Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
Neurobiol Aging ; 135: 60-69, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38185053

RESUMEN

Alzheimer's disease (AD) is more prevalent in women than men, supposing due to the decline of estrogens in menopause, accompanied by increased gonadotropins such as luteinizing hormone (LH). We and others found that the transcription factor early growth response-1 (EGR1) regulates cholinergic function including the expression of acetylcholinesterase (AChE) and plays a significant role in cognitive decline of AD. Here we investigated in APP/PS1 mice by ovariectomy (OVX) and estradiol (E2) supplementation or inhibition of LH the effect on hippocampus-related cognition and related molecular changes. We found that OVX-associated cognitive impairment was accompanied by increased dorsal hippocampal EGR1 expression, which was rescued by downregulating peripheral LH rather than by supplementing E2. We also found in postmortem AD brains a higher expression of pituitary LH-mRNA and higher EGR1 expression in the posterior hippocampus. Both, in human and mice, there was a significant positive correlation between respectively posterior/dorsal hippocampal EGR1 and peripheral LH expression. We conclude that peripheral increased LH and increased posterior hippocampal EGR1 plays a significant role in AD pathology.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Ratones , Femenino , Animales , Humanos , Hormona Luteinizante/metabolismo , Regulación hacia Abajo , Acetilcolinesterasa , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Enfermedad de Alzheimer/metabolismo , Cognición , Ovariectomía , Ratones Transgénicos , Modelos Animales de Enfermedad , Hipocampo/metabolismo
2.
Handb Clin Neurol ; 181: 427-443, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34238476

RESUMEN

Gender identity (an individual's perception of being male or female) and sexual orientation (heterosexuality, homosexuality, or bisexuality) are programmed into our brain during early development. During the intrauterine period in the second half of pregnancy, a testosterone surge masculinizes the fetal male brain. If such a testosterone surge does not occur, this will result in a feminine brain. As sexual differentiation of the brain takes place at a much later stage in development than sexual differentiation of the genitals, these two processes can be influenced independently of each other and can result in gender dysphoria. Nature produces a great variability for all aspects of sexual differentiation of the brain. Mechanisms involved in sexual differentiation of the brain include hormones, genetics, epigenetics, endocrine disruptors, immune response, and self-organization. Furthermore, structural and functional differences in the hypothalamus relating to gender dysphoria and sexual orientation are described in this review. All the genetic, postmortem, and in vivo scanning observations support the neurobiological theory about the origin of gender dysphoria, i.e., it is the sizes of brain structures, the neuron numbers, the molecular composition, functions, and connectivity of brain structures that determine our gender identity or sexual orientation. There is no evidence that one's postnatal social environment plays a crucial role in the development of gender identity or sexual orientation.


Asunto(s)
Identidad de Género , Transexualidad , Femenino , Humanos , Hipotálamo , Masculino , Embarazo , Diferenciación Sexual , Conducta Sexual
3.
Psychoneuroendocrinology ; 117: 104680, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32387876

RESUMEN

BACKGROUND: Classic nuclear-initiated estrogen signaling stimulates corticotropin-releasing hormone (CRH) gene expression as a transcription factor. However, the possible mechanism by which membrane-initiated estrogen signaling (MIES) influences CRH expression remains unclear. There are indications that MIES may upregulate nitric oxide (NO) production through the phosphatidylinositol 3-hydroxy kinase (PI3K) and potentially through the mitogen-activated protein kinase (MAPK) pathway. OBJECTIVES: We investigated the effect of MIES-mediated kinase pathways on CRH expression with or without NO synthesis. METHOD: In SK-N-SH cell culture, estradiol-bovine serum albumin (E2-BSA) was used as the specific membrane estrogen receptor activator, with a specific NO donor, and/or inhibitors for NO synthase (NOS), PI3K, MAPK, protein kinase A (PKA), and protein kinase C (PKC). RESULTS: E2-BSA significantly increased NO and CRH levels in the medium and NOS1-mRNA levels in the cells. In addition, NO donor up-regulated CRH expression, while NOS-inhibitor down-regulated it. When the inhibitor of MAPK and/or the inhibitor of PI3K was added to the medium, only the latter appeared to significantly block the stimulating effect of E2-BSA on NO synthesis, and this was accompanied by an increased CRH expression in the medium. We further studied the effect of the MIES-PKC-mediated pathway on CRH expression, with or without NOS-inhibitor, while the MIES-PKA(-PI3K) pathway served as a control. We found that MIES-PKC upregulated CRH expression independent of NO synthesis. CONCLUSION: MIES can efficiently upregulate CRH expression via various intracellular kinase pathways and may thus be a crucial component in the stress response.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Estradiol/farmacología , Estrógenos/metabolismo , Regulación de la Expresión Génica/fisiología , Óxido Nítrico/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Estrógenos/metabolismo , Albúmina Sérica Bovina/farmacología , Transducción de Señal/fisiología , Células Cultivadas , Humanos
4.
Neurosci Bull ; 35(2): 205-215, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30706412

RESUMEN

The locus coeruleus (LC) has been studied in major depressive disorder (MDD) and bipolar disorder (BD). A major problem of immunocytochemical studies in the human LC is interference with the staining of the immunocytochemical end-product by the omnipresent natural brown pigment neuromelanin. Here, we used a multispectral method to untangle the two colors: blue immunocytochemical staining and brown neuromelanin. We found significantly increased tyrosine hydroxylase (TH) in the LC of MDD patients-thus validating the method-but not in BD patients, and we did not find significant changes in the receptor tyrosine-protein kinase ErbB4 in the LC in MDD or BD patients. We observed clear co-localization of ErbB4, TH, and neuromelanin in the LC neurons. The different stress-related molecular changes in the LC may contribute to the different clinical symptoms in MDD and BD.


Asunto(s)
Trastorno Bipolar/metabolismo , Trastorno Depresivo Mayor/metabolismo , Locus Coeruleus/metabolismo , Melaninas/metabolismo , Receptor ErbB-4/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Anciano , Anciano de 80 o más Años , Trastorno Bipolar/patología , Trastorno Depresivo Mayor/patología , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica/métodos , Locus Coeruleus/patología , Masculino , Microscopía/métodos , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/patología , Sensibilidad y Especificidad , Análisis Espectral/métodos
5.
Elife ; 72018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30179154

RESUMEN

Dysfunction of the noradrenergic (NE) neurons is implicated in the pathogenesis of bipolar disorder (BPD). ErbB4 is highly expressed in NE neurons, and its genetic variation has been linked to BPD; however, how ErbB4 regulates NE neuronal function and contributes to BPD pathogenesis is unclear. Here we find that conditional deletion of ErbB4 in locus coeruleus (LC) NE neurons increases neuronal spontaneous firing through NMDA receptor hyperfunction, and elevates catecholamines in the cerebrospinal fluid (CSF). Furthermore, Erbb4-deficient mice present mania-like behaviors, including hyperactivity, reduced anxiety and depression, and increased sucrose preference. These behaviors are completely rescued by the anti-manic drug lithium or antagonists of catecholaminergic receptors. Our study demonstrates the critical role of ErbB4 signaling in regulating LC-NE neuronal function, reinforcing the view that dysfunction of the NE system may contribute to the pathogenesis of mania-associated disorder.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Conducta Animal , Trastorno Bipolar/metabolismo , Catecolaminas/metabolismo , Eliminación de Gen , Locus Coeruleus/metabolismo , Receptor ErbB-4/metabolismo , Potenciales de Acción/efectos de los fármacos , Neuronas Adrenérgicas/efectos de los fármacos , Animales , Trastorno Bipolar/patología , Peso Corporal , Catecol O-Metiltransferasa/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/metabolismo , Litio/farmacología , Locus Coeruleus/efectos de los fármacos , Ratones , Norepinefrina/metabolismo , Fosforilación/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
6.
Psychoneuroendocrinology ; 95: 34-42, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29793095

RESUMEN

BACKGROUND: Alterations in peripheral sex hormones may play an important role in sex differences in terms of stress responses and mood disorders. It is not yet known whether and how stress-related brain systems and brain sex steroid levels fluctuate in relation to changes in peripheral sex hormone levels, or whether the different sexes show different patterns. We aimed to investigate systematically, in male and female rats, the effect of decreased circulating sex hormone levels following gonadectomy on acute and chronic stress responses, manifested as changes in plasma and hypothalamic sex steroids and hypothalamic stress-related molecules. METHOD: Experiment (Exp)-1: Rats (14 males, 14 females) were gonadectomized or sham-operated (intact); Exp-2: gonadectomized and intact rats (28 males, 28 females) were exposed to acute foot shock or no stressor; and Exp-3: gonadectomized and intact rats (32 males, 32 females) were exposed to chronic unpredictable mild stress (CUMS) or no stressor. For all rats, plasma and hypothalamic testosterone (T), estradiol (E2), and the expression of stress-related molecules were determined, including corticotropin-releasing hormone, vasopressin, oxytocin, aromatase, and the receptors for estrogens, androgens, glucocorticoids, and mineralocorticoids. RESULTS: Surprisingly, no significant correlation was observed in terms of plasma sex hormones, brain sex steroids, and hypothalamic stress-related molecule mRNAs (p > 0.113) in intact or gonadectomized, male or female, rats. Male and female rats, either intact or gonadectomized and exposed to acute or chronic stress, showed different patterns of stress-related molecule changes. CONCLUSION: Diminished peripheral sex hormone levels lead to different peripheral and central patterns of change in the stress response systems in male and female rats. This has implications for the choice of models for the study of the different types of mood disorders which also show sex differences.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Hormonas Esteroides Gonadales/fisiología , Estrés Fisiológico/fisiología , Animales , Aromatasa , Encéfalo/metabolismo , Hormona Liberadora de Corticotropina , Depresión , Trastorno Depresivo , Estradiol/análisis , Femenino , Hipotálamo/metabolismo , Hipotálamo/fisiología , Masculino , Orquiectomía , Ovariectomía , Oxitocina , Ratas , Ratas Sprague-Dawley , Receptores de Esteroides/análisis , Caracteres Sexuales , Factores Sexuales , Testosterona/análisis , Vasopresinas
7.
Neuropsychopharmacology ; 42(10): 2064-2071, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28447621

RESUMEN

Oxytocin (OXT), synthesized in the hypothalamic paraventricular nucleus (PVN) and then released into different brain areas, may play a crucial role in various behaviors and neuropsychiatric disorders, including depression. Testosterone has been proposed by clinical studies to have the opposite effect of oxytocin in these disorders. We began by studying, in the postmortem hypothalamus of fifteen patients with mood disorders and fifteen matched controls, the expression of OXT in the PVN by means of immunocytochemistry (ICC) and the co-localization of OXT and androgen receptor (AR) by means of double labeling ICC. Subsequently, the regulatory effect of AR on OXT gene expression was studied in vitro. We found a higher expression of PVN OXT in the mood disorder patients than in the control subjects, and observed a clear co-localization of AR in OXT-expressing neurons, both in the cytoplasm and in the nucleus. In addition, a significant decrease in OXT-mRNA levels was observed after pre-incubation of the SK-N-SH cells with testosterone. A further potential androgen-responsive element in the human OXT gene promotor was revealed by electrophoretic mobility shift assays and co-transfections in neuroblastoma cells. Finally, in vitro studies demonstrated that AR mediated the down-regulation of OXT gene expression. These results suggest that the fact that OXT and testosterone appear to have opposite effects in neuropsychiatric disorders might be based upon a direct inhibition of AR on OXT transcription, which may provide a novel target for therapeutic strategies in depression.


Asunto(s)
Hipotálamo/metabolismo , Trastornos del Humor/metabolismo , Oxitocina/metabolismo , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/patología , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Citoplasma/patología , Expresión Génica , Humanos , Hipotálamo/patología , Inmunohistoquímica , Trastornos del Humor/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Oxitocina/genética , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Testosterona/administración & dosificación , Testosterona/metabolismo
8.
Physiol Behav ; 145: 118-21, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25846436

RESUMEN

BACKGROUND: Anesthesia administration before sacrificing animals is a common practice in stress-related studies, but the effect of anesthesia on the results remains understudied. We aimed to reveal the interference of different anesthetics, i.e. intraperitoneal (i.p.) sodium-pentobarbital injection or isoflurane inhalation, with the acute stress responses in rats. METHODS: Rats were randomly divided into foot shock (FS) and non-stressed control groups, and further grouped according to the sacrificing procedure: direct decapitation, decapitation after i.p. sodium-pentobarbital injection, or isoflurane inhalation. There was also a non-stressed group sacrificed by decapitation following i.p. saline injection. Plasma levels of corticosterone (CORT), testosterone and estradiol, hypothalamic stress-related molecule mRNA expression of corticotropin-releasing hormone, arginine vasopressin and oxytocin, and frontal lobe stress-related molecule mRNA expression of NMDA receptor subunit NR2B, GABAA receptor and the neuronal-type nicotinic acetylcholine receptor were measured. RESULTS: FS significantly increased plasma CORT levels in direct decapitation and isoflurane groups, while this stress response 'disappeared' following i.p. sodium-pentobarbital injection. In control animals, both the injection of saline and pentobarbital caused a significant increase of plasma CORT. Neither the sex hormone levels nor the mRNA expression of stress-related molecules in the brain showed significant differences among the groups. CONCLUSION: The injection of the anesthetic compound rather than the compound itself may cause extra stress which interferes with the plasma CORT levels, but not with plasma sex hormone levels nor with the brain mRNA expression. Isoflurane inhalation leaves the stress response intact and is also optimal from an ethical point of view.


Asunto(s)
Hipnóticos y Sedantes/uso terapéutico , Isoflurano/uso terapéutico , Pentobarbital/uso terapéutico , Estrés Psicológico/tratamiento farmacológico , Animales , Arginina Vasopresina/genética , Arginina Vasopresina/metabolismo , Corticosterona/sangre , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Oxitocina/genética , Oxitocina/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Estrés Psicológico/sangre
9.
Behav Brain Res ; 284: 231-7, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25687843

RESUMEN

Sex differences play an important role in depression, the basis of which is an excessive stress response. We aimed at revealing the neurobiological sex differences in the same study in acute- and chronically-stressed rats. Female Sprague-Dawley (SD) rats were randomly divided into 6 groups: chronic unpredictable mild stress (CUMS), acute foot shock (FS) and controls, animals in all 3 groups were sacrificed in proestrus or diestrus. Male SD rats were randomly divided into 3 groups: CUMS, FS and controls. Comparisons were made of behavioral changes in CUMS and control rats, plasma levels of corticosterone (CORT), testosterone (T) and estradiol (E2), and of the hypothalamic mRNA-expression of stress-related molecules, i.e. estrogen receptor α and ß, androgen receptor, aromatase, mineralocorticoid receptor, glucocorticoid receptor, corticotropin-releasing hormone, arginine vasopressin and oxytocin. CUMS resulted in disordered estrus cycles, more behavioral and hypothalamic stress-related molecules changes and a stronger CORT response in female rats compared with male rats. Female rats also showed decreased E2 and T levels after FS and CUMS, while male FS rats showed increased E2 and male CUMS rats showed decreased T levels. Stress affects the behavioral, endocrine and the molecular response of the stress systems in the hypothalamus of SD rats in a clear sexual dimorphic way, which has parallels in human data on stress and depression.


Asunto(s)
Ratas Sprague-Dawley/fisiología , Ratas Sprague-Dawley/psicología , Caracteres Sexuales , Estrés Psicológico/fisiopatología , Enfermedad Aguda , Animales , Peso Corporal/fisiología , Enfermedad Crónica , Corticosterona/sangre , Electrochoque , Estradiol/sangre , Ciclo Estral/fisiología , Femenino , Pie , Hipotálamo/fisiopatología , Masculino , ARN Mensajero/metabolismo , Distribución Aleatoria , Testosterona/sangre
10.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 41(2): 125-31, 2012 03.
Artículo en Chino | MEDLINE | ID: mdl-22499507

RESUMEN

OBJECTIVE: To investigate the effect of maternal deprivation on the activity of hypothalamo-pituitary-adrenal (HPA) axis, acute stress response and the sex hormone receptors expression in hypothalamic paraventricular nucleus (PVN) in female rats. METHODS: Maternal deprivation model was induced in female Sprague-Dawley (SD) rats. Foot shock was given at different stages of estrus cycle during the adulthood. Plasma estradiol, testosterone and adrenocorticotropin (ACTH) levels were determined by radioimmunoassay; and plasma corticosterone level was measured by enzyme linked immunosorbent assay. The expression of androgen receptor (AR) and estrogen receptor (ER-ß) in the hypothalamic PVN was detected by immunohistochemistry. RESULTS: Decreased plasma ACTH and corticosterone levels were found in the proestrus of female rats with maternal deprivation (P=0.012 and P=0.019, respectively). A significant down-regulation (P=0.008) of PVN-AR, but not PVN-ER-ß expression was found in female rats with maternal deprivation. CONCLUSION: Maternal deprivation may reduce the HPA axis activity in female SD rats, which is closely correlated with the fluctuation of the circulating sex hormones. The androgen in the hypothalamus seems to play a more important role than the estrogen in this procedure.


Asunto(s)
Sistema Hipotálamo-Hipofisario/fisiopatología , Privación Materna , Sistema Hipófiso-Suprarrenal/fisiopatología , Estrés Fisiológico , Hormona Adrenocorticotrópica/sangre , Animales , Corticosterona/sangre , Estradiol/sangre , Femenino , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Testosterona/sangre
11.
Biol Psychiatry ; 66(9): 832-9, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19596122

RESUMEN

BACKGROUND: Corticotropin-releasing hormone (CRH) is considered the central driving force in the stress response and plays a key role in the pathogenesis of depression. Retinoic acid (RA) has been suggested by clinical studies to be associated with affective disorders. METHODS: First, hypothalamic tissues of 12 patients with affective disorders and 12 matched control subjects were studied by double-label immunofluorescence to analyze the expression of CRH and retinoic acid receptor-alpha (RAR-alpha). Second, critical genes involved in the RA signaling pathways were analyzed in a rat model of depression. Finally, the regulatory effect of RAR-alpha on CRH gene expression was studied in vitro. RESULTS: We found that the expression of RAR-alpha was colocalized with CRH neurons in human hypothalamic paraventricular nucleus (PVN). The density of RAR-alpha-immunoreactive neurons and CRH-RAR-alpha double-staining neurons was significantly increased in the PVN of patients with affective disorders. The ratio of the CRH-RAR-alpha double-staining neurons to the CRH-immunoreactive neurons in affective disorder patients was also increased. Recruitment of RAR-alpha by the CRH promoter was observed in the rat hypothalamus. A dysregulated RA metabolism and signaling was also found in the hypothalamus of a rat model for depression. Finally, in vitro studies demonstrated that RAR-alpha mediated an upregulation of CRH gene expression. CONCLUSIONS: These results suggest that RAR-alpha might contribute to regulating the activity of CRH neurons in vivo, and the vulnerable character of the critical proteins in RA signaling pathways might provide novel targets for therapeutic strategies for depression.


Asunto(s)
Hormona Liberadora de Corticotropina/genética , Trastornos del Humor/metabolismo , Trastornos del Humor/fisiopatología , Receptores de Ácido Retinoico/metabolismo , Regulación hacia Arriba , Adulto , Anciano , Anciano de 80 o más Años , Familia de Aldehído Deshidrogenasa 1 , Animales , Arginina Vasopresina/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Hipotálamo/metabolismo , Masculino , Persona de Mediana Edad , Trastornos del Humor/genética , Neuronas/metabolismo , Ratas , Receptores de Ácido Retinoico/fisiología , Retinal-Deshidrogenasa/metabolismo , Receptor alfa de Ácido Retinoico , Transducción de Señal/genética , Estrés Fisiológico/fisiología , Tretinoina/metabolismo
12.
Neuroendocrinology ; 85(1): 27-36, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17308368

RESUMEN

Previous studies have shown that the total number of corticotropin-releasing hormone (CRH)-stained neurons in the human hypothalamic paraventricular nucleus (PVN) increases with age. To determine whether this age-related change depends on gender and whether circulating sex hormones play a role, we analyzed the total number of CRH-immunoreactive neurons by means of immunocytochemistry and image analysis in the postmortem hypothalamic PVN of 22 control subjects (11 males and 11 females) between the ages of 22 and 89 years, and of 10 subjects with abnormal sex hormone status. Our data show that men have a significantly larger number of CRH neurons than women (p = 0.004) and that the total number of CRH neurons increases significantly with age, but only in male controls (p = 0.032), not in female controls (p = 0.733). Female controls do not show a significant change in the total number of CRH neurons either before or after the age (50 years) of menopause (p = 0.792). Male subjects with low testosterone levels due to castration showed significantly fewer CRH neurons than well-matched intact males (p = 0.008), while castrated male-to-female (M-F) transsexuals with estrogen replacement showed normal numbers of CRH neurons. One male case, who had high estrogen levels due to an estrogen-producing tumor, showed a large number of CRH neurons. Thus, although circulating androgens and estrogens both seem to play a stimulatory role with respect to CRH neurons, the age-dependent increase in the number of CRH neurons in the PVN of men, which has been interpreted to reflect activation of the CRH neurons with age, seems to result from factors other than age-related changes of circulating sex hormone levels.


Asunto(s)
Envejecimiento , Hormona Liberadora de Corticotropina/metabolismo , Hormonas Esteroides Gonadales/fisiología , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Caracteres Sexuales , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Núcleo Hipotalámico Paraventricular/patología
13.
Ageing Res Rev ; 4(2): 141-94, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15996533

RESUMEN

Corticotropin-releasing hormone (CRH) plays a central role in the regulation of the hypothalamic-pituitary-adrenal (HPA)-axis, i.e., the final common pathway in the stress response. The action of CRH on ACTH release is strongly potentiated by vasopressin, that is co-produced in increasing amounts when the hypothalamic paraventricular neurons are chronically activated. Whereas vasopressin stimulates ACTH release in humans, oxytocin inhibits it. ACTH release results in the release of corticosteroids from the adrenal that, subsequently, through mineralocorticoid and glucocorticoid receptors, exert negative feedback on, among other things, the hippocampus, the pituitary and the hypothalamus. The most important glucocorticoid in humans is cortisol, present in higher levels in women than in men. During aging, the activation of the CRH neurons is modest compared to the extra activation observed in Alzheimer's disease (AD) and the even stronger increase in major depression. The HPA-axis is hyperactive in depression, due to genetic factors or due to aversive stimuli that may occur during early development or adult life. At least five interacting hypothalamic peptidergic systems are involved in the symptoms of major depression. Increased production of vasopressin in depression does not only occur in neurons that colocalize CRH, but also in neurons of the supraoptic nucleus (SON), which may lead to increased plasma levels of vasopressin, that have been related to an enhanced suicide risk. The increased activity of oxytocin neurons in the paraventricular nucleus (PVN) may be related to the eating disorders in depression. The suprachiasmatic nucleus (SCN), i.e., the biological clock of the brain, shows lower vasopressin production and a smaller circadian amplitude in depression, which may explain the sleeping problems in this disorder and may contribute to the strong CRH activation. The hypothalamo-pituitary thyroid (HPT)-axis is inhibited in depression. These hypothalamic peptidergic systems, i.e., the HPA-axis, the SCN, the SON and the HPT-axis, have many interactions with aminergic systems that are also implicated in depression. CRH neurons are strongly activated in depressed patients, and so is their HPA-axis, at all levels, but the individual variability is large. It is hypothesized that particularly a subgroup of CRH neurons that projects into the brain is activated in depression and induces the symptoms of this disorder. On the other hand, there is also a lot of evidence for a direct involvement of glucocorticoids in the etiology and symptoms of depression. Although there is a close association between cerebrospinal fluid (CSF) levels of CRH and alterations in the HPA-axis in depression, much of the CRH in CSF is likely to be derived from sources other than the PVN. Furthermore, a close interaction between the HPA-axis and the hypothalamic-pituitary-gonadal (HPG)-axis exists. Organizing effects during fetal life as well as activating effects of sex hormones on the HPA-axis have been reported. Such mechanisms may be a basis for the higher prevalence of mood disorders in women as compared to men. In addition, the stress system is affected by changing levels of sex hormones, as found, e.g., in the premenstrual period, ante- and postpartum, during the transition phase to the menopause and during the use of oral contraceptives. In depressed women, plasma levels of estrogen are usually lower and plasma levels of androgens are increased, while testosterone levels are decreased in depressed men. This is explained by the fact that both in depressed males and females the HPA-axis is increased in activity, parallel to a diminished HPG-axis, while the major source of androgens in women is the adrenal, whereas in men it is the testes. It is speculated, however, that in the etiology of depression the relative levels of sex hormones play a more important role than their absolute levels. Sex hormone replacement therapy indeed seems to improve mood in elderly people and AD patients. Studies of rats have shown that high levels of cumulative corticosteroid exposure and rather extreme chronic stress induce neuronal damage that selectively affects hippocampal structure. Studies performed under less extreme circumstances have so far provided conflicting data. The corticosteroid neurotoxicity hypothesis that evolved as a result of these initial observations is, however, not supported by clinical and experimental observations. In a few recent postmortem studies in patients treated with corticosteroids and patients who had been seriously and chronically depressed no indications for AD neuropathology, massive cell loss, or loss of plasticity could be found, while the incidence of apoptosis was extremely rare and only seen outside regions expected to be at risk for steroid overexposure. In addition, various recent experimental studies using good stereological methods failed to find massive cell loss in the hippocampus following exposure to stress or steroids, but rather showed adaptive and reversible changes in structural parameters after stress. Thus, the HPA-axis in AD is only moderately activated, possibly due to the initial (primary) hippocampal degeneration in this condition. There are no convincing arguments to presume a causal, primary role for cortisol in the pathogenesis of AD. Although cortisol and CRH may well be causally involved in the signs and symptoms of depression, there is so far no evidence for any major irreversible damage in the human hippocampus in this disorder.


Asunto(s)
Envejecimiento/patología , Encéfalo/patología , Trastorno Depresivo/patología , Degeneración Nerviosa/patología , Estrés Fisiológico/patología , Trastorno Depresivo/fisiopatología , Humanos , Degeneración Nerviosa/fisiopatología , Estrés Fisiológico/fisiopatología
14.
Brain ; 128(Pt 6): 1301-13, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15705605

RESUMEN

Oestrogens may modulate the activity of the hypothalamic-pituitary-adrenal (HPA) axis. The present study was to investigate whether the activity of the HPA axis in mood disorders might be directly modulated by oestrogens via oestrogen receptors (ORs) in the corticotropin-releasing hormone (CRH) neurons of the human hypothalamic paraventricular nucleus (PVN). Brains of 13 subjects ranging in age between 45 and 79 years suffering from major depression/major depressive disorder (eight cases) or bipolar disorder (five cases) and of 13 controls, matched for sex, age, brain weight, post-mortem delay, fixation time and season and clock time at death, were studied with double-label immunocytochemistry. The total number of CRH-immunoreactive (IR) neurons, CRH neurons that colocalized ORalpha in the neuronal nucleus and the number of only nuclear ORalpha-containing neurons in the PVN were measured using an image analysis system. In addition, the volume of the PVN delineated on the basis of CRH neurons was determined. It was found that the total number of CRH-IR neurons in patients with mood disorders was nearly 1.7 times higher than in controls (P = 0.034). A novel finding was that the total number of CRH-IR neurons and the number of CRH-nuclear ORalpha double-staining neurons in the PVN were strongly correlated both in controls and in patients with mood disorders (P < 0.001 and P = 0.022, respectively). The ratio of the CRH-nuclear-ORalpha double-staining neurons to the total CRH-IR neurons in patients with mood disorders was similar to that in the controls (P = 0.448). The volume of the sub-region of the PVN that was delineated on the basis of CRH neurons was significantly larger in patients with mood disorders than in controls (P = 0.022). Another novel finding was the large population of extra-hypothalamic CRH neurons that was found in the thalamus. In summary, oestrogens may directly influence CRH neurons in the human PVN. The increased numbers of neurons expressing CRH in mood disorders is accompanied by increased ORalpha colocalization in the nucleus of these neurons. These changes seem to be trait- rather than state-related.


Asunto(s)
Hormona Liberadora de Corticotropina/análisis , Receptor alfa de Estrógeno/análisis , Trastornos del Humor/metabolismo , Núcleo Hipotalámico Paraventricular/química , Anciano , Encéfalo/patología , Núcleo Celular/química , Femenino , Humanos , Sistema Hipotálamo-Hipofisario/fisiopatología , Procesamiento de Imagen Asistido por Computador , Masculino , Persona de Mediana Edad , Trastornos del Humor/patología , Trastornos del Humor/fisiopatología , Neuronas/química , Neuronas/patología , Tamaño de los Órganos , Núcleo Hipotalámico Paraventricular/patología , Sistema Hipófiso-Suprarrenal/fisiopatología , Coloración y Etiquetado/métodos
15.
Horm Behav ; 45(2): 93-102, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15019795

RESUMEN

To investigate whether depression is accompanied by changes in diurnal rhythms of free estradiol and cortisol in different phases of the menstrual cycle, we measured these two hormone levels in saliva samples collected every 2 h for 24 h from 15 healthy normally cycling women and 12 age-matched normally cycling women suffering from major depression taking antidepressants. The assessments were repeated four times over one menstrual cycle: during menstruation and in the late follicular/peri-ovulating, early to mid-luteal and late luteal phases, respectively. Quantification with a nonlinear periodic regression model revealed distinct diurnal rhythms in free estradiol and free cortisol in all subjects. For the diurnal cortisol rhythm, significant differences were found in the peak-width and ultradian amplitude among different menstrual phases, both in controls and depressed patients, while no significant differences were found between the two groups. The diurnal estradiol rhythm, on the other hand, was quite consistent among different menstrual phases within both groups, while the depressed patients had overall larger amplitudes than controls, which is negatively correlated with disease duration. Significant positive correlations between the two hormone rhythms were found for 24-h mean level (mesor), peak, and trough in late luteal phase, and for ultradian harmonics in early to mid-luteal phase in controls, but only for ultradian harmonics in late follicular/peri-ovulating phase and for acrophase in the menstruation phase in depressed patients. A sub-analysis was also performed in patients who received Fluoxetine (n = 7). The findings implicate a close correlation between the hypothalamic-pituitary-adrenal axis and the hypothalamic-pituitary-gonadal axis, both of which may be involved in depression.


Asunto(s)
Ritmo Circadiano/fisiología , Trastorno Depresivo Mayor/fisiopatología , Estradiol/metabolismo , Hidrocortisona/metabolismo , Ciclo Menstrual/metabolismo , Adulto , Análisis de Varianza , Estradiol/análisis , Femenino , Humanos , Hidrocortisona/análisis , Análisis por Apareamiento , Valores de Referencia , Saliva/química
16.
Eur J Endocrinol ; 148(2): 227-32, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12590642

RESUMEN

OBJECTIVE: To investigate the diurnal rhythm of estrogens in normally cyclic women during reproductive life. DESIGN: Multiple saliva sampling in normally cyclic healthy women during reproductive life at different phases of their menstrual cycles was carried out. METHODS: Salivary estradiol was measured by radioimmunoassay in samples collected every 2 h for 24 h from 15 normally cyclic healthy women during reproductive life during the menstrual phase, the late follicular/peri-ovulation phase, the early to mid luteal phase and the late luteal phase, respectively, of their menstrual cycles. The levels of salivary estradiol were analyzed by means of periodic regression. RESULTS: A daily biological rhythm of free estradiol was found after quantification with a nonlinear periodic regression model. The observed diurnal free estradiol rhythm consists of two major components: an asymmetrically peaked diurnal cycle and ultradian harmonics in the range of 6 to 12 h. The diurnal and ultradian rhythms were remarkably consistent throughout the menstrual cycle in terms of mesor (24 h mean level), peak width and amplitude. There was a tendency for the 24-h rhythm acrophases to converge in the early morning, while the acrophase of the menstrual phase occurred significantly later than in the late follicular/peri-ovulation phase. CONCLUSIONS: The diurnal rhythm of estradiol has a similar complex temporal organization for different menstrual phases. The menstrual cycle mainly modulates the acrophase of the diurnal rhythm.


Asunto(s)
Ritmo Circadiano , Estradiol/metabolismo , Ciclo Menstrual/metabolismo , Saliva/metabolismo , Ciclos de Actividad , Adulto , Femenino , Humanos , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA