Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Brain Behav Immun ; 77: 110-126, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30582962

RESUMO

Remyelination is an endogenous process by which functional recovery of damaged neurons is achieved by reinstating the myelin sheath around axons. Remyelination has been documented in multiple sclerosis (MS) lesions and experimental models, although it is often incomplete or fails to affect the integrity of the axon, thereby leading to progressive disability. Microglia play a crucial role in the clearance of the myelin debris produced by demyelination and in inflammation-dependent OPC activation, two processes necessary for remyelination to occur. We show here that following corpus callosum demyelination in the TMEV-IDD viral murine model of MS, there is spontaneous and partial remyelination that involves a temporal discordance between OPC mobilization and microglia activation. Pharmacological treatment with the endocannabinoid 2-AG enhances the clearance of myelin debris by microglia and OPC differentiation, resulting in complete remyelination and a thickening of the myelin sheath. These results highlight the importance of targeting microglia during the repair processes in order to enhance remyelination.


Assuntos
Ácidos Araquidônicos/farmacologia , Endocanabinoides/farmacologia , Glicerídeos/farmacologia , Microglia/efeitos dos fármacos , Remielinização/efeitos dos fármacos , Animais , Ácidos Araquidônicos/metabolismo , Axônios/metabolismo , Diferenciação Celular/fisiologia , Corpo Caloso/patologia , Corpo Caloso/fisiologia , Doenças Desmielinizantes/fisiopatologia , Modelos Animais de Doenças , Endocanabinoides/metabolismo , Feminino , Glicerídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Esclerose Múltipla/fisiopatologia , Bainha de Mielina/metabolismo , Células Precursoras de Oligodendrócitos/fisiologia , Oligodendroglia/metabolismo , Theilovirus/patogenicidade
2.
Neuroscience ; 191: 139-47, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21320576

RESUMO

Estradiol biosynthesis is catalyzed by the enzyme aromatase, the product of the CYP19A1 gene. Aromatase is expressed in the brain, where it is involved not only in the control of neuroendocrine events and reproduction, but also in the regulation of neural development, synaptic plasticity and cell survival. In this review we summarize the existing data related with the detection of aromatase in human brain, with particular emphasis in the so-called "non-primary reproductive" areas. Besides hypothalamus, amygdala and preoptic/septal areas, aromatase is expressed in certain regions of basal forebrain, cerebral cortex, hippocampus, thalamus, cerebellum and brainstem of the human brain. Aromatase in human brain is produced by neurons, but there is also an astrocyte subpopulation that constitutively expresses the enzyme. The use of different methodological approaches, including the in vivo analysis by positron emission tomography of human subjects, has permitted to draw a general map of human brain aromatase, but the detailed distribution map is still far to be completed. On the other hand, despite the fact that there is only one aromatase protein, there are multiple mRNA transcripts that differ in the 5'-untranslated region, where regulatory elements reside. To date, some of the aromatase transcripts characteristic of cerebral cortex, as well as of human cell lines of neural origin, have been identified. This characteristic may confer tissue or even region-specific regulation of the expression and therefore it is conceivable to develop selective aromatase modulators to regulate the expression of the enzyme in the human brain. This article is part of a Special Issue entitled: Neuroactive Steroids: Focus on Human Brain.


Assuntos
Aromatase/metabolismo , Encéfalo/metabolismo , Estradiol/metabolismo , Aromatase/genética , Encéfalo/anatomia & histologia , Humanos , RNA Mensageiro
3.
Neuroscience ; 138(2): 389-401, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16426763

RESUMO

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.


Assuntos
Aromatase/genética , Lobo Temporal/enzimologia , Adulto , Idoso , Autopsia , Sequência de Bases , Primers do DNA , Humanos , Masculino , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , RNA/genética , RNA/isolamento & purificação , Transcrição Gênica
4.
Neuroscience ; 138(3): 741-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16338087

RESUMO

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.


Assuntos
Encéfalo/fisiologia , Fosfoproteínas/fisiologia , Animais , Astrócitos/fisiologia , Regulação da Expressão Gênica , Glioma/genética , Humanos , Fosfoproteínas/genética , RNA Mensageiro/genética , Células de Schwann/fisiologia
5.
Neuroscience ; 138(3): 801-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16310968

RESUMO

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.


Assuntos
Androgênios/fisiologia , Prosencéfalo/fisiologia , Receptores Androgênicos/fisiologia , Androgênios/farmacologia , Animais , Astrócitos/fisiologia , Axônios/fisiologia , Córtex Cerebral/fisiologia , Humanos , Neurônios/fisiologia , Prosencéfalo/efeitos dos fármacos , Ratos
6.
J Neural Transm (Vienna) ; 112(1): 171-6, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15599615

RESUMO

Decreasing levels of gonadal steroids with aging are associated to an increase in cognitive, neurological and psychiatric disturbances. Estradiol is neuroprotective in animal models of neurodegeneration. However, the effects of hormonal replacement therapy on brain function in postmenopausal women are controversial. A possible alternative to hormonal replacement therapy is to increase local brain steroidogenesis, since experimental studies indicate that local estrogen formation in the brain is neuroprotective.


Assuntos
Encéfalo/metabolismo , Degeneração Neural/metabolismo , Degeneração Neural/prevenção & controle , Esteroides/biossíntese , Animais , Encéfalo/efeitos dos fármacos , Terapia de Reposição Hormonal/métodos , Humanos , Esteroides/farmacologia , Esteroides/uso terapêutico
7.
Rev Neurol ; 39(11): 1043-51, 2004.
Artigo em Espanhol | MEDLINE | ID: mdl-15597268

RESUMO

INTRODUCTION: The nervous system is a target for steroid hormones as well as a steroidogenic tissue, and it produces steroids that have a paracrine or autocrine effect on neurons and glial cells. Steroids formed in nervous tissue are called neurosteroids in order to differentiate them, in terms of their origin, from the peripheral steroids, although they both share the same molecular structure. DEVELOPMENT: We analyse the capacity of neurons and glial cells to synthesise steroids and describe the role played in steroidogenesis by certain key molecules, such as steroidogenic acute regulatory protein, peripheral benzodiazepine receptor and aromatase enzyme, which acts as a catalyst in the conversion of testosterone into estradiol. We also provide a description of the different mechanisms of action of the hormonal steroids and neurosteroids in the nervous system. These include both the regulation of protein synthesis by neurons and glial cells, by acting on nuclear receptors, and rapid effects mediated by membrane receptors or the allosteric modulation of neurotransmitter receptors. We review the clinical and experimental evidence for the neuroprotective effects of sex steroids and neurosteroids, and the limitations of hormone replacement therapy following menopause. CONCLUSIONS: Given the restraints involved in the systemic use of hormones as neuroprotective therapy, alternative strategies that take advantage of the neuroprotective properties of steroids must be sought. These could involve locally increasing their synthesis inside the brain or developing molecules that activate the steroid receptors in the nervous system and not in the peripheral organs.


Assuntos
Hormônios Esteroides Gonadais/metabolismo , Hormônios/metabolismo , Fenômenos Fisiológicos do Sistema Nervoso , Neuroglia , Neurônios , Fármacos Neuroprotetores/metabolismo , Animais , Hormônios Esteroides Gonadais/uso terapêutico , Hormônios/uso terapêutico , Humanos , Estrutura Molecular , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/fisiopatologia , Neuroglia/química , Neuroglia/metabolismo , Neurônios/química , Neurônios/metabolismo , Fármacos Neuroprotetores/uso terapêutico
8.
Rev. neurol. (Ed. impr.) ; 39(11): 1043-1051, 1 dic., 2004. ilus
Artigo em Es | IBECS | ID: ibc-37307

RESUMO

Introducción. El sistema nervioso es una diana de las hormonas esteroides y también un tejido esteroidogénico, y produce esteroides que actúan de una forma paracrina o autocrina sobre neuronas y glía. Los esteroides formados en el tejido nervioso se denominan neuroesteroides, para diferenciarlos, por su origen, de los esteroides periféricos, aunque tienen la misma estructura molecular que éstos. Desarrollo. Se analiza la capacidad de las neuronas y las células de glía de sintetizar esteroides, y se describe el papel de algunas moléculas clave en la esteroidogénesis, como la proteína de regulación aguda de esteroidogénesis, el receptor periférico de benzodiacepinas y la enzima aromatasa, que cataliza la conversión de testosterona en estradiol. También se describen los diversos mecanismos de acción de los esteroides hormonales y los neuroesteroides en el sistema nervioso. Éstos incluyen, tanto la regulación de la síntesis de proteínas por neuronas y glía, mediante acciones sobre receptores nucleares, como efectos rápidos mediados por receptores de membrana o la modulación alostérica de receptores para neurotransmisores. Se revisan las evidencias clínicas y experimentales de los efectos neuroprotectores de los esteroides sexuales y neuroesteroides y las limitaciones de la terapia hormonal sustitutiva, tras la menopausia. Conclusiones. Dada la limitación que plantea el uso sistémico de hormonas como terapia neuroprotectora, se necesitan encontrar estrategias alternativas que aprovechen las propiedades neuroprotectoras de los esteroides, tales como aumentar localmente su síntesis en el cerebro o desarrollar moléculas que activen a los receptores de esteroides en el sistema nervioso y no en los órganos periféricos (AU)


The nervous system is a target for steroid hormones as well as a steroidogenic tissue, and it produces steroids that have a paracrine or autocrine effect on neurons and glial cells. Steroids formed in nervous tissue are called neurosteroids in order to differentiate them, in terms of their origin, from the peripheral steroids, although they both share the same molecular structure. Development. We analyse the capacity of neurons and glial cells to synthesise steroids and describe the role played in steroidogenesis by certain key molecules, such as steroidogenic acute regulatory protein, peripheral benzodiazepine receptor and aromatase enzyme, which acts as a catalyst in the conversion of testosterone into estradiol. We also provide a description of the different mechanisms of action of the hormonal steroids and neurosteroids in the nervous system. These include both the regulation of protein synthesis by neurons and glial cells, by acting on nuclear receptors, and rapid effects mediated by membrane receptors or the allosteric modulation of neurotransmitter receptors. We review the clinical and experimental evidence for the neuroprotective effects of sex steroids and neurosteroids, and the limitations of hormone replacement therapy following menopause. Conclusions. Given the restraints involved in the systemic use of hormones as neuroprotective therapy, alternative strategies that take advantage of the neuroprotective properties of steroids must be sought. These could involve locally increasing their synthesis inside the brain or developing molecules that activate the steroid receptors in the nervous system and not in the peripheral organs (AU)


Assuntos
Animais , Axotomia , Sobrevivência Celular , Apoptose , Células Ganglionares da Retina , Nervo Óptico
9.
J Neuroendocrinol ; 16(1): 58-63, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14962077

RESUMO

The ovarian hormone progesterone is neuroprotective in some animal models of neurodegeneration. Progesterone actions in the brain may partly be mediated by the locally produced metabolites 5alpha-dihydroprogesterone and 3alpha,5alpha-tetrahydroprogesterone. The neuroprotective effects of these two metabolites of progesterone were assessed in this study. Ovariectomized Wistar rats were injected with kainic acid, to induce excitotoxic neuronal death in the hippocampus, and with different doses of 5alpha-dihydroprogesterone and 3alpha,5alpha-tetrahydroprogesterone. The number of surviving neurones in the hilus of the dentate gyrus of the hippocampus was assessed with the optical disector method. The administration of kainic acid resulted in a significant decrease in the number of hilar neurones and in the induction of vimentin expression in reactive astrocytes, a sign of neural damage. Low doses of 5alpha-dihydroprogesterone (0.25 and 0.5 mg/kg body weight, b.w.) prevented the loss of hilar neurones and the appearance of vimentin immunoreactivity in astrocytes. Higher doses (1-2 mg/kg b.w.) were not neuroprotective. By contrast, low doses of 3alpha,5alpha-tetrahydroprogesterone (0.25-1 mg/kg b.w.) were unable to protect the hilus from kainic acid while higher doses (2-4 mg/kg b.w.) were protective. The different optimal neuroprotective doses of 5alpha-dihydroprogesterone and 3alpha,5alpha-tetrahydroprogesterone suggest that these two steroids may protect neurones using different mechanisms. The neuroprotective effects of 3alpha,5alpha-tetrahydroprogesterone may be exerted by the inhibition of neuronal activity via the GABAA receptor. This latter possibility is supported by the observation that 3beta,5alpha-tetrahydroprogesterone, an isomer of 3alpha,5alpha-tetrahydroprogesterone that does not bind to GABAA receptor, was not neuroprotective. In summary, our findings suggest that progesterone neuroprotective effects may be, at least in part, mediated by its reduced metabolites 5alpha-dihydroprogesterone and 3alpha,5alpha-tetrahydroprogesterone.


Assuntos
Giro Denteado/fisiologia , Di-Hidrotestosterona/metabolismo , Fármacos Neuroprotetores/metabolismo , Pregnanolona/metabolismo , Animais , Morte Celular/fisiologia , Giro Denteado/efeitos dos fármacos , Giro Denteado/patologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Feminino , Ácido Caínico/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/patologia , Progesterona/metabolismo , Ratos , Ratos Wistar
10.
Prog Neurobiol ; 71(1): 57-66, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14611868

RESUMO

The process of aging deeply influences morphological and functional parameters of peripheral nerves. The observations summarized here indicate that the deterioration of myelin occurring in the peripheral nerves during aging may be explained by the fall of the levels of the major peripheral myelin proteins [e.g., glycoprotein Po (Po) and peripheral myelin protein 22 (PMP22)]. Neuroactive steroids, such as progesterone (PROG), dihydroprogesterone (5alpha-DH PROG), and tetrahydroprogesterone (3alpha,5alpha-TH PROG), are able to stimulate the low expression of these two myelin proteins present in the sciatic nerve of aged male rats. Since Po and PMP22 play an important physiological role in the maintenance of the multilamellar structure of PNS myelin, we have evaluated the effect of PROG and its neuroactive derivatives, 5alpha-DH PROG and 3alpha,5alpha-TH PROG, on the morphological alterations of myelinated fibers in the sciatic nerve of 22-24-month-old male rats. Data obtained clearly indicate that neuroactive steroids are able to reduce aging-associated morphological abnormalities of myelin and aging-associated myelin fiber loss in the sciatic nerve.


Assuntos
Envelhecimento , Bainha de Mielina/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/prevenção & controle , Progesterona/farmacologia , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Masculino , Proteína P0 da Mielina/efeitos dos fármacos , Proteína P0 da Mielina/fisiologia , Proteínas da Mielina/efeitos dos fármacos , Proteínas da Mielina/fisiologia , Doenças do Sistema Nervoso Periférico/patologia , Progesterona/análogos & derivados
11.
Homo ; 54(2): 132-41, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14740363

RESUMO

In elderly people, vascular alterations and degenerative alterations of the Central Nervous System (CNS) are two of the most common reasons for illness and death. Lipid pattern modifications and menopause in women are some of the causes for the appearance of these alterations. Vascular endothelium is in part responsible for vascular homeostasis, through the production of several vasoactive factors. Growth hormone (GH) exerts effects on the CNS and on the vascular endothelium, since GH deficient subjects exhibit endothelium-dependent alterations, which recover under substitutive GH treatment. Growth hormone has important actions on lipid metabolism that also play a role on vascular and endothelial function. Moreover, cardiac function improves when GH is associated to angiotensin II receptor blockers. Elderly people exhibit a physiological GH deficiency that could affect their vascular and cerebral functions. A study was carried out using old Wistar rats to clarify the effects of GH on the vessels under chronic "in vivo" conditions. The response to various vasoactive substances in aortic rings has been evaluated. An increase in the aortic media thickness was seen in old rats, which showed also a reduction in the vasodilator response to isoprenaline as compared to young animals. GH treatment partially restored the vasodilator response and reduced media thickness. Neuronal population was reduced in the hypocampus of old rats as compared to young ones and GH treatment was able to significantly enhance the number. Neurotransmitters were measured in several cerebral areas to establish differences between young and old GH-treated or untreated animals. Glutamine, Arginine and Aspartate were reduced in old animals whereas Citruline was increased. GH treatment restored in all cases the levels corresponding to young rats.


Assuntos
Envelhecimento/fisiologia , Hormônio do Crescimento Humano/farmacologia , Idoso , Animais , Feminino , Hipocampo/patologia , Hipocampo/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios , Neurotransmissores/análise , Ratos , Ratos Wistar
12.
Brain Res Brain Res Rev ; 37(1-3): 320-34, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11744097

RESUMO

Data from epidemiological studies suggest that the decline in estrogen following menopause could increase the risk of neurodegenerative diseases. Furthermore, experimental studies on different animal models have shown that estrogen is neuroprotective. The mechanisms involved in the neuroprotective effects of estrogen are still unclear. Anti-oxidant effects, activation of different membrane-associated intracellular signaling pathways, and activation of classical nuclear estrogen receptors (ERs) could contribute to neuroprotection. Interactions with neurotrophins and other growth factors may also be important for the neuroprotective effects of estradiol. In this review we focus on the interaction between insulin-like growth factor-I (IGF-I) and estrogen signaling in the brain and on the implications of this interaction for neuroprotection. During the development of the nervous system, IGF-I promotes the differentiation and survival of specific neuronal populations. In the adult brain, IGF-I is a neuromodulator, regulates synaptic plasticity, is involved in the response of neural tissue to injury and protects neurons against different neurodegenerative stimuli. As an endocrine signal, IGF-I represents a link between the growth and reproductive axes and the interaction between estradiol and IGF-I is of particular physiological relevance for the regulation of growth, sexual maturation and adult neuroendocrine function. There are several potential points of convergence between estradiol and IGF-I receptor (IGF-IR) signaling in the brain. Estrogen activates the mitogen-activated protein kinase (MAPK) pathway and has a synergistic effect with IGF-I on the activation of Akt, a kinase downstream of phosphoinositol-3 kinase. In addition, IGF-IR is necessary for the estradiol induced expression of the anti-apoptotic molecule Bcl-2 in hypothalamic neurons. The interaction of ERs and IGF-IR in the brain may depend on interactions between neural cells expressing ERs with neural cells expressing IGF-IR, or on direct interactions of the signaling pathways of alpha and beta ERs and IGF-IR in the same cell, since most neurons expressing IGF-IR also express at least one of the ER subtypes. In addition, studies on adult ovariectomized rats given intracerebroventricular (i.c.v.) infusions with antagonists for ERs or IGF-IR or with IGF-I have shown that there is a cross-regulation of the expression of ERs and IGF-IR in the brain. The interaction of estradiol and IGF-I and their receptors may be involved in different neural events. In the developing brain, ERs and IGF-IR are interdependent in the promotion of neuronal differentiation. In the adult, ERs and IGF-IR interact in the induction of synaptic plasticity. Furthermore, both in vitro and in vivo studies have shown that there is an interaction between ERs and IGF-IR in the promotion of neuronal survival and in the response of neural tissue to injury, suggesting that a parallel activation or co-activation of ERs and IGF-IR mediates neuroprotection.


Assuntos
Encéfalo/metabolismo , Citoproteção/fisiologia , Estrogênios/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Estrogênios/farmacologia , Feminino , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Ratos , Receptor IGF Tipo 1/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais/fisiologia
14.
J Neurobiol ; 47(4): 318-29, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11351342

RESUMO

The expression of aromatase, the enzyme that catalyzes the biosynthesis of estrogens from precursor androgens, is increased in the brain after injury, suggesting that aromatase may be involved in neuroprotection. In the present study, the effect of inactivating aromatase has been assessed in a model of neurodegeneration induced by the systemic administration of neurotoxins. Domoic acid, at a dose that is not neurotoxic in intact male mice, induced significant neuronal loss in the hilus of the hippocampal formation of mice with reduced levels of aromatase substrates as a result of gonadectomy. Furthermore, the aromatase substrate testosterone, as well as its metabolite estradiol, the product of aromatase, were able to protect hilar neurons from domoic acid. In contrast, dihydrotestosterone, the 5 alpha-reduced metabolite of testosterone and a nonaromatizable androgen, was not. These findings suggest that aromatization of testosterone to estradiol may be involved in the neuroprotective action of testosterone in this experimental model. In addition, aromatase knock-out mice showed significant neuronal loss after injection of a low dose of domoic acid, while control littermates did not, indicating that aromatase deficiency increases the vulnerability of hilar neurons to neurotoxic degeneration. The effect of aromatase on neuroprotection was also tested in male rats treated systemically with the specific aromatase inhibitor fadrozole and injected with kainic acid, a well characterized neurotoxin for hilar neurons in the rat. Fadrozole enhanced the neurodegenerative effect of kainic acid in intact male rats and this effect was counterbalanced by the administration of estradiol. Furthermore, the neuroprotective effect of testosterone against kainic acid in castrated male rats was blocked by fadrozole. These findings suggest that neuroprotection by aromatase is due to the formation of estradiol from its precursor testosterone. Finally, a role for local cerebral aromatase in neuroprotection is indicated by the fact that intracerebral administration of fadrozole enhanced kainic acid induced neurodegeneration in the hippocampus of intact male rats. These findings indicate that aromatase deficiency decreases the threshold for neurodegeneration and that local cerebral aromatase is neuroprotective. Brain aromatase may therefore represent a new target for therapeutic approaches to neurodegenerative diseases.


Assuntos
Aromatase/fisiologia , Encéfalo/enzimologia , Ácido Caínico/análogos & derivados , Fármacos Neuroprotetores/metabolismo , Animais , Aromatase/deficiência , Inibidores da Aromatase , Encéfalo/citologia , Morte Celular/efeitos dos fármacos , Di-Hidrotestosterona/farmacologia , Inibidores Enzimáticos/farmacologia , Estradiol/farmacologia , Fadrozol/farmacologia , Ácido Caínico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/patologia , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/farmacologia , Orquiectomia , Testosterona/farmacologia
15.
Prog Neurobiol ; 63(1): 29-60, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11040417

RESUMO

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.


Assuntos
Estradiol/farmacologia , Fármacos Neuroprotetores/farmacologia , Estradiol/fisiologia , Humanos , Doenças do Sistema Nervoso/etiologia , Fármacos Neuroprotetores/metabolismo , Receptores de Estrogênio/fisiologia , Fatores de Risco , Psicologia do Esquizofrênico
16.
J Pediatr Endocrinol Metab ; 13(8): 1045-66, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11085182

RESUMO

Gonadal steroid hormones have multiple effects throughout development on steroid responsive tissues in the brain. The belief that the cellular morphology of the adult brain cannot be modulated or that the synaptic connectivity is "hard-wired" is being rapidly refuted by abundant and growing evidence. Indeed, the brain is capable of undergoing many morphological changes throughout life and gonadal steroids play an important role in many of these processes. Gonadal steroids are implicated in the development of sexually dimorphic structures in the brain, in the control of physiological behaviors and functions and the brain's response to physiological or harmful substances. The effect of sex steroids on neuroprotection and neuroregeneration is an important and expanding area of investigation. Astroglia are targets for estrogen and testosterone and are apparently involved in the actions of sex steroids on the central nervous system. Sex hormones induce changes in the expression of glial fibrillary acidic protein, the growth of astrocytic processes and the extent to which neuronal membranes are covered by astroglial processes. These changes are linked to modifications in the number of synaptic inputs to neurons and suggest that astrocytes may participate in the genesis of gonadal steroid-induced sex differences in synaptic connectivity and synaptic plasticity in the adult brain. Astrocytes and tanycytes may also participate in the cellular effects of sex steroids by releasing neuroactive substances and by regulating the local accumulation of specific growth factors, such as insulin-like growth factor-I, that are involved in estrogen-induced synaptic plasticity and estrogen-mediated neuroendocrine control. Astroglia may also be involved in the regenerative and neuroprotective effects of sex steroids since astroglial activation after brain injury or after peripheral nerve axotomy is regulated by sex hormones.


Assuntos
Encéfalo/fisiologia , Hormônios Esteroides Gonadais/fisiologia , Animais , Substâncias de Crescimento/fisiologia , Neuroglia/fisiologia , Plasticidade Neuronal/fisiologia , Fármacos Neuroprotetores/metabolismo , Cicatrização/fisiologia
17.
J Neurocytol ; 29(5-6): 425-37, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11424959

RESUMO

Several in vitro and in vivo studies have shown that estrogen has neuroprotective properties. The neuroprotective effects of estrogen are probably exerted through several mechanisms. It is established that estrogen can provide neuroprotection by actions that are independent of estrogen receptor activation. In addition, in several experimental models, activation of estrogen receptors appears to be indispensable for neuroprotection. This review focuses on neuroprotection mediated by estrogen receptors. The interaction of estrogen with growth factor receptor signaling to induce neuroprotection is discussed. Evidence is presented that estrogen receptors and insulin-like growth factor-1 receptors interact in the promotion of neuronal survival and neuroprotection.


Assuntos
Neurônios/citologia , Neurônios/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Sobrevivência Celular/fisiologia , Fármacos Neuroprotetores/metabolismo
18.
J Neurosci Res ; 58(6): 815-22, 1999 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-10583912

RESUMO

We have previously shown that 17-beta-estradiol protects neurons in the dentate gyrus from kainic acid-induced death in vivo. To analyse whether this effect is mediated through estrogen receptors and through cross-talk between steroid and insulin-like growth factor (IGF) systems, we have concomitantly administered antagonists of estrogen receptor (ICI 182,780) or the IGF-I receptor (JB1) with estradiol. In addition, we have also administered IGF-I with or without the estrogen receptor antagonist. JB1 (20 microg/ml), ICI 182,780 (10(-7) M), and IGF-I (100 microg/ml) were delivered into the left lateral ventricle of young ovariectomized rats via an Alzet osmotic minipump (0.5 microl/hr) for 2 weeks. All rats received kainic acid (7 mg/Kg b.w.) or vehicle i.p. injections at day 7 after minipump implant. Also on day 7, rats treated i.c. v.with only ICI 182,780 or JB1 received a single i.p. injection of 17-beta-estradiol (150 microg/rat) or vehicle. On day 14 after minipump implant, the rats were killed, brains processed, and the number of surviving hilar neurons estimated by the optical disector technique. Both IGF-I and estradiol treatments resulted in over 90% survival of hilar neurons. The neuroprotective action of estradiol was blocked by ICI 182,780 and by JB1. Furthermore, IGF-I enhancement of neuronal survival was significantly reduced by ICI 182,780. These results indicate that in this model of hippocampal lesion, the neuroprotective effect of estradiol depends both on estrogen receptors and IGF-I receptors, while the protection exerted by IGF-I depends also on estrogen receptors. In conclusion, an interaction of estrogen receptor and IGF-I receptor signalling may mediate neuroprotection in the adult rat hippocampus.


Assuntos
Estradiol/farmacologia , Estradiol/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/fisiologia , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Animais , Contagem de Células , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Fulvestranto , Hipocampo/citologia , Ácido Caínico/farmacologia , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Somatostatina/análise
19.
J Neurobiol ; 40(4): 574-84, 1999 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10453057

RESUMO

Astroglia are targets for estrogen and testosterone and are apparently involved in the action of sex steroids on the brain. Sex hormones induce changes in the expression of glial fibrillary acidic protein, the growth of astrocytic processes, and the degree of apposition of astroglial processes to neuronal membranes in the rat hypothalamus. These changes are linked to modifications in the number of synaptic inputs to hypothalamic neurons. These findings suggest that astrocytes may participate in the genesis of androgen-induced sex differences in synaptic connectivity and in estrogen-induced synaptic plasticity in the adult brain. Astrocytes and tanycytes may also participate in the cellular effects of sex steroids by releasing neuroactive substances and by regulating the local accumulation of specific growth factors, such as insulin-like growth factor-I, that are involved in estrogen-induced synaptic plasticity and estrogen-mediated neuroendocrine control. Astroglia may also be involved in regenerative and neuroprotective effects of sex steroids, since astroglia formation after brain injury or after peripheral nerve axotomy is regulated by sex hormones. Furthermore, the expression of aromatase, the enzyme that produces estrogen, is induced de novo in astrocytes in lesioned brain areas of adult male and female rodents. Since astroglia do not express aromatase under normal circumstances, the induction of this enzyme may be part of the program of glial activation to cope with the new conditions of the neural tissue after injury. Given the neuroprotective and growth-promoting effects of estrogen after injury, the local production of this steroid may be a relevant component of the reparative process.


Assuntos
Astrócitos/fisiologia , Encéfalo/fisiologia , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Animais , Encéfalo/citologia , Hormônios Esteroides Gonadais/fisiologia , Humanos
20.
Glia ; 26(3): 260-7, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10340766

RESUMO

Estrogen receptors are direct regulators of transcription that function by binding to specific DNA sequences in promoters of target genes. The two cloned forms of estrogen receptors, alpha and beta, are expressed in the central nervous system by different neuronal populations. Astrocytes in vitro are also reported to express estrogen receptor alpha; however, this expression has not been confirmed in the rat brain in vivo. The apparent absence of estrogen receptors in glia in vivo contrasts with the well-known effects of this hormone on astrocytes of different brain areas, including the hippocampal formation. In this study, the expression of estrogen receptors in the hippocampal formation of adult male rats has been assessed by confocal microscopy. Estrogen receptor alpha-immunoreactivity was localized in neuronal nuclei in the pyramidal cell layer of CA1-CA3 fields. Estrogen receptor beta-immunoreactivity was observed in the perikarya, apical dendrites, and cell nuclei of pyramidal neurons in CA1 and CA2. Furthermore, estrogen receptor beta-immunoreactive glia were observed in CA1, CA2, CA3, and in the hilus of the dentate gyrus of male and female rats. Estrogen receptor beta-immunoreactivity was localized in glial processes and perikarya and, in some cases, in glial cell nuclei. Double immunocytochemical labeling of estrogen receptor beta and the specific astroglial marker glial fibrillary acidic protein revealed that estrogen receptor beta-immunoreactive glial cells were astrocytes. Estrogen receptor alpha was not co-localized with glial fibrillary acidic protein. The presence of estrogen receptor beta in astrocytes of adult male and female rats demonstrates a possible mechanism by which estrogen can directly modulate gene expression in these cells.


Assuntos
Astrócitos/metabolismo , Encéfalo/citologia , Receptores de Estrogênio/metabolismo , Animais , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Masculino , Microscopia Confocal , Células Piramidais/metabolismo , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...