Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Front Neuroendocrinol ; 71: 101102, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37689249

RESUMO

The brain synthesizes a variety of neurosteroids, including neuroestradiol. Inhibition of neuroestradiol synthesis results in alterations in basic neurodevelopmental processes, such as neurogenesis, neuroblast migration, neuritogenesis and synaptogenesis. Although the neurodevelopmental actions of neuroestradiol are exerted in both sexes, some of them are sex-specific, such as the well characterized effects of neuroestradiol derived from the metabolism of testicular testosterone during critical periods of male brain development. In addition, recent findings have shown sex-specific actions of neuroestradiol on neuroblast migration, neuritic growth and synaptogenesis in females. Among other factors, the epigenetic regulation exerted by X linked genes, such as Kdm6a/Utx, may determine sex-specific actions of neuroestradiol in the female brain. This review evidences the impact of neuroestradiol on brain formation in both sexes and highlights the interaction of neural steriodogenesis, hormones and sex chromosomes in sex-specific brain development.


Assuntos
Epigênese Genética , Neuroesteroides , Feminino , Masculino , Humanos , Neurônios/metabolismo , Neuroesteroides/metabolismo , Testosterona/metabolismo
2.
Neurobiol Dis ; 159: 105495, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34478848

RESUMO

Long-term high-fat diet (HFD) consumption commonly leads to obesity, a major health concern of western societies and a risk factor for Alzheimer's disease (AD). Both conditions present glial activation and inflammation and show sex differences in their incidence, clinical manifestation, and disease course. HFD intake has an important impact on gut microbiota, the bacteria present in the gut, and microbiota dysbiosis is associated with inflammation and certain mental disorders such as anxiety. In this study, we have analyzed the effects of a prolonged (18 weeks, starting at 7 months of age) HFD on male and female mice, both wild type (WT) and TgAPP mice, a model for AD, investigating the behavioral profile, gut microbiota composition and inflammatory/phagocytosis-related gene expression in hippocampus. In the open-field test, no overt differences in motor activity were observed between male and female or WT and TgAPP mice on a low-fat diet (LFD). However, HFD induced anxiety, as judged by decreased motor activity and increased time in the margins in the open-field, and a trend towards increased immobility time in the tail suspension test, with increased defecation. Intriguingly, female TgAPP mice on HFD showed less immobility and defecation compared to female WT mice on HFD. HFD induced dysbiosis of gut microbiota, resulting in reduced microbiota diversity and abundance compared with LFD fed mice, with some significant differences due to sex and little effect of genotype. Gene expression of pro-inflammatory/phagocytic markers in the hippocampus were not different between male and female WT mice, and in TgAPP mice of both sexes, some cytokines (IL-6 and IFNγ) were higher than in WT mice on LFD, more so in female TgAPP (IL-6). HFD induced few alterations in mRNA expression of inflammatory/phagocytosis-related genes in male mice, whether WT (IL-1ß, MHCII), or TgAPP (IL-6). However, in female TgAPP, altered gene expression returned towards control levels following prolonged HFD (IL-6, IL-12ß, TNFα, CD36, IRAK4, PYRY6). In summary, we demonstrate that HFD induces anxiogenic symptoms, marked alterations in gut microbiota, and increased expression of inflammatory genes, except for female TgAPP that appear to be resistant to the diet effects. Lifestyle interventions should be introduced to prevent AD onset or exacerbation by reducing inflammation and its associated symptoms; however, our results suggest that the eventual goal of developing prevention and treatment strategies should take sex into consideration.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Comportamento Animal/fisiologia , Dieta Hiperlipídica , Disbiose/genética , Microbioma Gastrointestinal/fisiologia , Inflamação/genética , Estresse Psicológico/genética , Doença de Alzheimer/fisiopatologia , Animais , Modelos Animais de Doenças , Disbiose/fisiopatologia , Feminino , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Interleucina-1beta/genética , Interleucina-6/genética , Masculino , Camundongos , Camundongos Transgênicos , Fagocitose/genética , RNA Mensageiro/metabolismo , Caracteres Sexuais , Estresse Psicológico/fisiopatologia
3.
Front Neuroendocrinol ; 55: 100787, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31513774

RESUMO

Estradiol, either from peripheral or central origin, activates multiple molecular neuroprotective and neuroreparative responses that, being mediated by estrogen receptors or by estrogen receptor independent mechanisms, are initiated at the membrane, the cytoplasm or the cell nucleus of neural cells. Estrogen-dependent signaling regulates a variety of cellular events, such as intracellular Ca2+ levels, mitochondrial respiratory capacity, ATP production, mitochondrial membrane potential, autophagy and apoptosis. In turn, these molecular and cellular actions of estradiol are integrated by neurons and non-neuronal cells to generate different tissue protective responses, decreasing blood-brain barrier permeability, oxidative stress, neuroinflammation and excitotoxicity and promoting synaptic plasticity, axonal growth, neurogenesis, remyelination and neuroregeneration. Recent findings indicate that the neuroprotective and neuroreparative actions of estradiol are different in males and females and further research is necessary to fully elucidate the causes for this sex difference.


Assuntos
Apoptose/fisiologia , Autofagia/fisiologia , Estradiol/metabolismo , Estradiol/farmacologia , Inflamação/metabolismo , Neurogênese/fisiologia , Plasticidade Neuronal/fisiologia , Neuroproteção/fisiologia , Receptores de Estrogênio/metabolismo , Caracteres Sexuais , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Feminino , Humanos , Masculino , Neurogênese/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
4.
Eur J Neurosci ; 52(1): 2756-2770, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32243028

RESUMO

Including sex is of paramount importance in preclinical and clinical stroke researches, and molecular studies dealing in depth with sex differences in stroke pathophysiology are needed. To gain insight into the molecular sex dimorphism of ischaemic stroke in rat cerebral cortex, male and female adult rats were subjected to transient middle cerebral artery occlusion. The expression of neuroglobin (Ngb) and other functionally related molecules involved in sex steroid signalling (oestrogen and androgen receptors), steroidogenesis (StAR, TSPO and aromatase) and autophagic activity (LC3B-II/LC3B-I ratio, UCP2 and HIF-1α) was assessed in the ipsilateral ischaemic and contralateral non-ischaemic hemispheres. An increased expression of Ngb was detected in the injured female cerebral cortex. In contrast, increased expression of oestrogen receptor α, GPER, StAR, TSPO and UCP2, and decreased androgen receptor expression were detected in the injured male cortex. In both sexes, the ischaemic insult induced an upregulation of LC3B-II/-I ratio, indicative of increased autophagy. Therefore, the cerebral cortex activates both sex-specific and common molecular responses with neuroprotective potential after ischaemia-reperfusion, which globally results in similar stroke outcome in both sexes. Nonetheless, these different potential molecular targets should be taken into account when neuroprotective drugs aiming to reduce brain damage in ischaemic stroke are investigated.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Animais , Autofagia , Córtex Cerebral , Modelos Animais de Doenças , Feminino , Infarto da Artéria Cerebral Média , Masculino , Neuroglobina , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Esteroides
5.
Nat Rev Neurosci ; 16(1): 17-29, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25423896

RESUMO

Hormones regulate homeostasis by communicating through the bloodstream to the body's organs, including the brain. As homeostatic regulators of brain function, some hormones exert neuroprotective actions. This is the case for the ovarian hormone 17ß-oestradiol, which signals through oestrogen receptors (ERs) that are widely distributed in the male and female brain. Recent discoveries have shown that oestradiol is not only a reproductive hormone but also a brain-derived neuroprotective factor in males and females and that ERs coordinate multiple signalling mechanisms that protect the brain from neurodegenerative diseases, affective disorders and cognitive decline.


Assuntos
Encéfalo/metabolismo , Estradiol , Neuroprostanos , Receptores de Estrogênio/fisiologia , Animais , Encéfalo/efeitos dos fármacos , Encefalopatias/tratamento farmacológico , Encefalopatias/metabolismo , Estradiol/metabolismo , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Humanos , Masculino , Neuroprostanos/metabolismo , Neuroprostanos/farmacologia , Neuroprostanos/uso terapêutico
6.
Glia ; 66(3): 522-537, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29139169

RESUMO

Sex differences in the incidence, clinical manifestation, disease course, and prognosis of neurological diseases, such as autism spectrum disorders or Alzheimer's disease, have been reported. Obesity has been postulated as a risk factor for cognitive decline and Alzheimer's disease and, during pregnancy, increases the risk of autism spectrum disorders in the offspring. Obesity is associated with increased serum and brain levels of free fatty acids, such as palmitic acid, which activate microglial cells triggering a potent inflammatory cascade. In this study, we have determined the effect of palmitic acid in the inflammatory profile, motility, and phagocytosis of primary male and female microglia, both in basal conditions and in the presence of a pro-inflammatory stimulus (interferon-γ). Male microglia in vitro showed higher migration than female microglia under basal and stimulated conditions. In contrast, female microglia had higher basal and stimulated phagocytic activity than male microglia. Palmitic acid did not affect basal migration or phagocytosis, but abolished the migration and phagocytic activity of male and female microglia in response to interferon-γ. These findings extend previous observations of sex differences in microglia and suggest that palmitic acid impairs the protective responses of these cells.


Assuntos
Movimento Celular/fisiologia , Microglia/metabolismo , Ácido Palmítico/toxicidade , Fagocitose/fisiologia , Caracteres Sexuais , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Inflamação/metabolismo , Inflamação/patologia , Interferon gama/administração & dosagem , Interferon gama/metabolismo , Masculino , Microglia/efeitos dos fármacos , Microglia/patologia , Fagocitose/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo , Prosencéfalo/patologia , RNA Mensageiro/metabolismo , Ratos Wistar
7.
J Neurogenet ; 31(4): 300-306, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29078716

RESUMO

Female mouse hippocampal and hypothalamic neurons growing in vitro show a faster development of neurites than male mouse neurons. This sex difference in neuritogenesis is determined by higher expression levels of the neuritogenic factor neurogenin 3 in female neurons. Experiments with the four core genotype mouse model, in which XX and XY animals with male gonads and XX and XY animals with female gonads are generated, indicate that higher levels of neurogenin 3 in developing neurons are determined by the presence of the XX chromosome complement. Female XX neurons express higher levels of estrogen receptors than male XY neurons. In female XX neurons, neuronal derived estradiol increases neurogenin 3 expression and neuritogenesis. In contrast, neuronal-derived estradiol is not able to upregulate neurogenin 3 in male XY neurons, resulting in decreased neuritogenesis compared to female neurons. However, exogenous testosterone increases neurogenin 3 expression and neuritogenesis in male XY neurons. These findings suggest that sex differences in neuronal development are determined by the interaction of sex chromosomes, neuronal derived estradiol and gonadal hormones.


Assuntos
Estradiol/biossíntese , Neurônios/metabolismo , Cromossomos Sexuais/fisiologia , Diferenciação Sexual/fisiologia , Animais , Feminino , Masculino , Camundongos , Neuritos/metabolismo , Neurogênese/fisiologia
8.
Cereb Cortex ; 25(1): 35-45, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23960212

RESUMO

Traumatic brain injury (TBI) and its consequences represent one of the leading causes of death in young adults. This lesion mediates glial activation and the release of harmful molecules and causes brain edema, axonal injury, and functional impairment. Since glial activation plays a key role in the development of this damage, it seems that controlling it could be beneficial and could lead to neuroprotective effects. Recent studies show that minocycline suppresses microglial activation, reduces the lesion volume, and decreases TBI-induced locomotor hyperactivity up to 3 months. The endocannabinoid system (ECS) plays an important role in reparative mechanisms and inflammation under pathological situations by controlling some mechanisms that are shared with minocycline pathways. We hypothesized that the ECS could be involved in the neuroprotective effects of minocycline. To address this hypothesis, we used a murine TBI model in combination with selective CB1 and CB2 receptor antagonists (AM251 and AM630, respectively). The results provided the first evidence for the involvement of ECS in the neuroprotective action of minocycline on brain edema, neurological impairment, diffuse axonal injury, and microglial activation, since all these effects were prevented by the CB1 and CB2 receptor antagonists.


Assuntos
Lesões Encefálicas/metabolismo , Encéfalo/efeitos dos fármacos , Microglia/efeitos dos fármacos , Minociclina/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Edema Encefálico/metabolismo , Lesões Encefálicas/patologia , Antagonistas de Receptores de Canabinoides/farmacologia , Indóis/farmacologia , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/antagonistas & inibidores
9.
Glia ; 63(11): 1966-1981, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26037411

RESUMO

Several brain disorders associated with neuroinflammation show sex differences in their incidence, onset, progression and/or outcome. The different regulation of the neuroinflammatory response in males and females could underlie these sex differences. In this study, we have explored whether reactive gliosis after a penetrating cortical injury exhibits sex differences. Males presented a higher density of Iba1 immunoreactive cells in the proximity of the wound (0-220 µm) than females. This sex difference was due to a higher number of Iba1 immunoreactive cells with nonreactive morphology. In addition microglia/macrophages in that region expressed arginase-1, marker of alternatively activated microglia, and the neuroprotective protein Neuroglobin, in a greater proportion in males than in females. No sex differences were found in the number of astrocytes around the lesion. However, the percentage of astrocytes expressing chemokine (C-C motif) ligand 2 (CCL2), involved in recruitment of immune cells and gliosis regulation, was higher in males. Males also presented a significantly higher density of neurons in the lesion edge than females. These findings indicate that male and female mice have different neuroinflammatory responses after a cortical stab wound injury and suggest that sex differences in reactive gliosis may contribute to sex differences in neuroinflammatory diseases. GLIA 2015;63:1966-1981.

10.
Front Neuroendocrinol ; 35(4): 459-72, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24704390

RESUMO

This review focuses on the neuroendocrine mechanisms in the mother and the newborn that are involved in the generation and consolidation of mother-child attachment. The role that different hormones and neurotransmitters play on the regulation of these mechanisms during parturition, the immediate postpartum period and lactation is discussed. Interferences in the initiation of mother-child attachment may have potential long-term effects for the behavior and affection of the newborn. Therefore, the possible consequences of alterations in the physiological neuroendocrine mechanisms of attachment, caused by elective Cesarean section, intrapartum hormonal manipulations, preterm delivery, mother-infant postpartum separation and bottle-feeding instead of breastfeeding are also discussed.


Assuntos
Aleitamento Materno , Sistema Endócrino , Relações Mãe-Filho , Doenças do Sistema Nervoso/etiologia , Animais , Aleitamento Materno/psicologia , Humanos , Período Pós-Parto/fisiologia
11.
Rev Endocr Metab Disord ; 16(3): 177-98, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26296373

RESUMO

Steroids are important physiological orchestrators of endocrine as well as peripheral and central nervous system functions. One of the key processes for regulation of these molecules lies in their enzymatic processing by a family of 5α-reductase (5α-Rs) isozymes. By catalyzing a key rate-limiting step in steroidogenesis, this family of enzymes exerts a crucial role not only in the physiological control but also in pathological events. Indeed, both 5α-R inhibition and supplementation of 5α-reduced metabolites are currently used or have been proposed as therapeutic strategies for a wide array of pathological conditions. In particular, the potent 5α-R inhibitors finasteride and dutasteride are used in the treatments of benign prostatic hyperplasia (BPH), as well as in male pattern hair loss (MPHL) known as androgenetic alopecia (AGA). Recent preclinical and clinical findings indicate that 5α-R inhibitors evoke not only beneficial, but also adverse effects. Future studies should investigate the biochemical and physiological mechanisms that underlie the persistence of the adverse sexual side effects to determine why a subset of patients is afflicted with such persistence or irreversible adverse effects. Also a better focus of clinical research is urgently needed to better define those subjects who are likely to be adversely affected by such agents. Furthermore, research on the non-sexual adverse effects such as diabetes, psychosis, depression, and cognitive function are needed to better understand the broad spectrum of the effects these drugs may elicit during their use in treatment of AGA or BPH. In this review, we will summarize the state of art on this topic, overview the key unresolved questions that have emerged on the pharmacological targeting of these enzymes and their products, and highlight the need for further studies to ascertain the severity and duration of the adverse effects of 5α-R inhibitors, as well as their biological underpinnings.


Assuntos
Inibidores de 5-alfa Redutase/efeitos adversos , Inibidores de 5-alfa Redutase/uso terapêutico , Alopecia/tratamento farmacológico , Animais , Sistema Nervoso Central/fisiologia , Colestenona 5 alfa-Redutase/deficiência , Colestenona 5 alfa-Redutase/fisiologia , Transtornos Cognitivos/induzido quimicamente , Depressão/induzido quimicamente , Diabetes Mellitus/induzido quimicamente , Dutasterida/efeitos adversos , Dutasterida/uso terapêutico , Finasterida/efeitos adversos , Finasterida/uso terapêutico , Humanos , Masculino , Hiperplasia Prostática/tratamento farmacológico , Neoplasias da Próstata/prevenção & controle , Psicoses Induzidas por Substâncias , Disfunções Sexuais Fisiológicas/induzido quimicamente , Esteroides/biossíntese
12.
Horm Behav ; 74: 19-27, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25921586

RESUMO

This article is part of a Special Issue "Estradiol and Cognition". Estradiol participates in the regulation of the function and plasticity of synaptic circuits in key cognitive brain regions, such as the prefrontal cortex and the hippocampus. The mechanisms elicited by estradiol are mediated by the regulation of transcriptional activity by nuclear estrogen receptors and by intracellular signaling cascades activated by estrogen receptors associated with the plasma membrane. In addition, the mechanisms include the interaction of estradiol with the signaling of other factors involved in the regulation of cognition, such as brain derived neurotrophic factor, insulin-like growth factor-1 and Wnt. Modifications in these signaling pathways by aging or by a long-lasting ovarian hormone deprivation after menopause may impair the enhancing effects of estradiol on synaptic plasticity and cognition.


Assuntos
Estradiol/farmacologia , Memória/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Masculino , Plasticidade Neuronal/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
13.
J Neurosci ; 33(20): 8891-7, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23678130

RESUMO

Ligands of the translocator protein (TSPO) elicit pleiotropic neuroprotective effects that represent emerging treatment strategies for several neurodegenerative conditions. To investigate the potential of TSPO as a therapeutic target for Alzheimer's disease (AD), the current study assessed the effects of the TSPO ligand Ro5-4864 on the development of neuropathology in 3xTgAD mice. The effects of the TSPO ligand on neurosteroidogenesis and AD-related neuropathology, including ß-amyloid accumulation, gliosis, and behavioral impairment, were examined under both early intervention (7-month-old young-adult male mice with low pathology) and treatment (24-month-old, aged male mice with advanced neuropathology) conditions. Ro5-4864 treatment not only effectively attenuated development of neuropathology and behavioral impairment in young-adult mice but also reversed these indices in aged 3xTgAD mice. Reduced levels of soluble ß-amyloid were also observed by the combination of TSPO ligands Ro5-4864 and PK11195 in nontransgenic mice. These findings suggest that TSPO is a promising target for the development of pleiotropic treatment strategies for the management of AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Receptores de GABA/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Benzodiazepinonas/uso terapêutico , Modelos Animais de Doenças , Humanos , Isoquinolinas/uso terapêutico , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Orquiectomia , Presenilina-1/genética , Esteroides/metabolismo , Proteínas tau/genética
14.
Toxicol Appl Pharmacol ; 272(2): 263-71, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23891859

RESUMO

Dimethoate is an organophosphorus insecticide extensively used in horticulture. Previous studies have shown that the administration of dimethoate to male rats, at a very low dose and during a sub-chronic period, increases the oxidation of lipids and proteins, reduces the levels of antioxidants and impairs mitochondrial function in various brain regions. In this study, we have assessed in C57Bl/6 adult male mice, whether sub-chronic (5weeks) intoxication with a low dose of dimethoate (1.4mg/kg) affects the expression of inflammatory molecules and the reactivity of microglia in the hippocampus and striatum under basal conditions and after an immune challenge caused by the systemic administration of lipopolysaccharide. Dimethoate increased mRNA levels of tumor necrosis factor α (TNFα) and interleukin (IL) 6 in the hippocampus, and increased the proportion of Iba1 immunoreactive cells with reactive phenotype in dentate gyrus and striatum. Lipopolysaccharide caused a significant increase in the mRNA levels of IL1ß, TNFα, IL6 and interferon-γ-inducible protein 10, and a significant increase in the proportion of microglia with reactive phenotype in the hippocampus and the striatum. Some of the effects of lipopolysaccharide (proportion of Iba1 immunoreactive cells with reactive phenotype and IL6 mRNA levels) were amplified in the animals treated with dimethoate, but only in the striatum. These findings indicate that a sub-chronic period of administration of a low dose of dimethoate, comparable to the levels of the pesticide present as residues in food, causes a proinflammatory status in the brain and enhances the neuroinflammatory response to the lipopolysaccharide challenge with regional specificity.


Assuntos
Corpo Estriado/efeitos dos fármacos , Dimetoato/toxicidade , Hipocampo/efeitos dos fármacos , Inflamação/induzido quimicamente , Inseticidas/toxicidade , Lipopolissacarídeos/farmacologia , Animais , Corpo Estriado/imunologia , Corpo Estriado/patologia , Hipocampo/imunologia , Hipocampo/patologia , Imuno-Histoquímica , Inflamação/imunologia , Inflamação/patologia , Interleucinas/biossíntese , Interleucinas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Testes de Toxicidade Subcrônica
15.
Brain Behav Immun ; 28: 196-206, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23207108

RESUMO

Early life experiences, such as prenatal stress, may result in permanent alterations in the function of the nervous and immune systems. In this study we have assessed whether prenatal stress affects the inflammatory response of the hippocampal formation of male mice to an inflammatory challenge during adulthood. Pregnant C57BL/6 mice were randomly assigned to stress (n=10) or non-stress (n=10) groups. Animals of the stress group were placed in plastic transparent cylinders and exposed to bright light for 3 sessions of 45min every day from gestational day 12 to parturition. Non-stressed pregnant mice were left undisturbed. At four months of age, non stressed and prenatally stressed male offspring were killed, 24h after the systemic administration of lipopolysaccharide (LPS) or vehicle. Under basal conditions, prenatally stressed animals showed increased expression of interleukin 1ß and tumor necrosis factor-α (TNF-α) in the hippocampus and an increased percentage of microglia cells with reactive morphology in CA1 compared to non-stressed males. Furthermore, prenatally stressed mice showed increased TNF-α immunoreactivity in CA1 and increased number of Iba-1 immunoreactive microglia and GFAP-immunoreactive astrocytes in the dentate gyrus after LPS administration. In contrast, LPS did not induce such changes in non-stressed animals. These findings indicate that prenatal stress induces a basal proinflammatory status in the hippocampal formation during adulthood that results in an enhanced activation of microglia and astrocytes in response to a proinflammatory insult.


Assuntos
Citocinas/fisiologia , Hipocampo/efeitos dos fármacos , Inflamação/induzido quimicamente , Lipopolissacarídeos/farmacologia , Estresse Psicológico/fisiopatologia , Animais , Corticosterona/sangue , Feminino , Hipocampo/fisiologia , Inflamação/imunologia , Inflamação/fisiopatologia , Interleucina-1beta/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real , Estresse Psicológico/imunologia , Fator de Necrose Tumoral alfa/fisiologia
16.
Horm Behav ; 63(2): 216-21, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22401743

RESUMO

Astrocytes and microglia respond to central nervous system (CNS) injury with changes in morphology, proliferation, migration and expression of inflammatory regulators. This phenomenon is known as reactive gliosis. Activation of astrocytes and microglia after acute neural insults, such as stroke or traumatic CNS injury, is considered to be an adaptive response that contributes to minimize neuronal damage. However, reactive gliosis may amplify CNS damage under chronic neurodegenerative conditions. Progesterone, estradiol and testosterone have been shown to control reactive gliosis in different models of CNS injury, modifying the number of reactive astrocytes and reactive microglia and the expression of anti-inflammatory and proinflammatory mediators. The actions of gonadal hormones on reactive gliosis involve different mechanisms, including the modulation of the activity of steroid receptors, such as estrogen receptors α and ß, the regulation of nuclear factor-κB mediated transcription of inflammatory molecules and the recruitment of the transcriptional corepressor c-terminal binding protein to proinflammatory promoters. In addition, the Parkinson's disease related gene parkin and the endocannabinoid system also participate in the regulation of reactive gliosis by estradiol. The control exerted by gonadal hormones on reactive gliosis may affect the response of neural tissue to trauma and neurodegeneration and may contribute to sex differences in the manifestation of neurodegenerative diseases. However, the precise functional consequences of the regulation of reactive gliosis by gonadal hormones under acute and chronic neurodegenerative conditions are still not fully clarified.


Assuntos
Lesões Encefálicas/complicações , Gliose/etiologia , Gliose/prevenção & controle , Hormônios Gonadais/fisiologia , Animais , Lesões Encefálicas/fisiopatologia , Gliose/fisiopatologia , Hormônios Gonadais/metabolismo , Hormônios Gonadais/farmacologia , Humanos , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
J Lipid Res ; 53(2): 300-10, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22158827

RESUMO

Diabetic peripheral neuropathy (DPN) is characterized by myelin abnormalities; however, the molecular mechanisms underlying such deficits remain obscure. To uncover the effects of diabetes on myelin alterations, we have analyzed myelin composition. In a streptozotocin-treated rat model of diabetic neuropathy, analysis of sciatic nerve myelin lipids revealed that diabetes alters myelin's phospholipid, FA, and cholesterol content in a pattern that can modify membrane fluidity. Reduced expression of relevant genes in the FA biosynthetic pathway and decreased levels of the transcriptionally active form of the lipogenic factor sterol-regulatory element binding factor-1c (SREBF-1c) were found in diabetic sciatic nerve. Expression of myelin's major protein, myelin protein zero (P0), was also suppressed by diabetes. In addition, we confirmed that diabetes induces sciatic nerve myelin abnormalities, primarily infoldings that have previously been associated with altered membrane fluidity. In a diabetic setting, synthetic activator of the nuclear receptor liver X receptor (LXR) increased SREBF-1c function and restored myelin lipid species and P0 expression levels to normal. These LXR-modulated improvements were associated with restored myelin structure in sciatic nerve and enhanced performance in functional tests such as thermal nociceptive threshold and nerve conduction velocity. These findings demonstrate an important role for the LXR-SREBF-1c axis in protection from diabetes-induced myelin abnormalities.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Bainha de Mielina/metabolismo , Receptores Nucleares Órfãos/metabolismo , Animais , Colesterol/metabolismo , Diabetes Mellitus Experimental/genética , Regulação da Expressão Gênica , Lipídeos/química , Receptores X do Fígado , Masculino , Proteína P0 da Mielina/metabolismo , Bainha de Mielina/química , Fosfolipídeos/metabolismo , Proteínas Quinases/genética , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/patologia , Estearoil-CoA Dessaturase/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Estreptozocina
18.
J Neuroinflammation ; 9: 71, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22520439

RESUMO

BACKGROUND: Stress during fetal life increases the risk of affective and immune disorders later in life. The altered peripheral immune response caused by prenatal stress may impact on brain function by the modification of local inflammation. In this study we have explored whether prenatal stress results in alterations in the immune response in the hippocampus of female mice during adult life. METHODS: Pregnant C57BL/6 mice were subjected three times/day during 45 minutes to restraint stress from gestational Day 12 to delivery. Control non-stressed pregnant mice remained undisturbed. At four months of age, non-stressed and prenatally stressed females were ovariectomized. Fifteen days after surgery, mice received an i.p. injection of vehicle or of 5 mg/kg of lipopolysaccharide (LPS). Mice were sacrificed 20 hours later by decapitation and the brains were removed. Levels of interleukin-1ß (IL1ß), interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), interferon γ-inducible protein 10 (IP10), and toll-like receptor 4 mRNA were assessed in the hippocampus by quantitative real-time polymerase chain reaction. Iba1 immunoreactivity was assessed by immunocytochemistry. Statistical significance was determined by one-way or two-way analysis of variance. RESULTS: Prenatal stress, per se, increased IL1ß mRNA levels in the hippocampus, increased the total number of Iba1-immunoreactive microglial cells and increased the proportion of microglial cells with large somas and retracted cellular processes. In addition, prenatally stressed and non-stressed animals showed different responses to peripheral inflammation induced by systemic administration of LPS. LPS induced a significant increase in mRNA levels of IL-6, TNF-α and IP10 in the hippocampus of prenatally stressed mice but not of non-stressed animals. In addition, after LPS treatment, prenatally stressed animals showed a higher proportion of Iba1-immunoreactive cells in the hippocampus with morphological characteristics of activated microglia compared to non-stressed animals. In contrast, LPS induced similar increases in expression of IL1ß and toll-like receptor 4 in both prenatally stressed and non-stressed animals. CONCLUSION: These findings indicate that prenatal stress induces long-lasting modifications in the inflammatory status of the hippocampus of female mice under basal conditions and alters the immune response of the hippocampus to peripheral inflammation.


Assuntos
Hipocampo/patologia , Mediadores da Inflamação , Microglia/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Estresse Psicológico/patologia , Fatores Etários , Animais , Feminino , Hipocampo/imunologia , Imunidade Celular , Mediadores da Inflamação/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Distribuição Aleatória , Estresse Psicológico/complicações , Estresse Psicológico/imunologia
19.
Cereb Cortex ; 21(9): 2046-55, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21258044

RESUMO

The neuroactive steroid estradiol reduces reactive astroglia after brain injury by mechanisms similar to those involved in the regulation of reactive gliosis by endocannabinoids. In this study, we have explored whether cannabinoid receptors are involved in the effects of estradiol on reactive astroglia. To test this hypothesis, the effects of estradiol, the cannabinoid CB1 antagonist/inverse agonist AM251, and the cannabinoid CB2 antagonist/inverse agonist AM630 were assessed in the cerebral cortex of male rats after a stab wound brain injury. Estradiol reduced the number of vimentin immunoreactive astrocytes and the number of glial fibrillary acidic protein immunoreactive astrocytes in the proximity of the wound. The effect of estradiol was significantly inhibited by the administration of either CB1 or CB2 receptor antagonists. The effect of estradiol may be in part mediated by alterations in endocannabinoid signaling because the hormone increased in the injured cerebral cortex the messenger RNA levels of CB2 receptors and of some of the enzymes involved in the synthesis and metabolism of endocannabinoids. These findings suggest that estradiol may decrease reactive astroglia in the injured brain by regulating the activity of the endocannabinoid system.


Assuntos
Lesões Encefálicas/patologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Estradiol/farmacologia , Gliose/patologia , Gliose/prevenção & controle , Receptores de Canabinoides/efeitos dos fármacos , Animais , Moduladores de Receptores de Canabinoides/biossíntese , Canabinoides/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Indóis/farmacologia , Masculino , Piperidinas/farmacologia , Pirazóis/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Receptor CB1 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/agonistas , Técnicas Estereotáxicas , Fixação de Tecidos , Vimentina/farmacologia , Ferimentos Perfurantes/patologia
20.
Front Neuroanat ; 16: 910477, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35958733

RESUMO

The propensity to develop neurodegenerative diseases is influenced by diverse factors including genetic background, sex, lifestyle, including dietary habits and being overweight, and age. Indeed, with aging, there is an increased incidence of obesity and neurodegenerative processes, both of which are associated with inflammatory responses, in a sex-specific manner. High fat diet (HFD) commonly leads to obesity and markedly affects metabolism, both peripherally and centrally. Here we analyzed the metabolic and inflammatory responses of middle-aged (11-12 months old) transgenic amyloid precursor protein (TgAPP) mice of both sexes to HFD for 18 weeks (starting at 7-8 months of age). We found clear sex differences with females gaining significantly more weight and fat mass than males, with a larger increase in circulating leptin levels and expression of inflammatory markers in visceral adipose tissue. Glycemia and insulin levels increased in HFD fed mice of both sexes, with TgAPP mice being more affected than wild type (WT) mice. In the hypothalamus, murine amyloid ß (Aß) levels were increased by HFD intake exclusively in males, reaching statistical significance in TgAPP males. On a low fat diet (LFD), TgAPP males had significantly lower mRNA levels of the anorexigenic neuropeptide proopiomelanocortin (POMC) than WT males, with HFD intake decreasing the expression of the orexigenic neuropeptides Agouti-related peptide (AgRP) and neuropeptide Y (NPY), especially in TgAPP mice. In females, HFD increased POMC mRNA levels but had no effect on AgRP or NPY mRNA levels, and with no effect on genotype. There was no effect of diet or genotype on the hypothalamic inflammatory markers analyzed or the astrogliosis marker glial acidic protein (GFAP); however, levels of the microglial marker Iba-1 increased selectively in male TgAPP mice. In summary, the response to HFD intake was significantly affected by sex, with fewer effects due to genotype. Hypothalamic inflammatory cytokine expression and astrogliosis were little affected by HFD in middle-aged mice, although in TgAPP males, which showed increased Aß, there was microglial activation. Thus, excess intake of diets high in fat should be avoided because of its possible detrimental consequences.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA