Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Exp Biol ; 227(Suppl_1)2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38449331

RESUMEN

Prenatal stress programmes long-lasting neuroendocrine and behavioural changes in the offspring. Often this programming is maladaptive and sex specific. For example, using a rat model of maternal social stress in late pregnancy, we have demonstrated that adult prenatally stressed male, but not prenatally stressed female offspring display heightened anxiety-like behaviour, whereas both sexes show hyperactive hypothalamo-pituitary-adrenal (HPA) axis responses to stress. Here, we review the current knowledge of the mechanisms underpinning dysregulated HPA axis responses, including evidence supporting a role for reduced neurosteroid-mediated GABAergic inhibitory signalling in the brains of prenatally stressed offspring. How maternal psychosocial stress is signalled from the mother to the fetuses is unclear. Direct transfer of maternal glucocorticoids to the fetuses is often considered to mediate the programming effects of maternal stress on the offspring. However, protective mechanisms including attenuated maternal stress responses and placental 11ß-hydroxysteroid dehydrogenase-2 (which inactivates glucocorticoids) should limit materno-fetal glucocorticoid transfer during pregnancy. Moreover, a lack of correlation between maternal stress, circulating maternal glucocorticoid levels and circulating fetal glucocorticoid levels is reported in several studies and across different species. Therefore, here we interrogate the evidence for a role for maternal glucocorticoids in mediating the effects of maternal stress on the offspring and consider the evidence for alternative mechanisms, including an indirect role for glucocorticoids and the contribution of changes in the placenta in signalling the stress status of the mother to the fetus.


Asunto(s)
Glucocorticoides , Madres , Animales , Femenino , Masculino , Embarazo , Ratas , Feto , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Placenta
2.
Front Neuroendocrinol ; 53: 100739, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30802468

RESUMEN

Successful pregnancy requires adjustments to multiple maternal homeostatic mechanisms, governed by the maternal brain to support and enable survival of the growing fetus and placenta. Such adjustments fit the concept of allostasis (stability through change) and have a cost: allostatic load. Allostasis is driven by ovarian, anterior pituitary, placental and feto-placental hormones acting on the maternal brain to promote adaptations that support the pregnancy and protect the fetus. Many women carry an existing allostatic load into pregnancy, from socio-economic circumstances, poor mental health and in 'developed' countries, also from obesity. These pregnancies have poorer outcomes indicating negative interactions (failing allostasis) between pre-pregnancy and pregnancy allostatic loads. Use of animal models, such as adult prenatally stressed female offspring with abnormal neuroendocrine, metabolic and behavioural phenotypes, to probe gene expression changes, and epigenetic mechanisms in the maternal brain in adverse pregnancies are discussed, with the prospect of ameliorating poor pregnancy outcomes.


Asunto(s)
Adaptación Fisiológica/fisiología , Encéfalo/crecimiento & desarrollo , Desarrollo Fetal/fisiología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Animales , Femenino , Humanos , Sistemas Neurosecretores/metabolismo , Embarazo
3.
Eur J Neurosci ; 52(1): 2487-2515, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31705553

RESUMEN

The hypothalamo-pituitary-adrenal (HPA) axis plays a key role in the neuroendocrine response to stress and in maintaining physiological homoeostasis. However, stress that is chronic in nature can lead to HPA axis dysfunction and increase the risk for developing affective disorders, particularly if the stress is experienced during vulnerable periods in life. Sex differences in how the HPA axis responds to stress are well established, with females typically displaying heightened responses. The underlying cause of these sex differences is important to understand, as many neuropsychiatric disorders disproportionately affect females. Much research has provided evidence for gonadal sex steroids in underpinning sex differences in HPA axis responsivity; however, we suggest that neuroactive metabolites of these steroids also play a key role in the brain in mediating sex differences in HPA axis responses to stress. The relationship between neuroactive steroids and stress is complex. Acute stress rapidly increases neuroactive steroid production, which can in turn modulate activity of the HPA axis. However, under chronic stress conditions, stress can impact the brain's capacity to generate steroids, and this in turn has corollary effects on HPA axis function that may increase the propensity for psychopathology, given both HPA axis dysfunction and deficits in neuroactive steroids are implicated in affective disorders. Hence, here we review the evidence from animal and human studies for sex differences in the interactions between neuroactive steroids and the stress axis at various stages of life, under physiological and pathophysiological stress conditions and consider the implications for health and disease.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Animales , Femenino , Hormonas Esteroides Gonadales , Humanos , Masculino , Caracteres Sexuales , Esteroides , Estrés Fisiológico , Estrés Psicológico
4.
Cell Tissue Res ; 375(1): 69-83, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30415283

RESUMEN

During pregnancy, the regulation of several neuroendocrine systems is altered to support the pregnancy and facilitate the transition to motherhood. These adaptations are organised by the mother's brain and include those that serve to optimise foetal growth, protect the foetus(es) from adverse prenatal programming by maternal stress, facilitate timely parturition and ensure the offspring are nourished and cared for after birth. Although pregnancy hormones are important in inducing and maintaining many of these adaptations, their effects are often mediated via interactions with neuropeptide systems in the brain. In particular, endogenous opioids in the maternal brain play key roles in altered regulation of the stress axis, the oxytocin system, the prolactin system and the neural circuits controlling maternal behaviour. Together, these adaptations maximise the likelihood of a successful pregnancy outcome.


Asunto(s)
Analgésicos Opioides/metabolismo , Encéfalo/metabolismo , Lactancia/metabolismo , Transducción de Señal , Animales , Femenino , Humanos , Conducta Materna , Embarazo , Prolactina/metabolismo
5.
J Neurosci ; 35(2): 666-77, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25589761

RESUMEN

Maternal social stress during late pregnancy programs hypothalamo-pituitary-adrenal (HPA) axis hyper-responsiveness to stressors, such that adult prenatally stressed (PNS) offspring display exaggerated HPA axis responses to a physical stressor (systemic interleukin-1ß; IL-1ß) in adulthood, compared with controls. IL-1ß acts via a noradrenergic relay from the nucleus tractus solitarii (NTS) to corticotropin releasing hormone neurons in the paraventricular nucleus (PVN). Neurosteroids can reduce HPA axis responses, so allopregnanolone and 3ß-androstanediol (3ß-diol; 5α-reduced metabolites of progesterone and testosterone, respectively) were given subacutely (over 24 h) to PNS rats to seek reversal of the "programmed" hyper-responsive HPA phenotype. Allopregnanolone attenuated ACTH responses to IL-1ß (500 ng/kg, i.v.) in PNS females, but not in PNS males. However, 3ß-diol normalized HPA axis responses to IL-1ß in PNS males. Impaired testosterone and progesterone metabolism or increased secretion in PNS rats was indicated by greater plasma testosterone and progesterone concentrations in male and female PNS rats, respectively. Deficits in central neurosteroid production were indicated by reduced 5α-reductase mRNA levels in both male and female PNS offspring in the NTS, and in the PVN in males. In PNS females, adenovirus-mediated gene transfer was used to upregulate expression of 5α-reductase and 3α-hydroxysteroid dehydrogenase mRNAs in the NTS, and this normalized hyperactive HPA axis responses to IL-1ß. Thus, downregulation of neurosteroid production in the brain may underlie HPA axis hyper-responsiveness in prenatally programmed offspring, and administration of 5α-reduced steroids acutely to PNS rats overrides programming of hyperactive HPA axis responses to immune challenge in a sex-dependent manner.


Asunto(s)
Androstano-3,17-diol/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Pregnanolona/farmacología , Estrés Psicológico/tratamiento farmacológico , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica)/genética , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica)/metabolismo , Androstano-3,17-diol/uso terapéutico , Animales , Femenino , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Interleucina-1beta/farmacología , Masculino , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/crecimiento & desarrollo , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/crecimiento & desarrollo , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Pregnanolona/uso terapéutico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/crecimiento & desarrollo , Núcleo Solitario/metabolismo
6.
Reproduction ; 151(6): 709-17, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27026714

RESUMEN

Social stress during pregnancy has profound effects on offspring physiology. This study examined whether an ethologically relevant social stress during late pregnancy in rats alters the reproductive axis and adrenal gland structure in post-pubertal male and female offspring. Prenatally stressed (PNS) pregnant rats (n=9) were exposed to an unfamiliar lactating rat for 10 min/day from day 16 to 20 of pregnancy inclusive, whereas control pregnant rats (n=9) remained in their home cages. Gonads, adrenal glands and blood samples were obtained from one female and one male from each litter at 11 to 12-weeks of age. Anogenital distance was measured. There was no treatment effect on body, adrenal or gonad weight at 11-12 weeks. PNS did not affect the number of primordial, secondary or tertiary ovarian follicles, numbers of corpora lutea or ovarian FSH receptor expression. There was an indication that PNS females had more primary follicles and greater ovarian aromatase expression compared with control females (both P=0.09). PNS males had longer anogenital distances (0.01±0.0 cm/g vs 0.008±0.00 cm/g; P=0.007) and higher plasma FSH concentrations (0.05 ng/mL vs 0.006 ng/mL; s.e.d.=0.023; P=0.043) compared with control males. There were no treatment effects on the number of Sertoli cells or seminiferous tubules, seminiferous tubule area, plasma testosterone concentration or testis expression of aromatase, FSH receptor or androgen receptor. PNS did not affect adrenal size. These data suggest that the developing male reproductive axis is more sensitive to maternal stress and that PNS may enhance aspects of male reproductive development.


Asunto(s)
Glándulas Suprarrenales/química , Conducta Animal , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Reproducción/fisiología , Conducta Social , Estrés Psicológico/fisiopatología , Glándulas Suprarrenales/fisiología , Animales , Animales Recién Nacidos , Femenino , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley , Factores Sexuales
7.
J Neurosci ; 34(29): 9665-76, 2014 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-25031406

RESUMEN

Maternal behavior ensures the proper development of the offspring. In lactating mammals, maternal behavior is impaired by stress, the physiological consequence of central corticotropin-releasing factor receptor (CRF-R) activation. However, which CRF-R subtype in which specific brain area(s) mediates this effect is unknown. Here we confirmed that an intracerebroventricularly injected nonselective CRF-R antagonist enhances, whereas an agonist impairs, maternal care. The agonist also prolonged the stress-induced decrease in nursing, reduced maternal aggression and increased anxiety-related behavior. Focusing on the bed nucleus of the stria terminalis (BNST), CRF-R1 and CRF-R2 mRNA expression did not differ in virgin versus lactating rats. However, CRF-R2 mRNA was more abundant in the posterior than in the medial BNST. Pharmacological manipulations within the medial-posterior BNST showed that both CRF-R1 and CRF-R2 agonists reduced arched back nursing (ABN) rapidly and after a delay, respectively. After stress, both antagonists prevented the stress-induced decrease in nursing, with the CRF-R2 antagonist actually increasing ABN. During the maternal defense test, maternal aggression was abolished by the CRF-R2, but not the CRF-R1, agonist. Anxiety-related behavior was increased by the CRF-R1 agonist and reduced by both antagonists. Both antagonists were also effective in virgin females but not in males, revealing a sexual dimorphism in the regulation of anxiety within the medial-posterior BNST. In conclusion, the detrimental effects of increased CRF-R activation on maternal behavior are mediated via CRF-R2 and, to a lesser extent, via CRF-R1 in the medial-posterior BNST in lactating rats. Moreover, both CRF-R1 and CRF-R2 regulate anxiety in females independently of their reproductive status.


Asunto(s)
Lactancia , Conducta Materna/psicología , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Núcleos Septales/metabolismo , Análisis de Varianza , Animales , Hormona Liberadora de Corticotropina/análogos & derivados , Hormona Liberadora de Corticotropina/farmacología , Femenino , Lactancia/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Motivación/fisiología , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/genética , Núcleos Septales/efectos de los fármacos , Factores Sexuales
8.
Neurobiol Stress ; 30: 100631, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38601362

RESUMEN

To ensure the unrestricted expression of maternal behaviour peripartum, activity of the corticotropin-releasing factor (CRF) system needs to be minimised. CRF binding protein (CRF-BP) might be crucial for this adaptation, as its primary function is to sequester freely available CRF and urocortin1, thereby dampening CRF receptor (CRF-R) signalling. So far, the role of CRF-BP in the maternal brain has barely been studied, and a potential role in curtailing activation of the stress axis is unknown. We studied gene expression for CRF-BP and both CRF-R within the paraventricular nucleus (PVN) of the hypothalamus. In lactating rats, Crh-bp expression in the parvocellular PVN was significantly higher and Crh-r1 expression in the PVN significantly lower compared to virgin rats. Acute CRF-BP inhibition in the PVN with infusion of CRF(6-33) increased basal plasma corticosterone concentrations under unstressed conditions in dams. Furthermore, while acute intra-PVN infusion of CRF increased corticosterone secretion in virgin rats, it was ineffective in vehicle (VEH)-pre-treated lactating rats, probably due to a buffering effect of CRF-BP. Indeed, pre-treatment with CRF(6-33) reinstated a corticosterone response to CRF in lactating rats, highlighting the critical role of CRF-BP in maintaining attenuated stress reactivity in lactation. To our knowledge, this is the first study linking hypothalamic CRF-BP activity to hypothalamic-pituitary-adrenal axis regulation in lactation. In terms of behaviour, acute CRF-BP inhibition in the PVN under non-stress conditions reduced blanket nursing 60 min and licking/grooming 90 min after infusion compared to VEH-treated rats, while increasing maternal aggression towards an intruder. Lastly, chronic intra-PVN inhibition of CRF-BP strongly reduced maternal aggression, with modest effects on maternal motivation and care. Taken together, intact activity of the CRF-BP in the PVN during the postpartum period is essential for the dampened responsiveness of the stress axis, as well as for the full expression of appropriate maternal behaviour.

9.
Nat Rev Neurosci ; 9(1): 11-25, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18073776

RESUMEN

A successful pregnancy requires multiple adaptations of the mother's physiology to optimize fetal growth and development, to protect the fetus from adverse programming, to provide impetus for timely parturition and to ensure that adequate maternal care is provided after parturition. Many of these adaptations are organized by the mother's brain, predominantly through changes in neuroendocrine systems, and these changes are primarily driven by the hormones of pregnancy. By contrast, adaptations in the mother's brain during lactation are maintained by external stimuli from the young. The changes in pregnancy are not necessarily innocuous: they may predispose the mother to post-partum mood disorders.


Asunto(s)
Adaptación Fisiológica , Encéfalo/fisiología , Lactancia/fisiología , Conducta Materna/fisiología , Embarazo/fisiología , Embarazo/psicología , Animales , Femenino , Desarrollo Fetal , Hormonas/metabolismo , Humanos , Trastornos del Humor/etiología , Trastornos del Humor/metabolismo , Trastornos del Humor/psicología , Sistemas Neurosecretores/fisiología , Trastornos Puerperales/etiología , Trastornos Puerperales/metabolismo , Trastornos Puerperales/psicología
10.
Reproduction ; 146(5): R175-89, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23901130

RESUMEN

A suboptimal in utero environment, for example, as a result of maternal stress, can have detrimental effects on the pregnancy and long-term adverse 'programming' effects on the offspring. This article focuses on the effects of prenatal social stress on the mother, her pregnancy and the offspring, since these issues have ethological relevance in both animals and humans. The consequences of social stress exposure depend on when during pregnancy the stress occurs, and many of the effects on the offspring are sex specific. Social stress during early pregnancy tends to result in pregnancy loss, whereas stress exposure later in pregnancy, when the mother has already invested considerable resources in the foetuses, results in programmed offspring of low birth weight: a risk factor for various adulthood diseases. Neuroendocrine and behavioural responses to stress in the offspring are particularly sensitive to foetal programming by prenatal stress, indicated by enhanced hypothalamo-pituitary-adrenal (HPA) axis responses and increased anxiety behaviour, which result from permanent changes in the offspring's brain. The dysregulation of HPA axis function may also interfere with other systems, for example, the hypothalamic-pituitary-gonadal axis, as there is evidence for alterations in steroidogenesis, reproductive potential and impaired reproductive/social behaviours in prenatally stressed offspring. Prenatal social stress also programmes future maternal behaviour, highlighting the potential for negative phenotypes to be transmitted to future generations. The possible mechanisms through which maternal stress during pregnancy is transmitted to the foetuses and the foetal brain is programmed by prenatal stress and the potential to overwrite programming of the offspring are discussed.


Asunto(s)
Desarrollo Fetal , Complicaciones del Embarazo/fisiopatología , Estrés Psicológico/fisiopatología , Adulto , Animales , Ansiedad/etiología , Niño , Conducta Infantil , Modelos Animales de Enfermedad , Pérdida del Embrión/etiología , Femenino , Retardo del Crecimiento Fetal/etiología , Humanos , Sistema Hipotálamo-Hipofisario/fisiopatología , Lactante , Conducta del Lactante , Masculino , Sistema Hipófiso-Suprarrenal/fisiopatología , Embarazo , Complicaciones del Embarazo/metabolismo , Medio Social , Estrés Psicológico/metabolismo
11.
Curr Opin Endocr Metab Res ; 25: 100367, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36561280

RESUMEN

Early-life stress can lead to detrimental offspring outcomes, including an increased risk for mood disorders and hypothalamic-pituitary-adrenal axis dysregulation. Neurosteroids bind to ligand-gated neurotransmitter receptors, rapidly modulating neuronal excitability and promoting termination of stress responses. Reduced neurosteroidogenesis underlies some of the aberrant neuroendocrine and behavioural phenotypes observed in adult prenatally stressed rodents. During development, disruptions in neurosteroid generation and action also lead to long-term programming effects on the off-spring's brain and behaviour. Here, we review recent advances in the field, focusing on the interaction between neurosteroids and early-life stress outcomes in adulthood and in the perinatal period. We also discuss the direction of future research, with emphasis on quantification methods, sex differences, and neurosteroids as targets for therapeutic intervention.

12.
J Endocrinol ; 255(3): 143-158, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36256689

RESUMEN

Stress during pregnancy negatively affects the fetus and increases the risk for affective disorders in adulthood. Excess maternal glucocorticoids are thought to mediate fetal programming; however, whether they exert their effects directly or indirectly remains unclear. During pregnancy, protective mechanisms including maternal hypothalamic-pituitary-adrenal (HPA) axis hyporesponsiveness and placental 11ß-hydroxysteroid dehydrogenase (11ßHSD) type 2, which inactivates glucocorticoids, limit mother-to-fetus glucocorticoid transfer. However, whether repeated stress negatively impacts these mechanisms is not known. Pregnant rats were exposed to repeated social stress on gestational days (GD) 16-20 and several aspects of HPA axis and glucocorticoid regulation, including concentrations of glucocorticoids, gene expression for their receptors (Nr3c1, Nr3c2), receptor chaperones (Fkbp51, Fkbp52) and enzymes that control local glucocorticoid availability (Hsd11b1, Hsd11b2), were investigated in the maternal, placental and fetal compartments on GD20. The maternal HPA axis was activated following stress, though the primary driver was vasopressin, rather than corticotropin-releasing hormone. Despite the stress-induced increase in circulating corticosterone in the dams, only a modest increase was detected in the circulation of female fetuses, with no change in the fetal brain of either sex. Moreover, there was no change in the expression of genes that mediate glucocorticoid actions or modulate local concentrations in the fetal brain. In the placenta labyrinth zone, stress increased Hsd11b2 expression only in males and Fkbp51 expression only in females. Our results indicate that any role glucocorticoids play in fetal programming is likely indirect, perhaps through sex-dependent alterations in placental gene expression, rather than exerting effects via direct crossover into the fetal brain.


Asunto(s)
Glucocorticoides , Sistema Hipotálamo-Hipofisario , Masculino , Femenino , Embarazo , Animales , Ratas , Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Placenta/metabolismo , Feto/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo
13.
Stress ; 14(1): 6-12, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20666638

RESUMEN

In rats, late pregnancy is associated with suppressed hypothalamo-pituitary-adrenal (HPA) axis responses to a variety of stressful stimuli. The result is reduced corticosterone secretion following stress, which is considered to give some protection to the fetuses from adverse glucocorticoid programming and limits the catabolic effect of corticosterone, hence minimizing maternal energy expenditure. We have used a model of immune challenge in which systemic administration of the cytokine, interleukin-1ß (IL-1ß), allows study of the mechanisms underlying HPA axis hyporesponsiveness in late pregnancy. Suppressed responsiveness of parvocellular paraventricular nucleus (pPVN) corticotropin-releasing hormone neurons, and hence the HPA axis, following IL-1ß in late pregnancy is evidently explained by presynaptic inhibition of noradrenaline release in the pPVN, owing to increased endogenous opioid peptide enkephalin production in brainstem nucleus tractus solitarii neurons. Allopregnanolone, a neurosteroid metabolite of progesterone, signals the pregnancy status of the animal to the brain and stimulates opioid production in the brainstem. In this review, we discuss the supporting evidence for these mechanisms.


Asunto(s)
Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Preñez/fisiología , Pregnanolona/farmacología , Hormona Adrenocorticotrópica/metabolismo , Analgésicos Opioides/metabolismo , Animales , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Estradiol/sangre , Femenino , Interleucina-1beta/farmacología , Norepinefrina/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/fisiología , Embarazo , Pregnanolona/sangre , Progesterona/sangre , Ratas , Estrés Fisiológico , betaendorfina/metabolismo
14.
Stress ; 14(6): 634-43, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21854167

RESUMEN

We recently reported that male, but not female, offspring born to mothers exposed to social stress during late gestation show heightened anxiety-type behaviour in adulthood. The amygdala organises anxious behaviour, which involves actions of corticotropin-releasing hormone (CRH). CRH gene expression and/or its release are increased in the amygdala in prenatally stressed (PNS) rats. CRH type 1 receptor (CRH-R1) mediates actions of CRH and urocortin I to promote anxiety-like behaviour, whereas the CRH type 2 receptor (CRH-R2) may mediate anxiolytic actions, through actions of urocortins 2 and 3. Here, using quantitative in situ hybridisation, we investigated whether altered CRH receptor mRNA expression in the amygdaloid nuclei may explain the sex differences in anxiety behaviour in adult male and female PNS rats. CRH-R1 mRNA expression was significantly greater in the central amygdala and basolateral amygdala (BLA) in male PNS rats compared with controls, with no change in the basomedial amygdala (BMA) or medial amygdala (MeA). In PNS females, CRH-R1 mRNA expression was greater than controls only in the MeA. Conversely, CRH-R2 mRNA expression was significantly lower in the BMA of male PNS rats compared with controls, but greater in female PNS rats, with no change in the BLA or MeA in either sex. The ratio of CRH-R1:CRH-R2 mRNA in the amygdaloid nuclei was generally increased in PNS males, but not in the PNS females. In conclusion, sex differences in anxiety-type behaviour in PNS rats may be explained by differential mRNA expression for CRH-R1 (pro-anxiogenic) and CRH-R2 (pro-anxiolytic) in the amygdaloid complex.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Efectos Tardíos de la Exposición Prenatal/genética , Receptores de Hormona Liberadora de Corticotropina/genética , Animales , Ansiedad , Trastornos de Ansiedad/genética , Femenino , Masculino , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales , Estrés Psicológico
15.
Stress ; 14(6): 652-64, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21995525

RESUMEN

Immune challenge during pregnancy is associated with preterm birth and poor perinatal development. The mechanisms of these effects are not known. 5α-Pregnan-3α-ol-20-one (3α,5α-THP), the neuroactive metabolite of progesterone, is critical for neurodevelopment and stress responses, and can influence cognition and affective behaviours. To develop an immune challenge model of preterm birth, pregnant Long-Evans rat dams were administered lipopolysaccharide [LPS; 30 µg/kg/ml, intraperitoneal (IP)], interleukin-1ß (IL-1ß; 1 µg/rat, IP) or vehicle (0.9% saline, IP) daily on gestational days 17-21. Compared to control treatment, prenatal LPS or IL-1ß reduced gestational length and the number of viable pups born. At 28-30 days of age, male and female offspring of mothers exposed to prenatal IL-1ß had reduced cognitive performance in the object recognition task compared to controls. In females, but not males, prenatal IL-1ß reduced anxiety-like behaviour, indicated by entries to the centre of an open field. In the hippocampus, progesterone turnover to its 5α-reduced metabolites was lower in prenatally exposed IL-1ß female, but not in male offspring. IL-1ß-exposed males and females had reduced oestradiol content in hippocampus, medial prefrontal cortex and diencephalon compared to controls. Thus, immune stress during late pregnancy reduced gestational length and negatively impacted birth outcomes, hippocampal function and central neurosteroid formation in the offspring.


Asunto(s)
Cognición/efectos de los fármacos , Fertilidad/efectos de los fármacos , Hipocampo/efectos de los fármacos , Interleucina-1beta/farmacología , Lipopolisacáridos/farmacología , Embarazo/inmunología , Animales , Estradiol/metabolismo , Femenino , Edad Gestacional , Hipocampo/metabolismo , Masculino , Efectos Tardíos de la Exposición Prenatal , Progesterona/metabolismo , Ratas , Ratas Long-Evans , Caracteres Sexuales
16.
Stress ; 14(1): 1-5, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21166559

RESUMEN

Steroid hormones play a critical role in the initiation and maintenance of pregnancy. In particular, the important role that the progesterone metabolite, and neurosteroid, allopregnanolone, may play in fetal and adolescent development is becoming increasingly evident. Unlike steroid hormones, neurosteroids act at nontraditional targets in the central and peripheral nervous systems, including GABA(A) receptor complexes. This commentary discusses the three works in this issue that elucidate the important role of allopregnanolone in the mechanisms that regulate stress hypo-sensitivity of rodents in late pregnancy, neuroprotective effects in fetal sheep exposed to a hypoxic insult, and the continuing role that prefrontal cortex formation of allopregnanolone may play on the cognitive development of gestationally stressed rat offspring, grown to adolescence. The narrative that these works comprise was facilitated by the 5(th) International Meeting on Steroids and the Nervous System (Torino, Italy), which is organized to update our knowledge on the relationships between steroid hormones synthesized in different organs and the nervous system. Topics covered in this most recent meeting included sex differences in, and hormonal influences on, cannabinoid-regulated biology; steroids and pain; the importance of co-regulatory factors for steroid receptor action in the brain; mechanism and role of estrogen-induced nonclassical signaling in the brain; vitamin D as the forgotten neurosteroid; neurosteroids and GABA(A) receptors; and pathogenic mechanisms mediated by glucocorticoid receptors in psychiatric disorders. The 6(th) International Meeting on Steroids and the Nervous System will be held in Torino, Italy in February 2011.


Asunto(s)
Neurotransmisores/fisiología , Embarazo/fisiología , Pregnanolona/fisiología , Animales , Sistema Nervioso Central/crecimiento & desarrollo , Femenino , Humanos , Progesterona/sangre
17.
J Neuroendocrinol ; 33(1): e12916, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33270955

RESUMEN

Acute swim stress results in the robust production of several neuroactive steroids, which act as mediators of the stress response. These steroids include glucocorticoids, and positive GABAA receptor modulatory steroids such as allopregnanolone and tetrahydrocorticosterone (THDOC), which potentiate inhibitory GABA signalling, thereby playing a role in the negative control of the hypothalamic-pituitary-adrenal (HPA) axis. Prenatally stressed (PNS) offspring exhibit increased vulnerability to stress-related disorders and frequently display exaggerated HPA axis responses to stressors during adulthood, which may be a result of reduced neuroactive steroid production and consequently inhibitory signalling. Here, we investigated whether exposure of rats to prenatal social stress from gestational day 16-20 altered neuroactive steroid production under non-stress conditions and in response to an acute stressor (swim stress) in adulthood. Using liquid chromatography-mass spectrometry, nine neuroactive steroids were quantified (corticosterone, deoxycorticosterone [DOC], dihydrodeoxycorticosterone, THDOC, progesterone, dihydroprogesterone, allopregnanolone, pregnenolone, testosterone) in plasma and in five brain regions (frontal cortex, hypothalamus, amygdala, hippocampus, brainstem) of male and female control and PNS rats. There was no difference in the neuroactive steroid profile between control and PNS rats under basal conditions. The increase in circulating corticosterone induced by acute swim stress was similar in control and PNS offspring. However, greater stress-induced corticosterone and DOC concentrations were observed in the brainstem of male PNS offspring, whereas DOC concentrations were lower in the hippocampus of PNS females compared to controls, following acute stress. Although PNS rats did not show deficits in allopregnanolone responses to acute stress, there were modest deficits in the production of THDOC in the brainstem, amygdala, and frontal cortex of PNS males and in the frontal cortex of PNS females. The data suggest that neuroactive steroid modulation of GABAergic signalling following stress exposure may be affected in a sex- and region-specific manner in PNS offspring.


Asunto(s)
Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Neuroesteroides/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Estrés Fisiológico/fisiología , Estrés Psicológico/metabolismo , Animales , Cromatografía Liquida , Femenino , Masculino , Espectrometría de Masas , Neuroesteroides/sangre , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Estrés Psicológico/sangre
18.
J Neurosci ; 29(20): 6449-60, 2009 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-19458216

RESUMEN

The hypothalamus-pituitary-adrenal (HPA) axis is the major neuroendocrine stress response system. Corticotropin-releasing hormone (CRH) neurons in the parvocellular paraventricular nucleus (pPVN) play a key role in coordinating responses of this system to stressors. The cytokine interleukin-1beta (IL-1beta), mimicking infection, robustly activates these CRH neurons via a noradrenergic input arising from the nucleus tractus solitarii (NTS). In late pregnancy, HPA axis responses to stressors, including IL-1beta, are attenuated by a central opioid mechanism that auto-inhibits noradrenaline release in the PVN. Here we show that the neuroactive progesterone metabolite allopregnanolone induces these changes in HPA responsiveness to IL-1beta in pregnancy. In late pregnancy, inhibition of 5alpha-reductase (an allopregnanolone-synthesizing enzyme) with finasteride restored HPA axis responses (rapidly increased pPVN CRH mRNA expression, ACTH, and corticosterone secretion) to IL-1beta. Conversely, allopregnanolone reduced HPA responses in virgin rats. In late pregnancy, activity of the allopregnanolone-synthesizing enzymes (5alpha-reductase and 3alpha-hydroxysteroid dehydrogenase) was increased in the hypothalamus as was mRNA expression in the NTS and PVN. Naloxone, an opioid antagonist, restores HPA axis responses to IL-1beta in pregnancy but had no additional effect after finasteride, indicating a causal connection between allopregnanolone and the endogenous opioid mechanism. Indeed, allopregnanolone induced opioid inhibition over HPA responses to IL-1beta in virgin rats. Furthermore, in virgin rats, allopregnanolone treatment increased, whereas in pregnant rats finasteride decreased proenkephalin-A mRNA expression in the NTS. Thus, in pregnancy, allopregnanolone induces opioid inhibition over HPA axis responses to immune challenge. This novel opioid-mediated mechanism of allopregnanolone action may alter regulation of other brain systems in pregnancy.


Asunto(s)
Analgésicos Opioides/metabolismo , Anestésicos/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Pregnanolona/farmacología , Estrés Fisiológico/efectos de los fármacos , 20-alfa-Dihidroprogesterona/farmacología , Hormona Adrenocorticotrópica/metabolismo , Análisis de Varianza , Animales , Cromatografía en Capa Delgada/métodos , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Finasterida/farmacología , Interleucina-1beta/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidorreductasas/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Embarazo , Preñez/efectos de los fármacos , Progesterona/farmacología , Progestinas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
20.
J Neuroendocrinol ; 30(11): e12644, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30194779

RESUMEN

Sex differences in hypothalamic-pituitary-adrenal (HPA) axis activity are well established in rodents. In addition to glucocorticoids, stress also stimulates the secretion of progesterone and deoxycorticosterone (DOC) from the adrenal gland. Neuroactive steroid metabolites of these precursors can modulate HPA axis function; however, it is not known whether levels of these steroids differ between male and females following stress. In the present study, we aimed to establish whether neuroactive steroid concentrations in the brain display sex- and/or region-specific differences under basal conditions and following exposure to acute stress. Brains were collected from male and female rats killed under nonstress conditions or following exposure to forced swimming. Liquid chromatography-mass spectrometry was used to quantify eight steroids: corticosterone, DOC, dihydrodeoxycorticosterone (DHDOC), pregnenolone, progesterone, dihydroprogesterone (DHP), allopregnanolone and testosterone in plasma, and in five brain regions (frontal cortex, hypothalamus, hippocampus, amygdala and brainstem). Corticosterone, DOC and progesterone concentrations were significantly greater in the plasma and brain of both sexes following stress; however, the responses in plasma were greater in females compared to males. This sex difference was also observed in the majority of brain regions for DOC and progesterone but not for corticosterone. Despite observing no stress-induced changes in circulating concentrations of pregnenolone, DHDOC or DHP, concentrations were significantly greater in the brain and this effect was more pronounced in females than males. Basal plasma and brain concentrations of allopregnanolone were significantly higher in females; moreover, stress had a greater impact on central allopregnanolone concentrations in females. Stress had no effect on circulating or brain concentrations of testosterone in males. These data indicate the existence of sex and regional differences in the generation of neuroactive steroids in the brain following acute stress, especially for the 5α-reduced steroids, and further suggest a sex-specific expression of steroidogenic enzymes in the brain. Thus, differential neurosteroidogenesis may contribute to sex differences in HPA axis responses to stress.


Asunto(s)
Encéfalo/metabolismo , Caracteres Sexuales , Esteroides/metabolismo , Estrés Psicológico/metabolismo , Animales , Corticosterona/sangre , Corticosterona/metabolismo , Femenino , Masculino , Pregnanolona/sangre , Pregnanolona/metabolismo , Pregnenolona/sangre , Pregnenolona/metabolismo , Progesterona/sangre , Progesterona/metabolismo , Ratas Sprague-Dawley , Esteroides/sangre , Estrés Psicológico/sangre , Natación , Testosterona/sangre , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA