and functional outcomes of this action in the brain remain unclear, particularly in model systems other than rodents. In the present study, we investigated the dynamics of CORT regulation in the zebra finch, an important model system for vocal learning, neuroplasticity and cognition. We tested the hypothesis that CORT is locally regulated in the zebra finch brain by quantifying regional and stress-related variation in total CORT across brain regions. In addition, we used an ex vivo slice culture system to test whether CORT regulates target gene expression uniquely in discrete regions of the brain. We documented a robust increase in brain CORT across regions after 30 minutes of restraint stress but, interestingly, baseline and stress-induced CORT levels varied between regions. In addition, CORT treatment of brain slice cultures differentially affected expression of three CORT target genes: it up-regulated expression of FKBP5 in most regions and SGK1 in the hypothalamus only, whereas GILZ was unaffected by CORT treatment across all brain regions investigated. The specific mechanisms producing regional variation in CORT and CORT-dependent downstream gene expression remain unknown, although these data provide additional support for the hypothesis that the songbird brain employs regulatory mechanisms that result in precise control over the influence of CORT on glucocorticoid-sensitive neural circuits.


Assuntos
Encéfalo/metabolismo , Corticosterona/metabolismo , Tentilhões/metabolismo , Regulação da Expressão Gênica , Estresse Fisiológico/fisiologia , Animais , Corticosterona/genética , Tentilhões/genética , Expressão Gênica
6.
Horm Behav ; 122: 104747, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32217065

RESUMO

Consistent between-individual differences in behaviour have been documented across the animal kingdom. Such variation between individuals has been shown to be the basis for selection and to act as a pacemaker for evolutionary change. Recently, equivocal evidence suggests that such consistent between-individual variation is also present in hormones. This observation has sparked interest in understanding the mechanisms shaping individual differences, temporal consistency and heritability of hormonal phenotypes and to understand, if and to what extent hormonal mechanisms are involved in mediating consistent variation in behaviour between individuals. Here, we used zebra finches of the fourth generation of bi-directionally selected lines for three independent behaviours: aggression, exploration and fearlessness. We investigated how these behaviours responded to artificial selection and tested their repeatability. We further tested for repeatability of corticosterone and testosterone across and within lines. Moreover, we are presenting the decomposed variance components for within-individual variance (i.e. flexibility) and between-individual variance (i.e. more or less pronounced differences between individuals) and investigate their contribution to repeatability estimates. Both hormones as well as the exploration and fearlessness but not aggressiveness, were repeatable. However, variance components and hence repeatability differed between lines and were often lower than in unselected control animals, mainly because of a reduction in between-individual variance. Our data show that artificial selection (including active selection and genetic drift) can affect the mean and variance of traits. We stress the importance for understanding how variable a trait is both between and within individuals to assess the selective value of a trait.


Assuntos
Agressão/fisiologia , Corticosterona/sangue , Medo , Tentilhões/fisiologia , Testosterona/sangue , Adaptação Psicológica/fisiologia , Animais , Comportamento Animal , Corticosterona/genética , Comportamento Exploratório/fisiologia , Medo/fisiologia , Medo/psicologia , Feminino , Tentilhões/sangue , Tentilhões/genética , Hierarquia Social , Masculino , Personalidade/genética , Personalidade/fisiologia , Fenótipo , Característica Quantitativa Herdável , Seleção Artificial , Territorialidade , Testosterona/genética
7.
Heredity (Edinb) ; 123(3): 337-348, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30837668

RESUMO

The hypothalamic-pituitary-adrenal (HPA) axis is responsible for the regulation of corticosterone, a hormone that is essential in the mediation of energy allocation and physiological stress. As a continuous source of challenge and stress for organisms, the environment has promoted the evolution of physiological adaptations and led to a great variation in corticosterone profiles within or among individuals, populations and species. In order to evolve via natural selection, corticosterone levels do not only depend on the strength of selection exerted on them, but also on the extent to which the regulation of corticosterone is heritable. Nevertheless, the heritability of corticosterone profiles in wild populations is still poorly understood. In this study, we estimated the heritability of baseline and stress-induced corticosterone levels in barn owl (Tyto alba) nestlings from 8 years of data, using a multivariate animal model based on a behavioural pedigree. We found that baseline and stress-induced corticosterone levels are strongly genetically correlated (r = 0.68-0.80) and that the heritability of stress-induced corticosterone levels (h2 = 0.24-0.33) was moderate and similar to the heritability of baseline corticosterone levels (h2 = 0.19-0.30). These findings suggest that the regulation of stress-induced corticosterone and baseline levels evolves at a similar pace when selection acts with the same intensity on both traits and that contrary to previous studies, the evolution of baseline and stress-induced level is interdependent in barn owls, as they may be strongly genetically correlated.


Assuntos
Adaptação Fisiológica/genética , Corticosterona/genética , Padrões de Herança , Característica Quantitativa Herdável , Estrigiformes/genética , Animais , Comportamento Animal/fisiologia , Evolução Biológica , Corticosterona/sangue , Feminino , Masculino , Modelos Genéticos , Fenótipo , Seleção Genética , Estresse Fisiológico/genética , Estrigiformes/sangue , Suíça
8.
PLoS One ; 13(5): e0197094, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29813096

RESUMO

In occidental societies, high fat and high sugar diets often coincide with episodes of stress. The association is likely to modify brain energy control. Brain insulin signalling is rarely studied in stressed individuals consuming high fat diets. Furthermore the effects of cinnamon supplement are not known in these conditions. Therefore, we exposed rats, over a 12-week period, to a control (C) or a high fat/high fructose (HF/HFr) diet that induces peripheral insulin resistance. A cinnamon supplement (C+CN and HF/HFr +CN) was added or not. After diet exposure, one group of rats was exposed to a 30-min restraint followed by a 10-min open-field test, their combination featuring a moderate stressor, the other rats staying unstressed in their home cages. The insulin signalling in hippocampus and frontal cortex was studied through the mRNA expression of the following genes: insulin receptor (Ir), insulin receptor substrate (Irs1), glucose transporters (Glut1 and Glut3), glycogen synthase (Gys1) and their modulators, Akt1 and Pten. In C rats, stress enhanced the expression of Ir, Irs1, Glut1, Gys1 and Akt1 mRNA. In C+CN rats, stress induced an increase in Pten but a decrease in Gys1 mRNA expression. In HF/HFr rats, stress was associated with an increase in Pten mRNA expression. In HF/HFr+CN rats, stress increased Pten mRNA expression but also decreased Gys1 mRNA expression. This suggests that a single moderate stress favours energy refilling mechanisms, an effect blunted by a previous HF/HFr diet and cinnamon supplement.


Assuntos
Encéfalo/metabolismo , Dieta Hiperlipídica/efeitos adversos , Extratos Vegetais/administração & dosagem , RNA Mensageiro/genética , Estresse Psicológico/metabolismo , Animais , Cinnamomum zeylanicum/química , Corticosterona/genética , Corticosterona/metabolismo , Dieta Ocidental/efeitos adversos , Suplementos Nutricionais , Frutose/administração & dosagem , Insulina/fisiologia , Resistência à Insulina , Masculino , RNA Mensageiro/metabolismo , Ratos Wistar , Transdução de Sinais , Transcriptoma
9.
FASEB J ; 32(6): 3321-3335, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29401624

RESUMO

The insulin family of growth factors (insulin, IGF1, and IGF2) are critical in sex determination, adrenal differentiation, and testicular function. Notably, the IGF system has been reported to mediate the proliferation of steroidogenic cells. However, the precise role and contribution of the membrane receptors mediating those effects, namely, insulin receptor (INSR) and type-I insulin-like growth factor receptor (IGF1R), have not, to our knowledge, been investigated. We show here that specific deletion of both Insr and Igf1r in steroidogenic cells in mice leads to severe alterations of adrenocortical and testicular development. Double-mutant mice display drastic size reduction of both adrenocortex and testes, with impaired corticosterone, testosterone, and sperm production. Detailed developmental analysis of the testes revealed that fetal Leydig cell (LC) function is normal, but there is a failure of adult LC maturation and steroidogenic function associated with accumulation of progenitor LCs (PLCs). Cell-lineage tracing revealed PLC enrichment is secondary to Insr and Igf1r deletion in differentiated adult LCs, suggesting a feedback mechanism between cells at different steps of differentiation. Taken together, these data reveal the cell-autonomous and nonautonomous roles of the IGF system for proper development and maintenance of steroidogenic lineages.-Neirijnck, Y., Calvel, P., Kilcoyne, K. R., Kühne, F., Stévant, I., Griffeth, R. J., Pitetti, J.-L., Andric, S. A., Hu, M.-C., Pralong, F., Smith, L. B., Nef, S. Insulin and IGF1 receptors are essential for the development and steroidogenic function of adult Leydig cells.


Assuntos
Diferenciação Celular , Células Intersticiais do Testículo/metabolismo , Receptor de Insulina/metabolismo , Receptores de Somatomedina/metabolismo , Células-Tronco/metabolismo , Córtex Suprarrenal/citologia , Córtex Suprarrenal/metabolismo , Animais , Corticosterona/genética , Corticosterona/metabolismo , Células Intersticiais do Testículo/citologia , Masculino , Camundongos , Camundongos Knockout , Receptor de Insulina/genética , Receptores de Somatomedina/genética , Células-Tronco/citologia , Testosterona/genética , Testosterona/metabolismo
10.
Can J Physiol Pharmacol ; 94(4): 363-72, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26761434

RESUMO

Phenylethanolamine N-methyltransferase (PNMT) is the terminal enzyme in the catecholamine biosynthetic pathway responsible for adrenaline biosynthesis. Adrenaline is involved in the sympathetic control of blood pressure; it augments cardiac function by increasing stroke volume and cardiac output. Genetic mapping studies have linked the PNMT gene to hypertension. This study examined the expression of cardiac PNMT and changes in its transcriptional regulators in the spontaneously hypertensive (SHR) and wild type Wistar-Kyoto (WKY) rats. SHR exhibit elevated levels of corticosterone, and lower levels of the cytokine IL-1ß, revealing systemic differences between SHR and WKY. PNMT mRNA was significantly increased in all chambers of the heart in the SHR, with the greatest increase in the right atrium. Transcriptional regulators of the PNMT promoter show elevated expression of Egr-1, Sp1, AP-2, and GR mRNA in all chambers of the SHR heart, while protein levels of Sp1, Egr-1, and GR were elevated only in the right atrium. Interestingly, only AP-2 protein-DNA binding was increased, suggesting it may be a key regulator of cardiac PNMT in SHR. This study provides the first insights into the molecular mechanisms involved in the dysregulation of cardiac PNMT in a genetic model of hypertension.


Assuntos
Expressão Gênica/genética , Átrios do Coração/metabolismo , Hipertensão/genética , Feniletanolamina N-Metiltransferase/metabolismo , Animais , Pressão Sanguínea/genética , Corticosterona/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Epinefrina , Regulação da Expressão Gênica , Hipertensão/metabolismo , Imunoglobulinas/genética , Interleucina-1beta/genética , Regiões Promotoras Genéticas/genética , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transcrição Gênica/genética
11.
Neuropharmacology ; 93: 116-23, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25656477

RESUMO

Previously we demonstrated that exposure of the central nucleus of the amygdala (CeA) to elevated corticosterone (CORT) induces nociceptive behaviors that are reversed by glucocorticoid and/or mineralocorticoid (GR/MR) receptor antagonism. Here we test the hypothesis that in a cholesterol (CHOL)-implanted control rat, selective knockdown of GR/MR in the CeA would, via a corticotropin-releasing factor (CRF)-mediated mechanism, replicate the nociceptive behaviors produced by elevated amygdala CORT. Micropellets of CHOL or CORT were stereotaxically placed on the dorsal margin of the CeA. Cannulae were implanted into the CeA for the delivery of vehicle or oligodeoxynucleotide (ODN) of either antisense (ASO) or random sequences (RSO) targeting GR or MR. Visceromotor behavioral response quantified visceral sensitivity in response to colonic distension, while von Frey filaments assessed somatic sensitivity. Receptor expression was determined with qRT-PCR. In CHOL implanted controls, knockdown of GR in the CeA increased both colonic and somatic sensitivity, whereas selective knockdown of MR in the CeA induced colonic hypersensitivity without affecting somatic sensitivity. CRF expression in the CeA was increased in CHOL-implanted rats treated with GR or MR ASO and resembled the augmented CRF expression seen in the CORT-implanted rats. This is the first study to demonstrate that decreasing either GR or MR within the CeA is sufficient to induce visceral hypersensitivity whereas somatic hypersensitivity developed after only GR knockdown. The loss of either GR or MR was associated with an increased CRF expression, and may represent a common mechanism for the development of CeA-mediated nociceptive behaviors.


Assuntos
Núcleo Central da Amígdala/fisiologia , Corticosterona/metabolismo , Nociceptividade/fisiologia , Dor/etiologia , Receptores de Glucocorticoides/deficiência , Análise de Variância , Animais , Núcleo Central da Amígdala/efeitos dos fármacos , Colesterol/administração & dosagem , Corticosterona/genética , Masculino , Mineralocorticoides/metabolismo , Nociceptividade/efeitos dos fármacos , Oligodesoxirribonucleotídeos Antissenso/administração & dosagem , Estimulação Física/efeitos adversos , Ratos , Ratos Endogâmicos F344 , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/deficiência , Receptores de Mineralocorticoides/genética , Dor Visceral/etiologia
12.
EMBO J ; 34(1): 36-54, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25430741

RESUMO

A hierarchical hormonal cascade along the hypothalamic-pituitary-adrenal axis orchestrates bodily responses to stress. Although corticotropin-releasing hormone (CRH), produced by parvocellular neurons of the hypothalamic paraventricular nucleus (PVN) and released into the portal circulation at the median eminence, is known to prime downstream hormone release, the molecular mechanism regulating phasic CRH release remains poorly understood. Here, we find a cohort of parvocellular cells interspersed with magnocellular PVN neurons expressing secretagogin. Single-cell transcriptome analysis combined with protein interactome profiling identifies secretagogin neurons as a distinct CRH-releasing neuron population reliant on secretagogin's Ca(2+) sensor properties and protein interactions with the vesicular traffic and exocytosis release machineries to liberate this key hypothalamic releasing hormone. Pharmacological tools combined with RNA interference demonstrate that secretagogin's loss of function occludes adrenocorticotropic hormone release from the pituitary and lowers peripheral corticosterone levels in response to acute stress. Cumulatively, these data define a novel secretagogin neuronal locus and molecular axis underpinning stress responsiveness.


Assuntos
Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Secretagoginas/metabolismo , Estresse Fisiológico/fisiologia , Animais , Corticosterona/genética , Hormônio Liberador da Corticotropina/genética , Masculino , Camundongos , Neurônios/citologia , Núcleo Hipotalâmico Paraventricular/citologia , Hipófise/citologia , Hipófise/metabolismo , Interferência de RNA , Secretagoginas/genética , Transcriptoma/fisiologia
13.
Eur J Immunol ; 45(3): 807-17, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25487064

RESUMO

Potentially life-threatening enterocolitis is the most frequent complication in children with colonic aganglionosis (Hirschsprung disease, HSCR), and little is known about the mechanisms leading to enterocolitis. Splenic lymphopenia has been reported in the Endothelin Receptor B (Ednrb)-null mouse model of HSCR that develops enterocolitis. In this study, we sought to identify molecular mechanisms underlying this immune phenotype. We employed the Ednrb(-/-) mouse, and the knockout of its ligand, Edn3 (Edn3(-/-)). The major finding is that enterocolitis in the Ednrb(-/-) and Edn3(-/-) mice lead to thymic involution, splenic lymphopenia, and suppression of B lymphopoiesis as a consequence of colonic aganglionosis, not an intrinsic Edn3-Ednrb signaling defect directly affecting the lymphoid organs. We showed that adoptive transfer of Ednrb(-/-) marrow repopulated the RAG2-null mice marrow, thymus and spleen without development of enterocolitis. We identified the glucocorticoid corticosterone, as a potential mediator of the immune phenotype. This previously unrecognized pattern of immune abnormalities in mouse is nearly identical to lymphoid depletion in neonatal sepsis during severe physiological stress, suggesting that the mouse model used here could be also used for sepsis studies.


Assuntos
Endotelina-3/imunologia , Enterocolite/imunologia , Doença de Hirschsprung/imunologia , Receptor de Endotelina B/imunologia , Animais , Colo/imunologia , Colo/patologia , Corticosterona/genética , Corticosterona/imunologia , Modelos Animais de Doenças , Endotelina-3/genética , Enterocolite/genética , Enterocolite/patologia , Doença de Hirschsprung/genética , Doença de Hirschsprung/patologia , Camundongos , Camundongos Knockout , Receptor de Endotelina B/genética , Baço/imunologia , Baço/patologia , Timo/imunologia , Timo/patologia
14.
Int J Mol Sci ; 15(11): 21028-44, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25405735

RESUMO

Leydig cells secrete testosterone, which is essential for male fertility and reproductive health. Stress increases the secretion of glucocorticoid (corticosterone, CORT; in rats), which decreases circulating testosterone levels in part through a direct action by binding to the glucocorticoid receptors (NR3C1) in Leydig cells. The intratesticular CORT level is dependent on oxidative inactivation of glucocorticoid by 11ß-hydroxysteroid dehydrogenase 1 (HSD11B1) in Leydig cells. In the present study, we investigated the time-course changes of steroidogenic gene expression levels after acute immobilization stress in rats. The plasma CORT levels were significantly increased 0.5, 1, 3 and 6 h after immobilization stress, while plasma testosterone levels were significantly reduced 3 and 6 h, after stress and luteinizing hormone (LH) did not change. Immobilization stress caused the down-regulation of Scarb1, Star and Cyp17a1 expression levels in the rat testis starting at the first hour of stress, ahead of the significant decreases of plasma testosterone levels. Other mRNA levels, including Cyp11a1, Hsd3b1 and Hsd17b3, began to decline after 3 h. Hsd11b1 and Nos2 mRNA levels did not change during the course of stress. Administration of glucocorticoid antagonist RU486 significantly restored plasma testosterone levels. In conclusion, Scarb1, Star and Cyp17a1 expression levels are more sensitive to acute stress, and acute immobilization stress causes the decline of the steroidogenic pathway via elevating the levels of glucocorticoid, which binds to NR3C1 in Leydig cells to inhibit steroidogenic gene expression.


Assuntos
Regulação da Expressão Gênica , Hormônios/sangue , Hormônios/genética , Células Intersticiais do Testículo/metabolismo , Animais , Células Cultivadas , Corticosterona/sangue , Corticosterona/genética , Masculino , Ratos , Ratos Sprague-Dawley , Restrição Física , Esteroides/sangue , Esteroides/metabolismo
15.
PLoS One ; 9(11): e111944, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25365210

RESUMO

The suprachiasmatic nucleus (SCN) is a circadian oscillator entrained to the day/night cycle via input from the retina. Serotonin (5-HT) afferents to the SCN modulate retinal signals via activation of 5-HT1B receptors, decreasing responsiveness to light. Consequently, 5-HT1B receptor knockout (KO) mice entrain to the day/night cycle with delayed activity onsets. Since circulating corticosterone levels exhibit a robust daily rhythm peaking around activity onset, we asked whether delayed entrainment of activity onsets affects rhythmic corticosterone secretion. Wheel-running activity and plasma corticosterone were monitored in mice housed under several different lighting regimens. Both duration of the light:dark cycle (T cycle) and the duration of light within that cycle was altered. 5-HT1B KO mice that entrained to a 9.5L:13.5D (short day in a T = 23 h) cycle with activity onsets delayed more than 4 h after light offset exhibited a corticosterone rhythm in phase with activity rhythms but reduced 50% in amplitude compared to animals that initiated daily activity <4 h after light offset. Wild type mice in 8L:14D (short day in a T = 22 h) conditions with highly delayed activity onsets also exhibited a 50% reduction in peak plasma corticosterone levels. Exogenous adrenocorticotropin (ACTH) stimulation in animals exhibiting highly delayed entrainment suggested that the endogenous rhythm of adrenal responsiveness to ACTH remained aligned with SCN-driven behavioral activity. Circadian clock gene expression in the adrenal cortex of these same animals suggested that the adrenal circadian clock was also aligned with SCN-driven behavior. Under T cycles <24 h, altered circadian entrainment to short day (winter-like) conditions, manifest as long delays in activity onset after light offset, severely reduces the amplitude of the diurnal rhythm of plasma corticosterone. Such a pronounced reduction in the glucocorticoid rhythm may alter rhythmic gene expression in the central nervous system and in peripheral organs contributing to an array of potential pathophysiologies.


Assuntos
Comportamento Animal/fisiologia , Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Corticosterona/metabolismo , Receptor 5-HT1B de Serotonina/metabolismo , Núcleo Supraquiasmático/metabolismo , Hormônio Adrenocorticotrópico/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Relógios Biológicos/efeitos dos fármacos , Ritmo Circadiano/efeitos dos fármacos , Corticosterona/genética , Camundongos , Camundongos Knockout , Receptor 5-HT1B de Serotonina/genética
16.
Am J Physiol Regul Integr Comp Physiol ; 307(9): R1085-91, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25163921

RESUMO

Following an immune challenge, there is two-way communication between the nervous and immune systems. It is proposed that a neural reflex--the inflammatory reflex--regulates the plasma levels of the key proinflammatory cytokine TNF-α, and that its efferent pathway is in the splanchnic sympathetic nerves. The evidence for this reflex is based on experiments on anesthetized animals, but anesthesia itself suppresses inflammation, confounding interpretation. Here, we show that previous section of the splanchnic nerves strongly enhances the levels of plasma TNF-α in conscious rats 90 min after they received intravenous LPS (60 µg/kg). The same reflex mechanism, therefore, applies in conscious as in anesthetized animals. In anesthetized rats, we then determined the longer-term effects of splanchnic nerve section on responses to LPS (60 µg/kg iv). We confirmed that prior splanchnic nerve section enhanced the early (90 min) peak in plasma TNF-α and found that it reduced the 90-min peak of the anti-inflammatory cytokine IL-10; both subsequently fell to low levels in all animals. Splanchnic nerve section also enhanced the delayed rise in two key proinflammatory cytokines IL-6 and interferon γ. That enhancement was undiminished after 6 h, when other measured cytokines had subsided. Finally, LPS treatment caused hypotensive shock in rats with cut splanchnic nerves but not in sham-operated animals. These findings demonstrate that reflex activation of the splanchnic anti-inflammatory pathway has a powerful and sustained restraining influence on inflammatory processes.


Assuntos
Anestesia , Inflamação/metabolismo , Reflexo/fisiologia , Transdução de Sinais/fisiologia , Nervos Esplâncnicos/fisiologia , Animais , Corticosterona/sangue , Corticosterona/genética , Corticosterona/metabolismo , Citocinas/sangue , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Lipopolissacarídeos/toxicidade , Masculino , Ratos , Ratos Sprague-Dawley
17.
Proc Biol Sci ; 281(1790)2014 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-25056627

RESUMO

Glucocorticoid hormones (CORT) are predicted to promote adaptation to variable environments, yet little is known about the potential for CORT secretion patterns to respond to selection in free-living populations. We assessed the heritable variation underlying differences in hormonal phenotypes using a cross-foster experimental design with nestling North American barn swallows (Hirundo rustica erythrogaster). Using a bivariate animal model, we partitioned variance in baseline and stress-induced CORT concentrations into their additive genetic and rearing environment components and estimated their genetic correlation. Both baseline and stress-induced CORT were heritable with heritability of 0.152 and 0.343, respectively. We found that the variation in baseline CORT was best explained by rearing environment, whereas the variation in stress-induced CORT was contributed to by a combination of genetic and environmental factors. Further, we did not detect a genetic correlation between these two hormonal traits. Although rearing environment appears to play an important role in the secretion of both types of CORT, our results suggest that stress-induced CORT levels are underlain by greater additive genetic variance compared with baseline CORT levels. Accordingly, we infer that the glucocorticoid response to stress has a greater potential for evolutionary change in response to selection compared with baseline glucocorticoid secretion patterns.


Assuntos
Glucocorticoides/sangue , Estresse Fisiológico/fisiologia , Andorinhas/genética , Andorinhas/fisiologia , Adaptação Fisiológica/fisiologia , Animais , Animais Recém-Nascidos , Corticosterona/sangue , Corticosterona/genética , Feminino , Genótipo , Glucocorticoides/genética , Masculino , Andorinhas/crescimento & desenvolvimento
18.
Artigo em Inglês | MEDLINE | ID: mdl-25038300

RESUMO

Chronic endogenous glucocorticoid (GC) excess in mammals is associated with metabolic dysfunction and dyslipidemia that are characterized by increased plasma triglyceride and total cholesterol (Tch) levels. However, the effects of chronic GC administration on cholesterol metabolism, particularly in muscle tissues of broiler chickens, are unknown. In this study, broiler chickens were treated chronically with vehicle (CON) or corticosterone (CORT) for 2 weeks. Chronic CORT treatment significantly increased Tch levels in pectoralis major muscle (PMC) (p<0.001) as well as in leg muscle (p<0.01), and CORT enhanced triglyceride levels in the PMC (p<0.001). Real-time PCR results showed that HMGCR (p<0.05) mRNA expression was up-regulated by CORT in PMC, and 11ß-HSD1 gene transcription (p=0.08) was not significantly downregulated, whereas glucocorticoid receptor (GR) mRNA expression, 11ß-HSD2, CYP7A1, CYP27A1, ApoB and LDLR were unchanged by CORT (p>0.05). Western blot results showed that the levels of total GR (p=0.08) tended to be increased and nuclear GR protein (p<0.05) was increased in PMC by CORT administration. Parallel to an increase in gene expression, HMGCR protein expression in PMC was significantly increased (p<0.05) by CORT. Moreover, LDLR (p<0.05), ApoA1 (p=0.06) and 11ß-HSD2 (p=0.07) protein expression in PMC tended to be increased by CORT compared to control. These results indicate that chronic CORT administration causes cholesterol accumulation in PMC tissues of broiler chickens by increasing cholesterol synthesis and uptake.


Assuntos
Galinhas/metabolismo , Galinhas/fisiologia , Colesterol/metabolismo , Corticosterona/metabolismo , Estresse Fisiológico/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Animais , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Galinhas/genética , Colestanotriol 26-Mono-Oxigenase/genética , Colestanotriol 26-Mono-Oxigenase/metabolismo , Colesterol/genética , Colesterol 7-alfa-Hidroxilase/genética , Colesterol 7-alfa-Hidroxilase/metabolismo , Corticosterona/genética , Regulação para Baixo/genética , Expressão Gênica/genética , Glucocorticoides/genética , Glucocorticoides/metabolismo , Músculo Esquelético/metabolismo , RNA Mensageiro/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Estresse Fisiológico/genética , Transcrição Gênica/genética , Triglicerídeos/genética , Triglicerídeos/metabolismo , Regulação para Cima/genética
19.
Sci Signal ; 7(314): ra20, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24570488

RESUMO

Intestinal crypt epithelial cells synthesize glucocorticoids, steroid hormones that protect against inflammatory bowel disease. To investigate how intestinal glucocorticoids are regulated during chronic inflammation, we induced chronic colitis in mice by exposing them to the chemical dextran sulfate sodium (DSS). We found that intestinal glucocorticoid secretion and expression of the genes Cyp11a1 and Cyp11b1 (which encode enzymes that synthesize glucocorticoids) were initially stimulated, but declined during the chronic phase, whereas tumor necrosis factor (TNF) and inflammatory cytokines secreted by T helper type 1 (TH1) and TH17 cells continuously increased in abundance in the inflamed colon. This suggested that inadequate intestinal glucocorticoid synthesis is a feature of chronic intestinal inflammation. We screened for cytokines that regulated intestinal glucocorticoid synthesis and found that TNF suppressed corticosterone secretion and Cyp11a1 and Cyp11b1 expression in an intestinal crypt epithelial cell line. TNF suppressed steroidogenesis by activating the transcription factors c-Jun and nuclear factor κB (NF-κB), which both interacted with the transcription factor NR5A2 and repressed Cyp11a1 reporter activity. This repression was relieved by expression of a dominant-negative form of c-Jun amino-terminal kinase 1 (JNK1), inhibitor of NF-κB, or by a JNK inhibitor. Furthermore, the dominant-negative TNF inhibitor XPro1595 inhibited c-Jun and NF-κB activation in mice, restored intestinal Cyp11a1 and Cyp11b1 expression, reduced colonic cell death, and rescued chronic colitis caused by DSS. Thus, during chronic colitis, TNF suppresses intestinal steroidogenic gene expression by inhibiting the activity of NR5A2, thus decreasing glucocorticoid synthesis and sustaining chronic inflammation.


Assuntos
Colite/metabolismo , Glucocorticoides/biossíntese , Mucosa Intestinal/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular , Enzima de Clivagem da Cadeia Lateral do Colesterol/biossíntese , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/imunologia , Doença Crônica , Colite/induzido quimicamente , Colite/genética , Colite/imunologia , Colite/patologia , Corticosterona/biossíntese , Corticosterona/genética , Corticosterona/imunologia , Sulfato de Dextrana/toxicidade , Regulação Enzimológica da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/imunologia , Glucocorticoides/genética , Glucocorticoides/imunologia , Humanos , Intestinos/imunologia , Intestinos/patologia , Camundongos , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/imunologia , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , NF-kappa B/genética , NF-kappa B/imunologia , NF-kappa B/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/imunologia , Esteroide 11-beta-Hidroxilase/biossíntese , Esteroide 11-beta-Hidroxilase/genética , Esteroide 11-beta-Hidroxilase/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/patologia , Células Th17/imunologia , Células Th17/metabolismo , Células Th17/patologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
20.
PLoS One ; 8(5): e63814, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23717489

RESUMO

Adrenal steroidogenesis is under a complex regulation involving extrinsic and intrinsic adrenal factors. TNF-α is an inflammatory cytokine produced in response to tissue injury and several other stimuli. We have previously demonstrated that TNF-R1 knockout (TNF-R1(-/-)) mice have a dysregulated synthesis of glucocorticoids (GCs) during Trypanosoma cruzi acute infection. Since TNF-α may influence GCs production, not only through the hypothalamus-pituitary axis, but also at the adrenal level, we now investigated the role of this cytokine on the adrenal GCs production. Wild type (WT) and TNF-R1(-/-) mice undergoing acute infection (Tc-WT and Tc-TNF-R1(-/-) groups), displayed adrenal hyperplasia together with increased GCs levels. Notably, systemic ACTH remained unchanged in Tc-WT and Tc-TNF-R1(-/-) compared with uninfected mice, suggesting some degree of ACTH-independence of GCs synthesis. TNF-α expression was increased within the adrenal gland from both infected mouse groups, with Tc-WT mice showing an augmented TNF-R1 expression. Tc-WT mice showed increased levels of P-p38 and P-ERK compared to uninfected WT animals, whereas Tc-TNF-R1(-/-) mice had increased p38 and JNK phosphorylation respect to Tc-WT mice. Strikingly, adrenal NF-κB and AP-1 activation during infection was blunted in Tc-TNF-R1(-/-) mice. The accumulation of mRNAs for steroidogenic acute regulatory protein and cytochrome P450 were significantly increased in both Tc-WT and Tc-TNF-R1(-/-) mice; being much more augmented in the latter group, which also had remarkably increased GCs levels. TNF-α emerges as a potent modulator of steroidogenesis in adrenocortical cells during T. cruzi infection in which MAPK pathways, NF-κB and AP-1 seem to play a role in the adrenal synthesis of pro-inflammatory cytokines and enzymes regulating GCs synthesis. These results suggest the existence of an intrinsic immune-adrenal interaction involved in the dysregulated synthesis of GCs during murine Chagas disease.


Assuntos
Glândulas Suprarrenais/metabolismo , Doença de Chagas/metabolismo , Doença de Chagas/parasitologia , Glucocorticoides/biossíntese , Glucocorticoides/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Animais , Doença de Chagas/sangue , Doença de Chagas/genética , Corticosterona/genética , Corticosterona/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Expressão Gênica/genética , Glucocorticoides/genética , Inflamação/genética , Inflamação/metabolismo , Inflamação/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação/genética , RNA Mensageiro/genética , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Trypanosoma cruzi , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...