Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 257
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Front Neuroendocrinol ; 65: 100974, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34995643

RESUMEN

Estradiol and hypothalamic paraventricular nucleus (PVN) help coordinate reproduction with body physiology, growth and metabolism. PVN integrates hormonal and neural signals originating in the periphery, generating an output mediated both by its long-distance neuronal projections, and by a variety of neurohormones produced by its magnocellular and parvocellular neurosecretory cells. Here we review the cyto-and chemo-architecture, the connectivity and function of PVN and the sex-specific regulation exerted by estradiol on PVN neurons and on the expression of neurotransmitters, neuromodulators, neuropeptides and neurohormones in PVN. Classical and non-classical estrogen receptors (ERs) are expressed in neuronal afferents to PVN and in specific PVN interneurons, projecting neurons, neurosecretory neurons and glial cells that are involved in the input-output integration and coordination of neurohormonal signals. Indeed, PVN ERs are known to modulate body homeostatic processes such as autonomic functions, stress response, reproduction, and metabolic control. Finally, the functional implications of the estrogenic modulation of the PVN for body homeostasis are discussed.


Asunto(s)
Neuropéptidos , Núcleo Hipotalámico Paraventricular , Estradiol/metabolismo , Femenino , Humanos , Masculino , Neuronas/metabolismo , Neuropéptidos/metabolismo , Sistemas Neurosecretores/metabolismo , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo
2.
Front Neuroendocrinol ; 52: 44-64, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30223003

RESUMEN

Synthetic selective modulators of the estrogen receptors (SERMs) have shown to protect neurons and glial cells against toxic insults. Among the most relevant beneficial effects attributed to these compounds are the regulation of inflammation, attenuation of astrogliosis and microglial activation, prevention of excitotoxicity and as a consequence the reduction of neuronal cell death. Under pathological conditions, the mechanism of action of the SERMs involves the activation of estrogen receptors (ERs) and G protein-coupled receptor for estrogens (GRP30). These receptors trigger neuroprotective responses such as increasing the expression of antioxidants and the activation of kinase-mediated survival signaling pathways. Despite the advances in the knowledge of the pathways activated by the SERMs, their mechanism of action is still not entirely clear, and there are several controversies. In this review, we focused on the molecular pathways activated by SERMs in brain cells, mainly astrocytes, as a response to treatment with raloxifene and tamoxifen.


Asunto(s)
Astrocitos/efectos de los fármacos , Encefalopatías/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Clorhidrato de Raloxifeno/farmacología , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Animales , Humanos
3.
J Neuroinflammation ; 15(1): 337, 2018 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518432

RESUMEN

BACKGROUND: Perinatal maternal malnutrition is related to altered growth of tissues and organs. The nervous system development is very sensitive to environmental insults, being the hippocampus a vulnerable structure, in which altered number of neurons and granular cells has been observed. Moreover, glial cells are also affected, and increased expression of proinflammatory mediators has been observed. We studied the effect of Glucagon-like peptide-1 receptor (GLP-1R) agonists, liraglutide, which have very potent metabolic and neuroprotective effects, in order to ameliorate/prevent the glial alterations present in the hippocampus of the pups from mothers with food restriction during pregnancy and lactation (maternal perinatal food restriction-MPFR). METHODS: Pregnant Sprague-Dawley rats were randomly assigned to 50% food restriction (FR; n = 12) or ad libitum controls (CT, n = 12) groups at day of pregnancy 12 (GD12). From GD14 to parturition, pregnant FR and CT rats were treated with liraglutide (100 µg/kg) or vehicle. At postnatal day 21 and before weaning, 48 males and 45 females (CT and MPFR) were sacrificed. mRNA expression levels of interleukin-1ß (IL1ß), interleukin-6 (IL-6), nuclear factor-κß, major histocompatibility complex-II (MHCII), interleukin 10 (IL10), arginase 1 (Arg1), and transforming growth factor (TGFß) were assessed in the hippocampus by quantitative real-time polymerase chain reaction. Iba1 and GFAP-immunoreactivity were assessed by immunocytochemistry. RESULTS: The mRNA expression IL1ß, IL6, NF-κB, and MHCII increased in the hippocampus of male but not in female pups from MPFR. In addition, there was an increase in the percentage of GFAP and Iba1-immupositive cells in the dentate gyrus compared to controls, indicating an inflammatory response in the brain. On the other hand, liraglutide treatment prevented the neuroinflammatory process, promoting the production of anti-inflammatory molecules such as IL10, TGFß, and arginase 1, and decreasing the number and reactivity of microglial cells and astrocytes in the hippocampus of male pups. CONCLUSION: Therefore, the GLP-1 analog, liraglutide, emerges as neuroprotective drug that minimizes the harmful effects of maternal food restriction, decreasing neuroinflammation in the hippocampus in a very early stage.


Asunto(s)
Antiinflamatorios/uso terapéutico , Privación de Alimentos , Hipocampo/metabolismo , Liraglutida/uso terapéutico , Efectos Tardíos de la Exposición Prenatal/patología , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Peso Corporal/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Citocinas/metabolismo , Femenino , Edad Gestacional , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Proteínas de Microfilamentos/metabolismo , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores Sexuales
4.
Horm Behav ; 61(4): 512-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22285935

RESUMEN

Estradiol and some selective estrogen receptor modulators (SERMs) are neuroprotective in a variety of experimental models of neurodegeneration, reduce the inflammatory response of glial cells, reduce anxiety and depression, promote cognition and modulate synaptic plasticity in the hippocampus of rodents. In this study we have assessed whether estradiol and two SERMs currently used in clinics, tamoxifen and raloxifene, affect medial prefrontal cortex function and morphology. Rats were ovariectomized and six days later some animals received a subcutaneous injection of the estrogenic compounds. In a first experiment animals were treated with estradiol benzoate or sesame oil vehicle. In a second experiment animals received raloxifene, tamoxifen or dimethyl sulfoxide as vehicle. Twenty four hours after the pharmacological treatment, animals were challenged to solve an allocentric working memory paradigm in a "Y" maze. Twenty trials consisting of a study phase and a test phase were conducted according to a delayed match-to-sample procedure in a single one-day session. Animals that were not submitted to behavioral test were used for Golgi analysis of the prefrontal cortex. Rats treated with estradiol benzoate, tamoxifen or raloxifene performed better in the Y maze and showed a significant increase in the numerical density of dendritic spines in secondary apical dendrites of layer III pyramidal neurons from the prelimbic/infralimbic prefrontal cortex, compared to their respective control groups. These findings suggest that estradiol, tamoxifen and raloxifene improve prefrontal cortex-related cognitive performance and modulate prefrontal cortex morphology in ovariectomized rats.


Asunto(s)
Espinas Dendríticas/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Ovariectomía , Corteza Prefrontal/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Animales , Conducta Animal/efectos de los fármacos , Estradiol/análogos & derivados , Estradiol/farmacología , Femenino , Aprendizaje por Laberinto/efectos de los fármacos , Corteza Prefrontal/citología , Desempeño Psicomotor/efectos de los fármacos , Clorhidrato de Raloxifeno/farmacología , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos , Tamoxifeno/farmacología
5.
Glia ; 58(1): 93-102, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19533603

RESUMEN

Expression of proinflammatory molecules by glial cells is involved in the pathophysiological changes associated with chronic neurological diseases. Under pathological conditions, astrocytes release a number of proinflammatory molecules, such as interleukin-6 (IL-6) and interferon-gamma-inducible protein-10 (IP-10). The ovarian hormone estradiol exerts protective effects in the central nervous system that, at least in part, may be mediated by a reduction of local inflammation. This study was designed to assess whether estradiol affects the production of IL-6 and IP-10 by primary cultures of newborn mice astrocytes exposed to lipopolysaccharide (LPS), a bacterial endotoxin known to cause neuroinflammation. In addition, the possible anti-inflammatory effect of several selective estrogen receptor modulators (SERMs) was also assessed. LPS induced an increase in the expression of IL-6 and IP-10 mRNA levels in astrocytes and an increase in IL-6 and IP-10 protein levels in the culture medium. These effects of LPS were impaired by estradiol and by the four SERMs tested in our study: tamoxifen, raloxifene, ospemifene, and bazedoxifene. All SERMs tested showed a similar effect on IL-6 and IP-10 mRNA levels, but raloxifene and ospemifene were more effective than tamoxifen and bazedoxifene in reducing protein levels in LPS-treated cultures. Finally, we report that news SERMs, ospemifene and bazedoxifene, exert anti-inflammatory actions by a mechanism involving classical estrogen receptors and by the inhibition of LPS-induced NFkappaB p65 transactivation. The results suggest that estrogenic compounds may be candidates to counteract brain inflammation under neurodegenerative conditions by targeting the production and release of proinflammatory molecules by astrocytes.


Asunto(s)
Astrocitos , Quimiocina CXCL10/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/metabolismo , Receptores de Estrógenos/fisiología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Astrocitos/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Quimiocina CXCL10/genética , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Estrógenos/farmacología , Regulación de la Expresión Génica/inmunología , Interleucina-6/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Receptores de Estrógenos/clasificación
6.
J Steroid Biochem Mol Biol ; 203: 105732, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32777355

RESUMEN

Gastrointestinal function is known to be regulated by steroid molecules produced by the gonads, the adrenal glands and the gut microbiota. However, we have a limited knowledge on the functional significance of local steroid production by gastrointestinal tract tissue. On this basis, we have here evaluated, as a first methodological approach, the expression of steroidogenic molecules and the local levels of key steroids in the male rat colon. Our findings indicate that the colon tissue expresses molecules involved in the early steps of steroidogenesis and in the consecutive synthesis and metabolism of steroid hormones, such as progesterone, testosterone and 17ß-estradiol. In addition, the levels of the steroid hormone precursor pregnenolone and the levels of active metabolites of progesterone and testosterone, such as dihydroprogesterone, tetrahydroprogesterone, dihydrotestosterone and 17ß-estradiol, were higher in colon than in plasma. Higher levels of the androgen metabolite 3α-diol were detected in the colon in comparison with another non-classical steroidogenic tissue, such as the cerebral cortex. These findings suggest the existence of local steroid synthesis and metabolism in the colon, with the production of active steroid metabolites that may impact on the activity of the enteric nervous system and on the composition of the gut microbiota.


Asunto(s)
Colon/metabolismo , Esteroides/metabolismo , Animales , Corteza Cerebral/metabolismo , Masculino , Ratas Sprague-Dawley , Esteroides/sangre
7.
J Steroid Biochem Mol Biol ; 199: 105596, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31958635

RESUMEN

Neuroactive steroids, molecules produced from cholesterol in steroidogenic cells (i.e., peripheral glands and nervous system) are physiological modulators and protective agents of nervous function. A possible role for neuroactive steroids in the sex-dimorphic clinical manifestation, onset and progression of Multiple Sclerosis (MS) has been recently suggested. To explore this possibility, we assessed the synthesis of the first steroidogenic product (pregnenolone; PREG) in the spinal cord of experimental autoimmune encephalomyelitis rats, a MS model. Data obtained indicate that the synthesis of PREG in the spinal cord is altered by the pathology in a sex-dimorphic way and depending on the pathological progression. Indeed, in male spinal cord the synthesis was already decreased at the acute phase of the disease (i.e., 14 days post induction - dpi) and maintained low during the chronic phase (i.e., 45 dpi), while in females this effect was observed only at the chronic phase. Substrate availability had also a role in the sex-dimorphic kinetics. Indeed, at the chronic phase, male animals showed a reduction in the levels of free cholesterol coupled to alteration of cholesterol metabolism into oxysterols; these effects were not observed in female animals. These findings suggest that the comprehension of the neurosteroidogenic processes could be relevant to better understand the sexual dimorphism of MS and to possibly design sex-oriented therapeutic strategies based on neuroactive steroids.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Esclerosis Múltiple/metabolismo , Pregnenolona/metabolismo , Médula Espinal/metabolismo , Animales , Colesterol/biosíntesis , Colesterol/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Cinética , Masculino , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Neuroesteroides/metabolismo , Pregnenolona/biosíntesis , Ratas , Caracteres Sexuales , Médula Espinal/patología , Especificidad por Sustrato
8.
Ageing Res Rev ; 54: 100943, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31430564

RESUMEN

The increase in life expectancy of the world population is associated with a higher prevalence of neurodegenerative diseases. Alzheimer's Disease (AD) is the most common neurodegenerative disease, affecting currently 43 million people over the world. To date, most of the pharmacological interventions in AD are intended for the alleviation of some of its symptoms, and there are no effective treatments to inhibit the progression of the disease. Translocator protein (TSPO) is present in contact points between the outer and the inner mitochondrial membranes and is involved in the control of steroidogenesis, inflammation and apoptosis. In the last decade, studies have shown that TSPO ligands present neuroprotective effects in different experimental models of AD, both in vitro and in vivo. The aim of this review is to analyze the data provided by these studies and to discuss if TSPO could be a viable therapeutic target for the development of new treatments for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Inflamación , Fármacos Neuroprotectores/farmacología , Receptores de GABA/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Apoptosis , Humanos , Ligandos , Fármacos Neuroprotectores/uso terapéutico , Receptores de GABA/efectos de los fármacos
9.
Physiol Behav ; 199: 386-394, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30529512

RESUMEN

It is well known that inflammatory challenge during the prenatal period results in permanent changes in glial cells and behavior in adulthood. However, it is unknown whether inflammatory challenge during the infantile period may have permanent sexually-dimorphic effects on microglia and astrocytes in vivo, which in turn may be associated with sex differences in adult behavior. In this study, we have evaluated whether postnatal injection of lipopolysaccharide (LPS; 250µg/kg, i.p. on postnatal day 14) induces depressive and less anxiety-like behaviors, glial cell activation, pro-inflammatory cytokine (TNF-alpha) secretion and sexually dimorphic responses in adulthood. Postnatal day 14 (P14) male and female Wistar rats received an intraperitoneal (ip) injection of LPS or PBS. Three months later, animals were tested in the Open Field (OF), the Elevated Plus Maze (EPM) and the Forced Swimming Test (FST) to assess the level of anxiety and depression-like behavior. Hippocampal proinflammatory cytokine TNF-alpha concentration and the number of astrocytes and microglia were estimated in the dentate gyrus, CA1, and CA3 in two regions of the hippocampus (ventral and dorsal). Our results showed that the administration of LPS resulted in less anxiety and depression-like behavior in males but not in females. However, the LPS-administration increased the number of microglia in the dorsal and ventral hippocampus areas in females more than male, while no significant differences in TNFα level had been detected between the LPS-rats treated and their controls. Interestingly, LPS resulted in an increase in the number of astrocytes in both areas of the hippocampus in a female. While in a male, our results showed a decrease in astrocytes number in the dorsal hippocampus, but no significant differences observed in ventral hippocampus. These findings indicate that an immune challenge in infantile rats induces a ventral and dorsal hippocampus damage in female more than in male, without affecting significantly the affective behavior changes in the female. The results also showed that small changes in the male hippocampus can affect the behavior and induce a depression-like behavior.


Asunto(s)
Astrocitos/efectos de los fármacos , Hipocampo/efectos de los fármacos , Microglía/efectos de los fármacos , Caracteres Sexuales , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Ansiedad/metabolismo , Astrocitos/metabolismo , Conducta Animal/efectos de los fármacos , Recuento de Células , Depresión/metabolismo , Femenino , Hipocampo/metabolismo , Lipopolisacáridos/farmacología , Masculino , Microglía/metabolismo , Embarazo , Ratas , Ratas Wistar
10.
J Neuroendocrinol ; 30(2)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28891264

RESUMEN

Brain expression of the enzyme P450-aromatase has been studied extensively. Subsequent to the aromatisation hypothesis having established brain aromatase as a key factor to convert gonadal testosterone to oestradiol, several studies have investigated the regulation of aromatase during the critical period of brain sexual differentiation. We review previous and recent findings concerning regulation of aromatase. The role of gonadal hormones, sex chromosome genes and neurosteroids is analysed in terms of their contribution to aromatase expression, as well as implications for the organisational effect of steroids during development.


Asunto(s)
Aromatasa/metabolismo , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hormonas Esteroides Gonadales/metabolismo , Diferenciación Sexual/fisiología , Animales , Aromatasa/genética , Encéfalo/embriología , Femenino , Masculino
11.
Biomed Pharmacother ; 99: 655-663, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29710462

RESUMEN

Thymelaea lythroides extract is widely used as a traditional folk medicine in Morocco, especially for the treatment of diabetes, rheumatism and Inflammatory disease. The aim of the study is to evaluate the possible effect of methanolic extract of Thymelaea lythroides in repressing the inflammatory responses and long-lasting depression-like behavior associated with neuroinflammation in adult rats after neonatal LPS exposure. Male rat pups were treated systemically with either LPS (250??g/kg) or vehicle (phosphate buffer saline) on postnatal day 14. Six hours later, the LPS groups were assigned to intraperitoneal (ip) injection of Minocycline (50?mg/kg) or Thymelaea lythroides (200?mg/kg). Thereafter, in adulthood (postnatal days 90-97), the spontaneous locomotor activity and depression-like behavior were assessed successively in open field and forced swim tests. The levels of proinflammatory cytokines, oxidative damage, and activation of microglia were determined in the hippocampus (HP) of male rats on (PND90-97). Our results showed that open field hypoactivity and increased immobility period in LPS-induced adult rats were normalized on treatment with Thymelaea lythroides and minocycline. Both treatments attenuate the overactivated microglial cells in the CA1 and CA3 of hippocampus (HP) and significantly reduced the oxidative-nitrosative stress markers and cytokine (TNF ?) production in the HP. Thymelaea lythroides seems to have similar neuroprotective effects to Minocycline, and such protection may be due to: reduction of oxidative stress, upregulation of inflammatory mediators production, antidepressant behavior which all are associated with neuroinflammation.


Asunto(s)
Depresión/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Extractos Vegetales/farmacología , Thymelaeaceae/química , Animales , Antidepresivos/aislamiento & purificación , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Citocinas/metabolismo , Depresión/fisiopatología , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Inflamación/patología , Lipopolisacáridos , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Minociclina/farmacología , Fármacos Neuroprotectores/aislamiento & purificación , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar
12.
Neuroscience ; 144(4): 1288-92, 2007 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-17161546

RESUMEN

The hypothalamic arcuate nucleus integrates different hormonal and neural signals to control neuroendocrine events, feeding, energy balance and reproduction. Previous studies have shown that in adult female rats the arcuate nucleus undergoes a cyclic fluctuation in the number of axo-somatic synapses during the estrous cycle, in parallel to the variation of ovarian hormone levels in plasma. In the present study we have used an unbiased stereological analysis in conjunction with postembedding immunocytochemistry to assess whether the synaptic remodeling during the estrous cycle in rats is specific for certain types of synapses. Our findings indicate that there is a significant decrease in the number of GABAergic axo-somatic synapses on proestrus afternoon and estrus day compared with other days of the estrous cycle. This decrease in GABAergic synapses is accompanied by an increase in the number of dendritic spine synapses. The synaptic density appears to cycle back to proestrus morning values on metestrus day. In contrast, the number of synapses on dendritic shafts does not change during the cycle. These results indicate that a rapid and selective synaptic turnover of arcuate synapses occurs in physiological circumstances.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Ciclo Estral/fisiología , Hormonas Esteroides Gonadales/metabolismo , Plasticidad Neuronal/fisiología , Sinapsis/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/ultraestructura , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Femenino , Metestro/metabolismo , Microscopía Inmunoelectrónica , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Proestro/metabolismo , Ratas , Ratas Wistar , Sinapsis/ultraestructura , Ácido gamma-Aminobutírico/metabolismo
13.
Neuroscience ; 144(4): 1293-304, 2007 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-17187935

RESUMEN

One important complication of diabetes is damage to the peripheral nervous system. However, in spite of the number of studies on human and experimental diabetic neuropathy, the current therapeutic arsenal is meagre. Consequently, the search for substances to protect the nervous system from the degenerative effects of diabetes has high priority in biomedical research. Neuroactive steroids might be interesting since they have been recently identified as promising neuroprotective agents in several models of neurodegeneration. We have assessed whether chronic treatment with progesterone (P), dihydroprogesterone (DHP) or tetrahydroprogesterone (THP) had neuroprotective effects against streptozotocin (STZ)-induced diabetic neuropathy at the neurophysiological, functional, biochemical and neuropathological levels. Using gas chromatography coupled to mass-spectrometry, we found that three months of diabetes markedly lowered P plasma levels in male rats, and chronic treatment with P restored them, with protective effects on peripheral nerves. In the model of STZ-induced of diabetic neuropathy, chronic treatment for 1 month with P, or with its derivatives, DHP and THP, counteracted the impairment of nerve conduction velocity (NCV) and thermal threshold, restored skin innervation density, and improved Na(+),K(+)-ATPase activity and mRNA levels of myelin proteins, such as glycoprotein zero and peripheral myelin protein 22, suggesting that these neuroactive steroids, might be useful protective agents in diabetic neuropathy. Interestingly, different receptors seem to be involved in these effects. Thus, while the expression of myelin proteins and Na(+),K(+)-ATPase activity are only stimulated by P and DHP (i.e. two neuroactive steroids interacting with P receptor, PR), NCV, thermal nociceptive threshold and intra-epidermal nerve fiber (IENF) density are also affected by THP, which interacts with GABA-A receptor. Because, a therapeutic approach with specific synthetic receptor ligands could avoid the typical side effects of steroids, future experiments will be devoted to evaluating the role of PR and GABA-A receptor in these protective effects.


Asunto(s)
Neuropatías Diabéticas/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Nervios Periféricos/efectos de los fármacos , Nervios Periféricos/metabolismo , Progesterona/farmacología , 20-alfa-Dihidroprogesterona/farmacología , 20-alfa-Dihidroprogesterona/uso terapéutico , Animales , Diabetes Mellitus Experimental/complicaciones , Neuropatías Diabéticas/fisiopatología , Neuropatías Diabéticas/prevención & control , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Masculino , Proteínas de la Mielina/genética , Conducción Nerviosa/efectos de los fármacos , Conducción Nerviosa/fisiología , Fármacos Neuroprotectores/uso terapéutico , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Nervios Periféricos/fisiopatología , Pregnanolona/farmacología , Pregnanolona/uso terapéutico , Progesterona/sangre , Progesterona/uso terapéutico , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/metabolismo , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Piel/inervación , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Resultado del Tratamiento
14.
J Steroid Biochem Mol Biol ; 171: 281-287, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28442392

RESUMEN

The translocator protein (TSPO) is an outer mitochondrial membrane protein involved in the transport of cholesterol into the mitochondria, which is the first step for the synthesis of steroid hormones, as well as in the regulation of mitochondrial permeability transition pore opening and apoptosis. Studies have shown that the activation of TSPO may promote neuroprotective actions in experimental models of neurodegeneration and brain injury. In a previous study, our group showed that 4'-chlorodiazepam (4'-CD), a TSPO ligand, was neuroprotective against amyloid-beta (Aß) in SHSY-5Y neuroblastoma cells. The aim of this study was to evaluate if 4'-CD was also neuroprotective against Aß in organotypic hippocampal cultures and to identify its mechanisms of action. Aß decreased the cell viability of organotypic hippocampal cultures, while 4'-CD had a neuroprotective effect when administered at 100nM and 1000nM. The neuroprotective effects of 4'-CD against Aß were associated with an increased expression of superoxide dismutase (SOD). No differences were found in the expression of catalase, glial fibrillary acidic protein, Akt and procaspase-3. In summary, our results show that 4'-CD is neuroprotective against Aß by a mechanism involving the modulation of SOD protein expression.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Benzodiazepinonas/farmacología , Hipocampo/efectos de los fármacos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Péptidos beta-Amiloides/metabolismo , Animales , Antioxidantes/farmacología , Biomarcadores/metabolismo , Proteínas Portadoras/metabolismo , Supervivencia Celular/efectos de los fármacos , Hipocampo/metabolismo , Ligandos , Masculino , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Nootrópicos/farmacología , Concentración Osmolar , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo , Ratas Wistar , Receptores de GABA-A/metabolismo , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/metabolismo , Técnicas de Cultivo de Tejidos , Regulación hacia Arriba/efectos de los fármacos
15.
Neurosci Lett ; 639: 98-102, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28041963

RESUMEN

The translocator protein 18kDa (TSPO) is located in the outer mitochondrial membrane and is involved in the cholesterol transport into the mitochondria and in the regulation of steroidogenesis and other mitochondrial functions. It is known that steroid hormones, such as estradiol, testosterone and dihydrotestosterone are neuroprotective and regulate neuritogenesis in the CNS by different mechanisms. However, the developmental effects of TSPO ligands in the CNS are not known. Therefore, the aim of this study was to identify the developmental effects of 4'-chlorodiazepam (4'-CD), a TSPO ligand, in primary cultures of male and female mouse hippocampal neurons. We observed that female neurons showed an advanced neuritogenesis compared to male neurons after 2days in vitro. Moreover, it was shown that 4'-CD administration accelerated the maturation of male hippocampal neurons, without changing the development of female neurons. These findings, showing that 4'-CD modulates the development of hippocampal neurons in a sex-dependent manner, suggest that TSPO may be involved in the regulation of neuritogenesis.


Asunto(s)
Benzodiazepinonas/farmacología , Hipocampo/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Transporte Biológico , Femenino , Hipocampo/crecimiento & desarrollo , Masculino , Ratones , Mitocondrias/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Neuronas/metabolismo , Receptores de GABA/metabolismo , Caracteres Sexuales
16.
Neuroscience ; 138(3): 741-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16338087

RESUMEN

The nervous system synthesizes steroids that regulate the development and function of neurons and glia, and have neuroprotective properties. The first step in steroidogenesis involves the delivery of free cholesterol to the inner mitochondrial membrane where it can be converted into pregnenolone by the enzyme cytochrome P450side chain cleavage. The peripheral-type benzodiazepine receptor and the steroidogenic acute regulatory protein are involved in this process and appear to function in a coordinated manner. Steroidogenic acute regulatory protein mRNA and protein are widely expressed throughout the adult brain. Steroidogenic acute regulatory protein expression has been detected in many neuronal populations, in ependymocytes, in some astroglial cells, in Schwann cells from peripheral nerves and in proliferating cells of the developing and adult brain. Steroidogenic acute regulatory protein is colocalized in the same neural cells with P450side chain cleavage and with other steroidogenic enzymes. Steroidogenic acute regulatory protein expression in the brain shows marked changes with development, aging and injury. The steroidogenic acute regulatory protein gene may be under the control of diverse mechanisms in different neural cell types, since its expression is upregulated by cyclic AMP (cAMP) in gliomas and astrocytes in culture and downregulated by cyclic AMP (cAMP) in Schwann cells. In addition, activation of N-methyl-D-aspartate receptors, and the consequent rise in intracellular calcium levels, activates steroidogenic acute regulatory protein and steroidogenesis in hippocampal neurons. In conclusion, steroidogenic acute regulatory protein is regulated in the nervous system by different physiological and pathological conditions and may play an important role during brain development, aging and after injury.


Asunto(s)
Encéfalo/fisiología , Fosfoproteínas/fisiología , Animales , Astrocitos/fisiología , Regulación de la Expresión Génica , Glioma/genética , Humanos , Fosfoproteínas/genética , ARN Mensajero/genética , Células de Schwann/fisiología
17.
Neuroscience ; 138(3): 801-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16310968

RESUMEN

Historically, morphological studies of the distribution of androgen receptors in the brain led to conclusions that the major regional targets of androgen action are involved in reproduction, that the primary cellular targets are neurons, and that functional androgen receptors are exclusively nuclear, consistent with the classical view of steroid receptors as ligand-dependent transcription factors. In this review, we discuss three separate but interrelated recent studies highlighting observations made with newer methodologies while assessing the regional, cellular or subcellular distribution of androgen receptors containing cells in the forebrain. Regional studies demonstrated that the largest forebrain target for androgen action in terms of the number of androgen receptor expressing cells is the cerebral cortex, rather than the main hypothalamic and limbic centers for reproductive function. Cellular studies to determine the phenotype of androgen receptor expressing cells confirmed that most of these cells are neurons but also revealed that small subpopulations are astrocytes. The expression of androgen receptors in astrocytes is both region and age dependent. In contrast, reactive astrocytes in the lesioned adult rat brain do not express androgen receptors whereas reactive microglia do. Finally, androgen receptor immunoreactive axons were identified in the cerebral cortex of the rat and human. These observations do not overturn classical views of the cellular and subcellular locus of steroid action in the nervous system, but rather broaden our view of the potential direct impact of gonadal steroid hormones on cellular function and emphasize the regional and developmental specificity of these effects on the nervous system.


Asunto(s)
Andrógenos/fisiología , Prosencéfalo/fisiología , Receptores Androgénicos/fisiología , Andrógenos/farmacología , Animales , Astrocitos/fisiología , Axones/fisiología , Corteza Cerebral/fisiología , Humanos , Neuronas/fisiología , Prosencéfalo/efectos de los fármacos , Ratas
18.
Neuroscience ; 138(2): 389-401, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16426763

RESUMEN

The expression of the human cyp19 gene, encoding P450 aromatase, the key enzyme for estrogen biosynthesis, involves alternative splicing of multiple forms of exon I regulated by different promoters. Aromatase expression has been detected in the human cerebral cortex, although the precise cellular distribution and promoter regulation are not fully characterized. We examined the variants of exon I of cyp19 by PCR analysis and the cellular distribution of the enzyme using immunohistochemistry in the human temporal cortex. We detected four different variants of exon I, suggesting a complex regulation of cyp19 in the cerebral cortex. In addition, the enzyme was localized mainly in a large subpopulation of pyramidal neurons and in a subpopulation of astrocytes. However, the majority of GABAergic interneurons identified by their expression of the calcium-binding proteins calbindin, calretinin and parvalbumin, did not display aromatase immunoreactivity. The broad range of potential modulators of the cyp19 gene in the cortex and the widespread expression of the protein in specific neuronal and glial subpopulations suggest that local estrogen formation may play an important role in human cortical function.


Asunto(s)
Aromatasa/genética , Lóbulo Temporal/enzimología , Adulto , Anciano , Autopsia , Secuencia de Bases , Cartilla de ADN , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , ARN/genética , ARN/aislamiento & purificación , Transcripción Genética
19.
Prog Neurobiol ; 63(1): 29-60, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11040417

RESUMEN

This review highlights recent evidence from clinical and basic science studies supporting a role for estrogen in neuroprotection. Accumulated clinical evidence suggests that estrogen exposure decreases the risk and delays the onset and progression of Alzheimer's disease and schizophrenia, and may also enhance recovery from traumatic neurological injury such as stroke. Recent basic science studies show that not only does exogenous estradiol decrease the response to various forms of insult, but the brain itself upregulates both estrogen synthesis and estrogen receptor expression at sites of injury. Thus, our view of the role of estrogen in neural function must be broadened to include not only its function in neuroendocrine regulation and reproductive behaviors, but also to include a direct protective role in response to degenerative disease or injury. Estrogen may play this protective role through several routes. Key among these are estrogen dependent alterations in cell survival, axonal sprouting, regenerative responses, enhanced synaptic transmission and enhanced neurogenesis. Some of the mechanisms underlying these effects are independent of the classically defined nuclear estrogen receptors and involve unidentified membrane receptors, direct modulation of neurotransmitter receptor function, or the known anti-oxidant activities of estrogen. Other neuroprotective effects of estrogen do depend on the classical nuclear estrogen receptor, through which estrogen alters expression of estrogen responsive genes that play a role in apoptosis, axonal regeneration, or general trophic support. Yet another possibility is that estrogen receptors in the membrane or cytoplasm alter phosphorylation cascades through direct interactions with protein kinases or that estrogen receptor signaling may converge with signaling by other trophic molecules to confer resistance to injury. Although there is clear evidence that estradiol exposure can be deleterious to some neuronal populations, the potential clinical benefits of estrogen treatment for enhancing cognitive function may outweigh the associated central and peripheral risks. Exciting and important avenues for future investigation into the protective effects of estrogen include the optimal ligand and doses that can be used clinically to confer benefit without undue risk, modulation of neurotrophin and neurotrophin receptor expression, interaction of estrogen with regulated cofactors and coactivators that couple estrogen receptors to basal transcriptional machinery, interactions of estrogen with other survival and regeneration promoting factors, potential estrogenic effects on neuronal replenishment, and modulation of phenotypic choices by neural stem cells.


Asunto(s)
Estradiol/farmacología , Fármacos Neuroprotectores/farmacología , Estradiol/fisiología , Humanos , Enfermedades del Sistema Nervioso/etiología , Fármacos Neuroprotectores/metabolismo , Receptores de Estrógenos/fisiología , Factores de Riesgo , Psicología del Esquizofrénico
20.
Prog Neurobiol ; 44(3): 279-307, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7886228

RESUMEN

It is now obvious that the CNS is capable of undergoing a variety of plastic changes at all stages of development. Although the magnitude and distribution of these changes may be more dramatic in the immature animal, the adult brain retains a remarkable capacity for undergoing morphological and functional modifications. Throughout development, as well as in the postpubertal animal, gonadal steroids exert an important influence over the architecture of specific sex steroid-responsive areas, resulting in sexual dimorphisms at both morphological and physiological levels. We are only now beginning to gain insight into the mechanisms involved in gonadal steroid-induced synaptic changes. The number of synaptic inputs to specific neuronal populations is sexually dimorphic and this can be modulated by changes in the sex steroid environment. These modifications can be correlated with other morphological changes, such as glial cell activation, that are occurring simultaneously in the same anatomical area. Indeed, the close physical relationship between glial cells and neuronal synaptic contacts makes them an ideal candidate for participating in this process. Interestingly, not only can the morphology and immunoreactivity of glial cells be modulated by gonadal steroids, but a close negative correlation between the number of synapses and the amount of glial ensheathing of a neuron has been demonstrated, suggesting an active participation of these cells in this process. Glia have sex steroid receptors, are capable of producing and metabolizing steroids, and can produce other neuronal trophic factors in response to sex steroids. Hence, their role in gonadal steroid-induced synaptic plasticity is becoming more apparent. In addition, there is recent evidence that this process may involve certain cell surface molecules, such as the N-CAMs, since a specific isoform of this molecule, previously referred to as the embryonic form, is found in those areas of the brain which maintain the capacity to undergo synaptic remodelling. However, there is much work to be done in order to fully understand this phenomenon and before bringing it into a clinical setting in hopes of treating neurodegenerative diseases or injuries to the nervous system.


Asunto(s)
Andrógenos/fisiología , Estrógenos/fisiología , Hipotálamo Medio/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Animales , Astrocitos/fisiología , Supervivencia Celular , Estradiol/farmacología , Femenino , Humanos , Neuronas/citología , Ratas , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA