Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Geroscience ; 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38954130

RESUMO

Pain perception is influenced by sex and aging, with previous studies indicating the involvement of aromatase, the estradiol synthase enzyme, in regulating pain perception. Previous research has established the presence of aromatase in dorsal root ganglia sensory neurons and its role in modulating pain perception. The present study aims to explore the implications of aging and sex on the expression of aromatase and estrogen receptors in the trigeminal ganglion. The study examined mRNA levels of aromatase, ERs, and the androgen receptor (AR) in the trigeminal ganglion of 3-month-old and 27-month-old male and female mice, as well as 3-month-old mice from the four-core genotype (FCG) transgenic model. The latter facilitates the assessment of gonadal hormone and sex chromosome implications for sex-specific traits. Aromatase localization in the ganglion was further assessed through immunohistochemistry. Aromatase immunoreactivity was observed for the first time in sensory neurons within the trigeminal ganglion. Trigeminal ganglion gene expressions were detected for aromatase, ERs, and AR in both sexes. Aromatase, ERß, and GPER gene expressions were higher in young males versus young females. Analyses of the FCG model indicated that sex differences depended solely on gonadal sex. The aging process induced an enhancement in the expression of aromatase, ERs, and AR genes across both sexes, culminating in a reversal of the previously observed gender-based differences. the potential impact of estrogen synthesis and signaling in the trigeminal ganglion on age and sex differences warrants consideration, particularly in relation to trigeminal sensory functions and pain perception.

2.
J Steroid Biochem Mol Biol ; 241: 106520, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38614433

RESUMO

Gonadal hormone deprivation (GHD) and decline such as menopause and bilateral oophorectomy are associated with an increased risk of neurodegeneration. Yet, hormone therapies (HTs) show varying efficacy, influenced by factors such as sex, drug type, and timing of treatment relative to hormone decline. We hypothesize that the molecular environment of the brain undergoes a transition following GHD, impacting the effectiveness of HTs. Using a GHD model in mice treated with Tibolone, we conducted proteomic analysis and identified a reprogrammed response to Tibolone, a compound that stimulates estrogenic, progestogenic, and androgenic pathways. Through a comprehensive network pharmacological workflow, we identified a reprogrammed response to Tibolone, particularly within "Pathways of Neurodegeneration", as well as interconnected pathways including "cellular respiration", "carbon metabolism", and "cellular homeostasis". Analysis revealed 23 proteins whose Tibolone response depended on GHD and/or sex, implicating critical processes like oxidative phosphorylation and calcium signalling. Our findings suggest the therapeutic efficacy of HTs may depend on these variables, suggesting a need for greater precision medicine considerations whilst highlighting the need to uncover underlying mechanisms.


Assuntos
Norpregnenos , Animais , Norpregnenos/farmacologia , Feminino , Camundongos , Proteômica/métodos , Moduladores de Receptor Estrogênico/farmacologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Camundongos Endogâmicos C57BL , Masculino , Ovariectomia , Hormônios Gonadais/metabolismo , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia
3.
Front Neuroendocrinol ; 71: 101085, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37543184

RESUMO

Substance use disorder (SUD) is a chronic condition characterized by pathological drug-taking and seeking behaviors. Remarkably different between males and females, suggesting that drug addiction is a sexually differentiated disorder. The neurobiological bases of sex differences in SUD include sex-specific reward system activation, influenced by interactions between gonadal hormone level changes, dopaminergic reward circuits, and epigenetic modifications of key reward system genes. This systematic review, adhering to PICOS and PRISMA-P 2015 guidelines, highlights the sex-dependent roles of estrogens, progesterone, and testosterone in SUD. In particular, estradiol elevates and progesterone reduces dopaminergic activity in SUD females, whilst testosterone and progesterone augment SUD behavior in males. Finally, SUD is associated with a sex-specific increase in the rate of opioid and monoaminergic gene methylation. The study reveals the need for detailed research on gonadal hormone levels, dopaminergic or reward system activity, and epigenetic landscapes in both sexes for efficient SUD therapy development.


Assuntos
Progesterona , Transtornos Relacionados ao Uso de Substâncias , Feminino , Humanos , Masculino , Dopamina/fisiologia , Epigênese Genética , Hormônios Esteroides Gonadais , Metanálise como Assunto , Caracteres Sexuais , Transtornos Relacionados ao Uso de Substâncias/genética , Revisões Sistemáticas como Assunto , Testosterona
4.
Neuroendocrinology ; 111(7): 660-677, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32570260

RESUMO

INTRODUCTION: The membrane-associated G protein-coupled estrogen receptor 1 (GPER) mediates the regulation by estradiol of arginine-vasopressin immunoreactivity in the supraoptic and paraventricular hypothalamic nuclei of female rats and is involved in the estrogenic control of hypothalamic regulated functions, such as food intake, sexual receptivity, and lordosis behavior. OBJECTIVE: To assess GPER distribution in the rat hypothalamus. METHODS: GPER immunoreactivity was assessed in different anatomical subdivisions of five selected hypothalamic regions of young adult male and cycling female rats: the arcuate nucleus, the lateral hypothalamus, the paraventricular nucleus, the supraoptic nucleus, and the ventromedial hypothalamic nucleus. GPER immunoreactivity was colocalized with NeuN as a marker of mature neurons, GFAP as a marker of astrocytes, and CC1 as a marker of mature oligodendrocytes. RESULTS: GPER immunoreactivity was detected in hypothalamic neurons, astrocytes, and oligodendrocytes. Sex and regional differences and changes during the estrous cycle were detected in the total number of GPER-immunoreactive cells and in the proportion of neurons, astrocytes, and oligodendrocytes that were GPER-immunoreactive. CONCLUSIONS: These findings suggest that estrogenic regulation of hypothalamic function through GPER may be different in males and females and may fluctuate during the estrous cycle in females.


Assuntos
Astrócitos/metabolismo , Ciclo Estral/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Oligodendroglia/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Caracteres Sexuais , Animais , Feminino , Imuno-Histoquímica , Masculino , Ratos , Ratos Wistar
5.
Trends Endocrinol Metab ; 31(10): 742-759, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32507541

RESUMO

Tibolone (TIB), a selective tissue estrogenic activity regulator (STEAR) in clinical use by postmenopausal women, activates hormonal receptors in a tissue-specific manner. Estrogenic activity is present mostly in the brain, vagina, and bone, while the inactive forms predominate in the endometrium and breast. Conflicting literature on TIB's actions has been observed. While it has benefits for vasomotor symptoms, bone demineralization, and sexual health, a higher relative risk of hormone-sensitive cancer has been reported. In the brain, TIB can improve mood and cognition, neuroinflammation, and reactive gliosis. This review aims to discuss the systemic effects of TIB on peri- and post-menopausal women and its role in the brain. We suggest that TIB is a hormonal therapy with promising neuroprotective properties.


Assuntos
Encéfalo/efeitos dos fármacos , Moduladores de Receptor Estrogênico/farmacologia , Menopausa/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Norpregnenos/farmacologia , Encéfalo/imunologia , Encéfalo/metabolismo , Moduladores de Receptor Estrogênico/efeitos adversos , Feminino , Humanos , Menopausa/imunologia , Menopausa/metabolismo , Norpregnenos/efeitos adversos
6.
Sci Rep ; 10(1): 8223, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32427857

RESUMO

Hypothalamic neurons show sex differences in neuritogenesis, female neurons have longer axons and higher levels of the neuritogenic factor neurogenin 3 (Ngn3) than male neurons in vitro. Moreover, the effect of 17-ß-estradiol (E2) on axonal growth and Ngn3 expression is only found in male-derived neurons. To investigate whether sex chromosomes regulate these early sex differences in neuritogenesis by regulating the E2 effect on Ngn3, we evaluated the growth and differentiation of hypothalamic neurons derived from the "four core genotypes" mouse model, in which the factors of "gonadal sex" and "sex chromosome complement" are dissociated. We showed that sex differences in neurite outgrowth are determined by sex chromosome complement (XX > XY). Moreover, E2 increased the mRNA expression of Ngn3 and axonal length only in XY neurons. ERα/ß expressions are regulated by sex chromosome complement; however, E2-effect on Ngn3 expression in XY neurons was only fully reproduced by PPT, a specific ligand of ERα, and prevented by MPP, a specific antagonist of ERα. Together our data indicate that sex chromosomes regulate early development of hypothalamic neurons by orchestrating not only sex differences in neuritogenesis, but also regulating the effect of E2 on Ngn3 expression through activation of ERα in hypothalamic neurons.


Assuntos
Axônios , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Estradiol/fisiologia , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Cromossomos Sexuais , Animais , Feminino , Masculino , Camundongos
7.
Front Neuroendocrinol ; 57: 100836, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32217094

RESUMO

The enzymatic complex 5α-reductase (5α-R) and 3α/3ß-hydroxysteroid oxidoreductase (HSOR) is expressed in the nervous system, where it transforms progesterone (PROG) and testosterone (T) into neuroactive metabolites. These metabolites regulate myelination, brain maturation, neurotransmission, reproductive behavior and the stress response. The expression of 5α-R and 3α-HSOR and the levels of PROG and T reduced metabolites show regional and sex differences in the nervous system and are affected by changing physiological conditions as well as by neurodegenerative and psychiatric disorders. A decrease in their nervous tissue levels may negatively impact the course and outcome of some pathological events. However, in other pathological conditions their increased levels may have a negative impact. Thus, the use of synthetic analogues of these steroids or 5α-R modulation have been proposed as therapeutic approaches for several nervous system pathologies. However, further research is needed to fully understand the consequences of these manipulations, in particular with 5α-R inhibitors.


Assuntos
3-Hidroxiesteroide Desidrogenases/fisiologia , Colestenona 5 alfa-Redutase/fisiologia , Progesterona/metabolismo , Testosterona/metabolismo , 3-Hidroxiesteroide Desidrogenases/genética , Animais , Encéfalo/enzimologia , Colestenona 5 alfa-Redutase/genética , Feminino , Expressão Gênica , Humanos , Masculino , Transtornos Mentais/enzimologia , Doenças Neurodegenerativas/enzimologia , Fármacos Neuroprotetores , Caracteres Sexuais
8.
J Neuroinflammation ; 17(1): 37, 2020 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-31992325

RESUMO

BACKGROUND: Tibolone is a synthetic steroid used in clinical practice for the treatment of climacteric symptoms and osteoporosis. Active metabolites of tibolone, generated in target tissues, have an affinity for estrogen and androgen receptors. Astrocytes are direct targets for estrogenic compounds and previous studies have shown that tibolone protects brain cortical neurons in association with a reduction in reactive astrogliosis in a mouse model of traumatic brain injury. Since phagocytosis is a crucial component of the neuroprotective function exerted by astrocytes, in the present study, we have assessed whether tibolone regulates phagocytosis in primary astrocytes incubated with brain-derived cellular debris. METHODS: Male and female astrocyte cell cultures were obtained from newborn (P0-P2) female and male Wistar rats. Astrocytic phagocytosis was first characterized using carboxylate beads, Escherichia coli particles, or brain-derived cellular debris. Then, the effect of tibolone on the phagocytosis of Cy3-conjugated cellular debris was quantified by measuring the intensity of Cy3 dye-emitted fluorescence in a given GFAP immunoreactive area. Before the phagocytosis assays, astrocytes were incubated with tibolone in the presence or absence of estrogen or androgen receptor antagonists or an inhibitor of the enzyme that synthesizes estradiol. The effect of tibolone on phagocytosis was analyzed under basal conditions and after inflammatory stimulation with lipopolysaccharide. RESULTS: Tibolone stimulated phagocytosis of brain-derived cellular debris by male and female astrocytes, with the effect being more pronounced in females. The effect of tibolone in female astrocytes was blocked by a selective estrogen receptor ß antagonist and by an androgen receptor antagonist. None of these antagonists affected tibolone-induced phagocytosis in male astrocytes. In addition, the inhibition of estradiol synthesis in the cultures enhanced the stimulatory effect of tibolone on phagocytosis in male astrocytes but blocked the effect of the steroid in female cells under basal conditions. However, after inflammatory stimulation, the inhibition of estradiol synthesis highly potentiated the stimulation of phagocytosis by tibolone, particularly in female astrocytes. CONCLUSIONS: Tibolone exerts sex-specific regulation of phagocytosis in astrocytes of both sexes, both under basal conditions and after inflammatory stimulation.


Assuntos
Astrócitos/efeitos dos fármacos , Inflamação/patologia , Norpregnenos/farmacologia , Fagocitose/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Antagonistas de Receptores de Andrógenos/farmacologia , Animais , Estradiol/biossíntese , Antagonistas de Estrogênios/farmacologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Inflamação/induzido quimicamente , Lipopolissacarídeos , Masculino , Microglia/efeitos dos fármacos , Ratos , Ratos Wistar
9.
J Neuroendocrinol ; 32(1): e12776, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31334878

RESUMO

The high concentrations of free fatty acids as a consequence of obesity and being overweight have become risk factors for the development of different diseases, including neurodegenerative ailments. Free fatty acids are strongly related to inflammatory events, causing cellular and tissue alterations in the brain, including cell death, deficits in neurogenesis and gliogenesis, and cognitive decline. It has been reported that people with a high body mass index have a higher risk of suffering from Alzheimer's disease. Hormones such as oestradiol not only have beneficial effects on brain tissue, but also exert some adverse effects on peripheral tissues, including the ovary and breast. For this reason, some studies have evaluated the protective effect of oestrogen receptor (ER) agonists with more specific tissue activities, such as the neuroactive steroid tibolone. Activation of ERs positively affects the expression of pro-survival factors and cell signalling pathways, thus promoting cell survival. This review aims to discuss the relationship between lipotoxicity and the development of neurodegenerative diseases. We also elaborate on the cellular and molecular mechanisms involved in neuroprotection induced by oestrogens.


Assuntos
Encéfalo/metabolismo , Estrogênios/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Inflamação/metabolismo , Neuroglia/metabolismo , Animais , Encéfalo/patologia , Humanos , Inflamação/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neuroglia/patologia , Transdução de Sinais/fisiologia
10.
J Neuroendocrinol ; 32(1): e12756, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31179596

RESUMO

Obesity is one of the most important health problems facing developed countries because being overweight is associated with a higher incidence of type 2 diabetes, cardiovascular disease and cancer, as well as other comorbidities. Although increased weight gain results from a combination of poor dietary habits and decreased energy expenditure, not all individuals have equal propensities to gain weight or to develop secondary complications of obesity. This is partially a result not only of genetics, including sex, but also the time during which an individual is exposed to an obesogenic environment. In the present study, we have compared the response of male and female mice to short-term exposure to a high-fat diet (HFD) or a low-fat diet during the peripubertal period (starting at 42 days of age) because this is a stage of dramatic hormonal and metabolic modifications. After 1 week on a HFD, there was no significant increase in body weight, although females significantly increased their energy intake. Serum leptin levels increased in both sexes, even though no change in fat mass was detected. Glyceamia and homeostasis model assessment increased in males, suggesting a rapid change in glucose metabolism. Hypothalamic pro-opiomelanocortin mRNA levels were significantly higher in females on a HFD compared to all other groups, which may be an attempt to reduce their increased energy intake. Hypothalamic inflammation and gliosis have been implicated in the development of secondary complications of obesity; however, no indication of activation of inflammatory processes or gliosis was found in response to 1 week of HFD in the hypothalamus, hippocampus or cerebellum of these young mice. These results indicate that there are both sex and age effects in the response to poor dietary intake because peripubertal male and female mice respond differently to short-term dietary changes and this response is different from that reported in adult rodents.


Assuntos
Glicemia/metabolismo , Peso Corporal/fisiologia , Dieta Hiperlipídica , Hipotálamo/metabolismo , Maturidade Sexual/fisiologia , Adiposidade/fisiologia , Animais , Ingestão de Energia/fisiologia , Feminino , Insulina/sangue , Leptina/sangue , Masculino , Camundongos , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Fatores Sexuais
11.
Int J Mol Sci ; 20(10)2019 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-31109056

RESUMO

Developmental actions of estradiol in the hypothalamus are well characterized. This hormone generates sex differences in the development of hypothalamic neuronal circuits controlling neuroendocrine events, feeding, growth, reproduction and behavior. In vitro, estradiol promotes sexually dimorphic effects on hypothalamic neuritogenesis. Previous studies have shown that developmental actions of the phytoestrogen genistein result in permanent sexually dimorphic effects in some behaviors and neural circuits in vivo. In the present study, we have explored if genistein, like estradiol, affects neuritogenesis in primary hypothalamic neurons and investigated the estrogen receptors implicated in this action. Hypothalamic neuronal cultures, obtained from male or female embryonic day 14 (E14) CD1 mice, were treated with genistein (0.1 µM, 0.5 µM or 1 µM) or vehicle. Under basal conditions, female neurons had longer primary neurites, higher number of secondary neurites and higher neuritic arborization compared to male neurons. The treatment with genistein increased neuritic arborization and the number of primary neurites and decreased the number of secondary neurites in female neurons, but not in male neurons. In contrast, genistein resulted in a significant increase in primary neuritic length in male neurons, but not in female neurons. The use of selective estrogen receptor antagonists suggests that estrogen receptor α, estrogen receptor ß and G-protein-coupled estrogen receptors are involved in the neuritogenic action of genistein. In summary, these findings indicate that genistein exerts sexually dimorphic actions on the development of hypothalamic neurons, altering the normal pattern of sex differences in neuritogenesis.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Genisteína/farmacologia , Fitoestrógenos/farmacologia , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Caracteres Sexuais , Animais , Biomarcadores , Feminino , Masculino , Camundongos , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurogênese/efeitos dos fármacos , Células Piramidais/metabolismo , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo
12.
Mol Cell Endocrinol ; 486: 65-78, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30822454

RESUMO

Palmitic acid (PA) induces several metabolic and molecular changes in astrocytes, and, it is involved in pathological conditions related to neurodegenerative diseases. Previously, we demonstrated that tibolone, a synthetic steroid with estrogenic, progestogenic and androgenic actions, protects cells from mitochondrial damage and morphological changes induced by PA. Here, we have evaluated which estrogen receptor is involved in protective actions of tibolone and analyzed whether tibolone reverses gene expression changes induced by PA. Tibolone actions on astrocytic cells were mimicked by agonists of estrogen receptor α (ERα) and ß (ERß), but the blockade of both ERs suggested a predominance of ERß on mitochondria membrane potential. Expression analysis showed a significant effect of tibolone on genes associated with inflammation such as IL6, IL1B and miR155-3p. It is noteworthy that tibolone attenuated the increased expression of TERT, TERC and DNMT3B genes induced by palmitic acid. Our results suggest that tibolone has anti-inflammatory effects and can modulate pathways associated with DNA methylation and telomeric complex. However, future studies are needed to elucidate the role of epigenetic mechanisms and telomere-associated proteins on tibolone actions.


Assuntos
Astrócitos/metabolismo , Receptor alfa de Estrogênio/metabolismo , Inflamação/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Norpregnenos/farmacologia , Ácido Palmítico/toxicidade , Astrócitos/efeitos dos fármacos , Linhagem Celular , Epigênese Genética/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/metabolismo , Humanos , Inflamação/genética , Nitrilas/farmacologia , Fenóis , Substâncias Protetoras/farmacologia , Pirazóis , Telomerase/genética , Telomerase/metabolismo , Telômero/metabolismo , Fatores de Transcrição/metabolismo
13.
Neuroendocrinology ; 108(2): 142-160, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30391959

RESUMO

Traumatic brain injury (TBI) is a serious public health problem. It may result in severe neurological disabilities and in a variety of cellular metabolic alterations for which available therapeutic strategies are limited. In the last decade, the use of estrogenic compounds, which activate protective mechanisms in astrocytes, has been explored as a potential experimental therapeutic approach. Previous works have suggested estradiol (E2) as a neuroprotective hormone that acts in the brain by binding to estrogen receptors (ERs). Several steroidal and nonsteroidal estrogenic compounds can imitate the effects of estradiol on ERs. These include hormonal estrogens, phytoestrogens and synthetic estrogens, such as selective ER modulators or tibolone. Current evidence of the role of astrocytes in mediating protective actions of estrogenic compounds after TBI is reviewed in this paper. We conclude that the use of estrogenic compounds to modulate astrocytic properties is a promising therapeutic approach for the treatment of TBI.


Assuntos
Astrócitos/efeitos dos fármacos , Lesões Encefálicas Traumáticas/tratamento farmacológico , Estrogênios/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Receptores de Estrogênio/metabolismo , Animais , Astrócitos/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Estrogênios/farmacologia , Humanos , Fármacos Neuroprotetores/farmacologia
14.
Mol Neurobiol ; 55(7): 5462-5477, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28948468

RESUMO

High concentrations of palmitic acid in plasma increase both the inflammation associated with obesity and the susceptibility to develop a neurodegenerative event. In the brain, the inflammatory response is mediated by activated microglial cells, which undergo morphological and biochemical changes and can directly affect cell viability. Recent evidence shows that the use of estrogenic compounds can control microglia-induced inflammation with promising results. In this study, we explored the actions of the synthetic steroid tibolone on BV-2 microglia cells stimulated with palmitic acid. Our results demonstrated that tibolone increased cell viability and reduced nuclear fragmentation and the production of reactive oxygen species, as well as preserved mitochondrial membrane potential. These effects were accompanied by reduced nuclear translocation of NF-κB p65, upregulation of neuroglobin, and improved antioxidant defense. Furthermore, estrogen receptor beta (ERß) inhibition partially dampened tibolone's protective actions in BV-2 cells stimulated with palmitic acid. In conclusion, tibolone protects BV-2 cells by a mechanism involving ERß and upregulation of neuroglobin.


Assuntos
Receptor beta de Estrogênio/metabolismo , Inflamação/patologia , Microglia/metabolismo , Microglia/patologia , Norpregnenos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ácido Palmítico/toxicidade , Animais , Antioxidantes/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Receptor beta de Estrogênio/agonistas , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Microglia/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neuroglobina/metabolismo , Fármacos Neuroprotetores/farmacologia , Nitrilas/farmacologia , Oxirredução , Pirazóis/farmacologia , Pirimidinas/farmacologia , Receptores Androgênicos/metabolismo , Fator de Transcrição RelA/metabolismo
15.
Endocrinology ; 159(1): 368-387, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29077836

RESUMO

Astrocytes participate in both physiological and pathophysiological responses to metabolic and nutrient signals. Although most studies have focused on the astrocytic response to weight gain due to high-fat/high-carbohydrate intake, surplus intake of a balanced diet also induces excess weight gain. We have accessed the effects of neonatal overnutrition, which has both age- and sex-dependent effects on weight gain, on hypothalamic inflammation/gliosis. Although both male and female Wistar rats accumulate excessive fat mass as early as postnatal day (PND) 10 with neonatal overnutrition, no increase in hypothalamic cytokine levels, markers of astrocytes or microglia, or inflammatory signaling pathways were observed. At PND 50, no effect of neonatal overnutriton was found in either sex, whereas at PND 150, males again weighed significantly more than their controls, and this was coincident with an increase in markers of inflammation and astrogliosis in the hypothalamus. Circulating triglycerides and free fatty acids were also elevated in these males, but not in females or in either sex at PND 10. Thus, the effects of fatty acids and estrogens on astrocytes in vitro were analyzed. Our results indicate that changes in circulating fatty acid levels may be involved in the induction of hypothalamic inflammation/gliosis in excess weight gain, even on a normal diet, and that estrogens could participate in the protection of females from these processes. In conclusion, the interaction of developmental influences, dietary composition, age, and sex determines the central inflammatory response and the associated long-term outcomes of excess weight gain.


Assuntos
Astrócitos/metabolismo , Gliose/etiologia , Hiperfagia/fisiopatologia , Doenças Hipotalâmicas/etiologia , Hipotálamo/metabolismo , Microglia/metabolismo , Adiposidade , Fatores Etários , Animais , Animais Recém-Nascidos , Astrócitos/imunologia , Astrócitos/patologia , Biomarcadores/metabolismo , Células Cultivadas , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Gliose/imunologia , Gliose/metabolismo , Gliose/patologia , Doenças Hipotalâmicas/imunologia , Doenças Hipotalâmicas/metabolismo , Doenças Hipotalâmicas/patologia , Hipotálamo/imunologia , Hipotálamo/patologia , Mediadores da Inflamação/metabolismo , Masculino , Microglia/imunologia , Microglia/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ratos Wistar , Caracteres Sexuais , Transdução de Sinais , Aumento de Peso
16.
Mol Neurobiol ; 55(5): 4453-4462, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28667487

RESUMO

Obesity has been associated with increased chronic neuroinflammation and augmented risk of neurodegeneration. This is worsened during the normal aging process when the levels of endogenous gonadal hormones are reduced. In this study, we have assessed the protective actions of tibolone, a synthetic steroid with estrogenic actions, on T98G human astrocytic cells exposed to palmitic acid, a saturated fatty acid used to mimic obesity in vitro. Tibolone improved cell survival, and preserved mitochondrial membrane potential in palmitic acid-treated astrocytic cells. Although we did not find significant actions of tibolone on free radical production, it modulated astrocytic morphology after treatment with palmitic acid. These data suggest that tibolone protects astrocytic cells by preserving both mitochondrial functionality and morphological complexity.


Assuntos
Astrócitos/citologia , Forma Celular/efeitos dos fármacos , Mitocôndrias/metabolismo , Norpregnenos/farmacologia , Ácido Palmítico/toxicidade , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos
17.
Sci Rep ; 7(1): 5320, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28706210

RESUMO

During development sex differences in aromatase expression in limbic regions of mouse brain depend on sex chromosome factors. Genes on the sex chromosomes may affect the hormonal regulation of aromatase expression and this study was undertaken to explore that possibility. Male E15 anterior amygdala neuronal cultures expressed higher levels of aromatase (mRNA and protein) than female cultures. Furthermore, treatment with oestradiol (E2) or dihydrotestosterone (DHT) increased Cyp19a1 expression and aromatase protein levels only in female neuronal cultures. The effect of E2 on aromatase expression was not imitated by oestrogen receptor (ER) α agonist PPT or the GPER agonist G1, but it was fully reproduced by DPN, a specific ligand of ERß. By contrast, the effect of DHT on aromatase expression was not blocked by the anti-androgen flutamide, but completely abrogated by the ERß antagonist PHTPP. Experiments using the four core genotype model showed a sex chromosome effect in ERß expression (XY > XX) and regulation by E2 or DHT (only XX respond) in amygdala neurons. In conclusion, sex chromosome complement governs the hormonal regulation of aromatase expression through activation of ERß in developing mouse brain.


Assuntos
Tonsila do Cerebelo/embriologia , Tonsila do Cerebelo/enzimologia , Aromatase/biossíntese , Receptor beta de Estrogênio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/enzimologia , Cromossomos Sexuais , Animais , Células Cultivadas , Di-Hidrotestosterona/metabolismo , Estradiol/metabolismo , Feminino , Masculino , Camundongos
18.
Neuroendocrinology ; 104(1): 40-50, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-26862917

RESUMO

Obesity is associated with increased fever and sickness behavior in response to infection. The hypothalamic-pituitary-adrenal (HPA) axis plays a key role in the reaction to immune stimuli. Bacterial infection, or bacterial lipopolysaccharide (LPS), induces the expression of peripheral cytokines that stimulate the hypothalamus and the hippocampus and activate the HPA axis. In this study, we explored whether the hypothalamic and hippocampal responses to infection are altered during the development of diet-induced obesity. Male mice were exposed to a high-fat diet (HFD) or a low-fat diet (LFD) for 15 days. They were then administered a single intraperitoneal injection of bacterial LPS or vehicle and sacrificed 24 h later. LPS increased circulating levels of insulin and leptin, but only in LFD animals. LPS induced a significant decrease in hypothalamic corticotrophin-releasing hormone and glucocorticoid receptor mRNA levels in LFD animals but exerted the opposite effect in HFD-fed mice. LPS increased the hypothalamic expression of molecules involved in the leptin signaling pathway (SOCS3 and STAT3), nuclear factor-κB pathway members, inflammatory mediators (tumor necrosis factor-α and interleukin-6) and glial proliferation markers (Emr1 and CD68) in LFD animals. These effects were dampened in HFD-fed mice. In contrast, the hippocampal responses to LPS were largely insensitive to HFD. These results suggest that HFD feeding reduced the inflammatory response induced by LPS in the hypothalamus but not in the hippocampus.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hipocampo/metabolismo , Hipotálamo/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Obesidade/etiologia , Adiponectina/sangue , Análise de Variância , Animais , Peso Corporal/efeitos dos fármacos , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Citocinas/genética , Citocinas/metabolismo , Gorduras na Dieta , Modelos Animais de Doenças , Ingestão de Alimentos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Inflamação/induzido quimicamente , Insulina/sangue , Leptina/sangue , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/sangue , RNA Mensageiro , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos
19.
Neuroendocrinology ; 104(1): 94-104, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-26954778

RESUMO

Nitric oxide is produced in the brain by the neuronal nitric oxide synthase (nNOS) and carries out a wide range of functions by acting as a neurotransmitter-like molecule. Gonadal hormones are involved in the regulation of the brain nitrergic system. We have previously demonstrated that estradiol, via classical estrogen receptors (ERs), regulates NOS activity in the supraoptic (SON) and paraventricular (PVN) nuclei of the hypothalamus, acting through both ERα and ERß. Magnocellular and parvocellular neurons in the SON and PVN also express the G protein-coupled ER (GPER). In this study, we have assessed whether GPER is also involved in the regulation of nicotinamide adenine dinucleotide phosphate (NADPH)-diaphorase in the SON and PVN. Adult female ovariectomized rats were treated with G1, a selective GPER agonist, or with G1 in combination with G15, a selective GPER antagonist. G1 treatment decreased NADPH-diaphorase expression in the SON and in all PVN subnuclei. The treatment with G1 + G15 effectively rescued the G1-dependent decrease in NADPH-diaphorase expression in both brain regions. In addition, the activation of extracellular signal-regulated kinase (ERK) 1/2, one of the kinases involved in the GPER-dependent intracellular signaling pathway and in NOS phosphorylation, was assessed in the same brain nuclei. Treatment with G1 significantly decreased the number of p-ERK 1/2-positive cells in the SON and PVN, while the treatment with G1 + G15 significantly recovered its number to control values. These findings suggest that the activation of GPER in the SON and PVN inhibits the phosphorylation of ERK 1/2, which induces a decrease in NADPH-diaphorase expression.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , NADPH Desidrogenase/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Quinolinas/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Núcleo Supraóptico/efeitos dos fármacos , Animais , Benzodioxóis/farmacologia , Contagem de Células , Feminino , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ovariectomia , Ratos , Ratos Wistar , Núcleo Supraóptico/metabolismo
20.
J Steroid Biochem Mol Biol ; 167: 135-143, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27890531

RESUMO

Diabetes may induce neurophysiological and structural changes in the central nervous system (i.e., diabetic encephalopathy). We here explored whether the levels of neuroactive steroids (i.e., neuroprotective agents) in the hippocampus may be altered by short-term diabetes (i.e., one month). To this aim, by liquid chromatography-tandem mass spectrometry we observed that in the experimental model of the rat raised diabetic by streptozotocin injection, one month of pathology induced changes in the levels of several neuroactive steroids, such as pregnenolone, progesterone and its metabolites (i.e., tetrahydroprogesterone and isopregnanolone) and testosterone and its metabolites (i.e., dihydrotestosterone and 3α-diol). Interestingly these brain changes were not fully reflected by the plasma level changes, suggesting that early phase of diabetes directly affects steroidogenesis and/or steroid metabolism in the hippocampus. These concepts are also supported by the findings that crucial steps of steroidogenic machinery, such as the gene expression of steroidogenic acute regulatory protein (i.e., molecule involved in the translocation of cholesterol into mitochondria) and cytochrome P450 side chain cleavage (i.e., enzyme converting cholesterol into pregnenolone) and 5α-reductase (enzyme converting progesterone and testosterone into their metabolites) are also affected in the hippocampus. In addition, cholesterol homeostasis as well as the functionality of mitochondria, a key organelle in which the limiting step of neuroactive steroid synthesis takes place, are also affected. Data obtained indicate that short-term diabetes alters hippocampal steroidogenic machinery and that these changes are associated with impaired cholesterol homeostasis and mitochondrial dysfunction in the hippocampus, suggesting them as relevant factors for the development of diabetic encephalopathy.


Assuntos
Diabetes Mellitus/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Esteroides/biossíntese , Animais , Sistema Nervoso Central/metabolismo , Colesterol/metabolismo , Cromatografia Líquida , Di-Hidrotestosterona/metabolismo , Homeostase , Masculino , Mitocôndrias/metabolismo , Estresse Oxidativo , Pregnenolona/biossíntese , Progesterona/biossíntese , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem , Testosterona/biossíntese , Substâncias Reativas com Ácido Tiobarbitúrico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA