Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Endocrinol ; 258(3)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37399525

RESUMO

Estrogens regulate synaptic properties and influence hippocampus-related learning and memory via estrogen receptors, which include the G-protein-coupled estrogen receptor 1 (GPER1). Studying mice, in which the GPER1 gene is dysfunctional (GPER1-KO), we here provide evidence for sex-specific roles of GPER1 in these processes. GPER1-KO males showed reduced anxiety in the elevated plus maze, whereas the fear response ('freezing') was specifically increased in GPER1-KO females in a contextual fear conditioning paradigm. In the Morris water maze, spatial learning and memory consolidation was impaired by GPER1 deficiency in both sexes. Notably, in the females, spatial learning deficits and the fear response were more pronounced if mice were in a stage of the estrous cycle, in which E2 serum levels are high (proestrus) or rising (diestrus). On the physiological level, excitability at Schaffer collateral synapses in CA1 increased in GPER1-deficient males and in proestrus/diestrus ('E2 high') females, concordant with an increased hippocampal expression of the AMPA-receptor subunit GluA1 in GPER1-KO males and females as compared to wildtype males. Further changes included an augmented early long-term potentiation (E-LTP) maintenance specifically in GPER1-KO females and an increased hippocampal expression of spinophilin in metestrus/estrus ('E2 low') GPER1-KO females. Our findings suggest modulatory and sex-specific functions of GPER1 in the hippocampal network, which reduce rather than increase neuronal excitability. Dysregulation of these functions may underlie sex-specific cognitive deficits or mood disorders.


Assuntos
Hipocampo , Receptores de Estrogênio , Masculino , Feminino , Camundongos , Animais , Receptores de Estrogênio/genética , Potenciação de Longa Duração/genética , Sinapses/fisiologia , Cognição , Plasticidade Neuronal/genética
3.
Psychoneuroendocrinology ; 156: 106320, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37307791

RESUMO

In order to translate the findings from the vast animal literature on the effect of 17ß-estradiol (E2) on brain and behavior to humans, a placebo-controlled pharmacological enhancement of E2 levels for at least 24 h is necessary. However, an exogenous increase in E2 for such a prolonged period might affect the endogenous secretion of other (neuroactive) hormones. Such effects would be of relevance for the interpretation of the effects of this pharmacological regimen on cognition and its neural correlates as well as be of basic scientific interest. We therefore administered a double dose of 12 mg of estradiol-valerate (E2V) to men and of 8 mg to naturally cycling women in their low-hormone phase, and assessed the concentration of two steroids critical to hormone regulation: follicle stimulating hormone (FSH) and luteinizing hormone (LH). We also assessed any changes in concentration of the neuroactive hormones progesterone (P4), testosterone (TST), dihydrotestosterone (DHT) and immune-like growth factor 1 (IGF-1). This regimen resulted in similar E2 levels in both sexes (saliva and serum). FSH and LH levels in both sexes were down-regulated to the same degree. P4 concentration decreased in both sexes only in serum but not saliva. TST and DHT levels dropped only in men whereas sex-hormone binding globulin was not affected. Finally, the concentration of IGF-1 decreased in both sexes. Based on previous studies on the effects of these neuroactive hormones, only the degree of downregulation of TST and DHT levels in men might have an impact on brain and behavior, which should be considered when interpreting the effects of the presented E2V regimes.


Assuntos
Fator de Crescimento Insulin-Like I , Hormônio Luteinizante , Masculino , Animais , Humanos , Feminino , Estradiol/farmacologia , Hormônio Foliculoestimulante , Testosterona/farmacologia , Menopausa , Valeratos
4.
J Neuroendocrinol ; 35(5): e13276, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37170708

RESUMO

Microglia have been shown to sculpt postnatal circuitry from birth up to adulthood due to their role in both synapse formation, synaptic pruning, and the elimination of weak, redundant synapses. Microglia are differentiated in a sex-dependent manner. In this study, we tested whether sexual differentiation of microglia results in sex-dependent postnatal reorganization of CA1 synaptic connectivity in the hippocampus. The stereological counting of synapses in mice using electron microscopy showed a continuous rise in synapse density until the fourth week, followed by a plateau phase and loss of synapses from the eighth week onwards, with no difference between sexes. This course of alteration in synapse numbers did not differ between sexes. However, selectively, on postnatal day (P) 14 the density of synapses was significantly higher in the female than in the male hippocampus. Higher synapse density in females was paralleled by higher activity of microglia, as indicated by morphological changes, CD68 expression, and proximity of microglia to synaptic sites. In Thy1-GFP mice, consistent with increased synapse numbers, bouton density was also clearly increased in females at P14. At this time point, CD47 expression, the "don't eat me" signal of neurons, was similar in males and females. The decrease in bouton density thereafter in conjunction with increased synapse numbers argues for a role of microglia in the formation of multispine boutons (MSB). Our data in females at P14 support the regulatory role of microglia in synapse density. Sexual differentiation of microglia, however, does not substantially affect long-term synaptic reorganization in the hippocampus.


Assuntos
Hipocampo , Microglia , Camundongos , Masculino , Feminino , Animais , Microglia/metabolismo , Neurônios , Sinapses/metabolismo , Terminações Pré-Sinápticas
5.
J Neuroendocrinol ; 34(2): e13090, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35081672

RESUMO

Sex steroids, such as estradiol (E2 ) and dihydrotestosterone (DHT), regulate hippocampal plasticity and memory in a sex-dependent manner. Because the activity-regulated cytoskeleton protein Arc/Arg3.1 is essential for long-term memory formation and synaptic plasticity, we investigated the expression of Arc/Arg3.1 with respect to its responsiveness to E2 and DHT in male and female hippocampal neurons. For the first time, we show that, in hippocampal neurons, Arc/Arg3.1 expression is sex-dependently regulated by sex steroids. No difference in the expression between sexes was observed under control conditions. Using a quantitative real-time polymerase chain reaction, western blot analysis and quantitative immunoreactivity, upregulation of Arc/Arg3.1 protein expression was observed in specifically female hippocampal neurons after application of E2 to the cultures. Conversely, upregulation of Arc/Arg3.1 was seen in specifically male neurons after application of DHT. A quantitative real-time PCR revealed that the sex-dependency was most pronounced on the mRNA level. Most importantly, the effects of E2 in cultures of female animals were abolished when neuron-derived E2 synthesis was inhibited. Our results point to a potentially important role of Arc/Arg3.1 regarding sex-dependency in sex steroid-induced synaptic plasticity in the hippocampus.


Assuntos
Neuroesteroides , Animais , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Di-Hidrotestosterona/farmacologia , Estradiol/metabolismo , Estradiol/farmacologia , Feminino , Hipocampo/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo
6.
Neuroscience ; 472: 35-50, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34364953

RESUMO

G-protein-coupled-estrogen-receptor 1 (GPER1) is a membrane-bound receptor that mediates estrogen signaling via intracellular signaling cascades. We recently showed that GPER1 promotes the distal dendritic enrichment of hyperpolarization activated and cyclic nucleotide-gated (HCN)1 channels in CA1 stratum lacunosum-moleculare (SLM), suggesting a role of GPER1-mediated signaling in neuronal plasticity. Here we studied whether this role involves processes of structural plasticity, such as the regulation of spine and synapse density in SLM. In organotypic entorhino-hippocampal cultures from mice expressing eGFP, we analyzed spine densities in SLM after treatment with GPER1 agonist G1 (20 nM). G1 significantly increased the density of "non-stubby" spines (maturing spines with a spine head and a neck), but did so only in cultures from female mice. In support of this finding, the expression of synaptic proteins was sex-specifically altered in the cultures: G1 increased the protein (but not mRNA) expression of PSD95 and reduced the p-/n-cofilin ratio only in cultures from females. Application of E2 (2 nM) reproduced the sex-specific effect on spine density in SLM, but only partially on the expression of synaptic proteins. Spine synapse density was, however, not altered after G1-treatment, suggesting that the increased spine density did not translate into an increased spine synapse density in the culture model. Taken together, our results support a role of GPER1 in mediating structural plasticity in CA1 SLM, but suggest that in developing hippocampus, this role is sex-specific.


Assuntos
Hipocampo , Receptores de Estrogênio , Animais , Espinhas Dendríticas/metabolismo , Feminino , Proteínas de Ligação ao GTP , Hipocampo/metabolismo , Masculino , Camundongos , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G , Sinapses/metabolismo
7.
Neurosci Lett ; 753: 135849, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33775739

RESUMO

In general, hippocampal neurons are capable of synthesizing sex steroids de novo from cholesterol, since the brain is equipped with all the enzymes required for the synthesis of estradiol and testosterone, the end products of sex steroidogenesis. Regarding estradiol, its synthesis in hippocampal neurons is homeostatically controlled by Ca2+ transients and is regulated by GnRH. Locally synthesized estradiol and testosterone maintain synaptic transmission and synaptic connectivity. Remarkably, the neurosteroid estradiol is effective in females, but not in males, and vice versa dihydrotestosterone (DHT) is effective in males, but not in females. Experimentally induced inhibition of estradiol synthesis in females and DHT synthesis in males resp. results in synapse loss, impaired LTP, and downregulation of synaptic proteins. GnRH-induced increase in estradiol synthesis appears to provide a link between the hypothalamus and the hippocampus, which may underlie estrous cyclicity of spine density in the female hippocampus. Hippocampal neurons are sex-dependently differentiated with respect to the responsiveness of hippocampal neurons to sex neurosteroids.


Assuntos
Di-Hidrotestosterona/metabolismo , Estradiol/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Neuroesteroides/metabolismo , Animais , Diferenciação Celular , Ciclo Estral , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipocampo/citologia , Humanos , Hipotálamo/citologia , Masculino , Plasticidade Neuronal , Fatores Sexuais , Transmissão Sináptica
8.
Sci Rep ; 10(1): 11405, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647191

RESUMO

Previously, we found that in dissociated hippocampal cultures the proportion of large spines (head diameter ≥ 0.6 µm) was larger in cultures from female than from male animals. In order to rule out that this result is an in vitro phenomenon, we analyzed the density of large spines in fixed hippocampal vibratome sections of Thy1-GFP mice, in which GFP is expressed only in subpopulations of neurons. We compared spine numbers of the four estrus cycle stages in females with those of male mice. Remarkably, total spine numbers did not vary during the estrus cycle, while estrus cyclicity was evident regarding the number of large spines and was highest during diestrus, when estradiol levels start to rise. The average total spine number in females was identical with the spine number in male animals. The density of large spines, however, was significantly lower in male than in female animals in each stage of the estrus cycle. Interestingly, the number of spine apparatuses, a typical feature of large spines, did not differ between the sexes. Accordingly, NMDA-R1 and NMDA-R2A/B expression were lower in the hippocampus and in postsynaptic density fractions of adult male animals than in those of female animals. This difference could already be observed at birth for NMDA-R1, but not for NMDA-R2A/B expression. In dissociated embryonic hippocampal cultures, no difference was seen after 21 days in culture, while the difference was evident in postnatal cultures. Our data indicate that hippocampal neurons are differentiated in a sex-dependent manner, this differentiation being likely to develop during the perinatal period.


Assuntos
Região CA1 Hipocampal/citologia , Espinhas Dendríticas/ultraestrutura , Caracteres Sexuais , Envelhecimento , Animais , Animais Recém-Nascidos , Região CA1 Hipocampal/crescimento & desenvolvimento , Células Cultivadas , Estro , Feminino , Genes Reporter , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica , Cultura Primária de Células , Células Piramidais/ultraestrutura , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/análise
9.
iScience ; 23(5): 101077, 2020 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-32361597

RESUMO

In the rodent ventricular-subventricular zone (V-SVZ) neurons are generated throughout life. They migrate along the rostral migratory stream (RMS) into the olfactory bulb before their final differentiation into interneurons and integration into local circuits. Estrogen receptors (ERs) are steroid hormone receptors with important functions in neurogenesis and synaptic plasticity. In this study, we show that the ER GPER1 is expressed in subsets of cells within the V-SVZ of female animals and provide evidence for a potential local estrogen source from aromatase-positive astrocytes surrounding the RMS. Blocking of GPER1 in Matrigel cultures of female animals significantly impairs migration of V-SVZ-derived cells. This outgrowth is accompanied by regulation of phosphorylation of the actin-binding protein cofilin by GPER1 signaling including an involvement of the p21-Ras pathway. Our results point to a prominent role of GPER1 in the initiation of neuronal migration from the V-SVZ to the olfactory bulb.

10.
Eur J Neurosci ; 52(1): 2548-2559, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31403726

RESUMO

Sex-dependent differences in learning and memory formation in humans have been frequently shown. The mechanisms underlying the formation and retention of memories are assumed to involve synaptic plasticity in the hippocampus. Estradiol was shown to effect synaptic plasticity in the hippocampus of rodents. The effects after exogenous application of estradiol to animals frequently produce inconsistent results, in particular, if sex is not considered in the studies. Recently we provided evidence that locally synthesized estradiol plays an essential role on synaptic connectivity in the hippocampus of females but not of male mice. In females, inhibition of local estradiol synthesis leads to synapse loss, which results from impairment of long-term potentiation and dephosphorylation of cofilin, and thereby the destabilization of postsynaptic dendritic spines. This sex-dependency was also seen in the classical aromatase knock-out mouse. Intriguingly, no differences between sexes have been found in a conditional forebrain-specific aromatase knock-out mouse. Altogether, the findings underscore the necessity of including 'Sex as a Biological Variable' in studies of sex steroid-induced synaptic plasticity.


Assuntos
Aromatase , Estradiol , Animais , Aromatase/metabolismo , Espinhas Dendríticas/metabolismo , Estradiol/farmacologia , Estrogênios , Feminino , Hipocampo/metabolismo , Masculino , Camundongos , Plasticidade Neuronal , Roedores/metabolismo
11.
Cereb Cortex ; 30(4): 2627-2641, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-31800024

RESUMO

Numerous studies provide increasing evidence, which supports the ideas that every cell in the brain of males may differ from those in females due to differences in sex chromosome complement as well as in response to hormonal effects. In this study, we address the question as to whether actions of neurosteroids, thus steroids, which are synthesized and function within the brain, contribute to sex-specific hippocampal synaptic plasticity. We have previously shown that predominantly in the female hippocampus, does inhibition of the conversion of testosterone to estradiol affect synaptic transmission. In this study, we show that testosterone and its metabolite dihydrotestosterone are essential for hippocampal synaptic transmission specifically in males. This also holds true for the density of mushroom spines and of spine synapses. We obtained similar sex-dependent results using primary hippocampal cultures of male and female animals. Since these cultures originated from perinatal animals, our findings argue for sex-dependent differentiation of hippocampal neurons regarding their responsiveness to sex neurosteroids up to birth, which persist during adulthood. Hence, our in vitro findings may point to a developmental effect either directly induced by sex chromosomes or indirectly by fetal testosterone secretion during the perinatal critical period, when developmental sexual priming takes place.


Assuntos
Hipocampo/metabolismo , Plasticidade Neuronal/fisiologia , Neuroesteroides/metabolismo , Caracteres Sexuais , Sinapses/metabolismo , Animais , Células Cultivadas , Feminino , Hipocampo/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Sinapses/ultraestrutura
12.
Cereb Cortex ; 30(3): 1688-1707, 2020 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-31667489

RESUMO

Reelin is an extracellular matrix protein, known for its dual role in neuronal migration during brain development and in synaptic plasticity at adult stages. During the perinatal phase, Reelin expression switches from Cajal-Retzius (CR) cells, its main source before birth, to inhibitory interneurons (IN), the main source of Reelin in the adult forebrain. IN-derived Reelin has been associated with schizophrenia and temporal lobe epilepsy; however, the functional role of Reelin from INs is presently unclear. In this study, we used conditional knockout mice, which lack Reelin expression specifically in inhibitory INs, leading to a substantial reduction in total Reelin expression in the neocortex and dentate gyrus. Our results show that IN-specific Reelin knockout mice exhibit normal neuronal layering and normal behavior, including spatial reference memory. Although INs are the major source of Reelin within the adult stem cell niche, Reelin from INs does not contribute substantially to normal adult neurogenesis. While a closer look at the dentate gyrus revealed some unexpected alterations at the cellular level, including an increase in the number of Reelin expressing CR cells, overall our data suggest that Reelin derived from INs is less critical for cortex development and function than Reelin expressed by CR cells.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Giro Denteado/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Interneurônios/metabolismo , Neocórtex/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Serina Endopeptidases/metabolismo , Animais , Comportamento Animal/fisiologia , Movimento Celular/fisiologia , Giro Denteado/fisiopatologia , Hipocampo/metabolismo , Interneurônios/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/fisiologia , Neurônios/metabolismo , Folhas de Planta/metabolismo , Proteína Reelina
13.
J Am Soc Nephrol ; 30(5): 824-839, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30971456

RESUMO

BACKGROUND: About 3%-5% of adults with membranous nephropathy have autoantibodies directed against thrombospondin type 1 domain-containing 7A (THSD7A), a podocyte-expressed transmembrane protein. However, the temporal and spatial expression of THSD7A and its biologic function for podocytes are unknown, information that is needed to understand the effects of THSD7A autoantibodies in this disease. METHODS: Using a variety of microscopic techniques, we analyzed THSD7A localization in postnatal, adult, and autoantibody-injected mice as well as in human podocytes. We also analyzed THSD7A function in human podocytes using confocal microscopy; Western blotting; and adhesion and migration assays. RESULTS: We found that THSD7A expression begins on glomerular vascularization with slit diaphragm formation in development. THSD7A localizes to the basal aspect of foot processes, closely following the meanders of the slit diaphragm in human and mice. Autoantibodies binding to THSD7A localize to the slit diaphragm. In human podocytes, THSD7A expression is accentuated at filopodia and thin arborized protrusions, an expression pattern associated with decreased membrane activity of cytoskeletal regulators. We also found that, phenotypically, THSD7A expression in human podocytes is associated not only with increases in cell size, enhanced adhesion, and reduced detachment from collagen type IV-coated plates but also, with decreased ability to migrate. CONCLUSIONS: Our findings suggest that THSD7A functions as a foot process protein involved in the stabilization of the slit diaphragm of mature podocytes and that autoantibodies to THSD7A, on the basis of their localization, might structurally and functionally alter the slit diaphragm's permeability to protein.


Assuntos
Antígenos de Superfície/genética , Glomerulonefrite Membranosa/genética , Glomérulos Renais/metabolismo , Proteínas de Membrana/metabolismo , Trombospondinas/imunologia , Animais , Antígenos de Superfície/imunologia , Autoanticorpos/imunologia , Western Blotting , Células Cultivadas , Regulação da Expressão Gênica , Taxa de Filtração Glomerular , Glomerulonefrite Membranosa/fisiopatologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Podócitos/imunologia , Proteinúria/metabolismo , Sensibilidade e Especificidade , Trombospondinas/metabolismo
14.
J Neurochem ; 150(2): 173-187, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30790293

RESUMO

Clinical and animal studies have revealed sex-specific differences in histopathological and neurological outcome after traumatic brain injury (TBI). The impact of perioperative administration of sex steroid inhibitors on TBI is still elusive. Here, we subjected male and female C57Bl/6N mice to the controlled cortical impact (CCI) model of TBI and applied pharmacological inhibitors of steroid hormone synthesis, that is, letrozole (LET, inhibiting estradiol synthesis by aromatase) and finasteride (FIN, inhibiting dihydrotestosterone synthesis by 5α-reductase), respectively, starting 72 h prior CCI, and continuing for a further 48 h after CCI. Initial gene expression analyses showed that androgen (Ar) and estrogen receptors (Esr1) were sex-specifically altered 72 h after CCI. When examining brain lesion size, we found larger lesions in male than in female mice, but did not observe effects of FIN or LET treatment. However, LET treatment exacerbated neurological deficits 24 and 72 h after CCI. On the molecular level, FIN administration reduced calpain-dependent spectrin breakdown products, a proxy of excitotoxicity and disturbed Ca2+ homeostasis, specifically in males, whereas LET increased the reactive astrocyte marker glial fibrillary acid protein specifically in females. Examination of neurotrophins (brain-derived neurotrophic factor, neuronal growth factor, NT-3) and their receptors (p75NTR , TrkA, TrkB, TrkC) revealed CCI-induced down-regulation of TrkB and TrkC protein expression, which was reduced by LET in both sexes. Interestingly, FIN decreased neuronal growth factor mRNA expression and protein levels of its receptor TrkA only in males. Taken together, our data suggest a sex-specific impact on pathogenic processes in the injured brain after TBI. Sex hormones may thus modulate pathogenic processes in experimental TBI.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Encéfalo/efeitos dos fármacos , Di-Hidrotestosterona/antagonistas & inibidores , Estradiol/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Antagonistas de Estrogênios/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/efeitos dos fármacos , Caracteres Sexuais
15.
Hippocampus ; 29(6): 550-565, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30394609

RESUMO

Cajal-Retzius (CR) cells are early-born glutamatergic neurons that are primarily known as the early main source of the signal protein Reelin. In the reeler mutant, the absence of Reelin causes severe defects in the radial migration of neurons, resulting in abnormal cortical layering. To date, the exact morphological properties of CR-cells independent of Reelin are unknown. With this in view, we studied the ontogenesis, density, and distribution of CR-cells in reeler mice that were cross-bred with a CXCR4-EGFP reporter mouse line, thus enabling us to clearly identify CR-cells positions in the disorganized hippocampus of the reeler mouse. As evidenced by morphological analysis, differences were found regarding CR-cell distribution and density: generally, we found fewer CR-cells in the developing and adult reeler hippocampus as compared to the hippocampus of wild-type animals (WT); however, in reeler mice, CR-cells were much more closely associated to the hippocampal fissure (HF), resulting in relatively higher local CR-cell densities. This higher local cell density was accompanied by stronger immunoreactivity of the CXCR4 ligand, stroma-derived factor-1 (SDF-1) that is known to regulate CR-cell positioning. Importantly, confocal microscopy indicates an integration of CR-cells into the developing and adult hippocampal network in reeler mice, raising evidence that network integration of CR-cells might be independent of Reelin.


Assuntos
Moléculas de Adesão Celular Neuronais/deficiência , Moléculas de Adesão Celular Neuronais/genética , Proteínas da Matriz Extracelular/deficiência , Proteínas da Matriz Extracelular/genética , Hipocampo/patologia , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Serina Endopeptidases/deficiência , Serina Endopeptidases/genética , Animais , Contagem de Células , Movimento Celular , Quimiocina CXCL12/metabolismo , Giro Denteado/metabolismo , Giro Denteado/patologia , Ácido Glutâmico/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Camundongos , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Microscopia Confocal , Rede Nervosa/metabolismo , Rede Nervosa/patologia , Neurogênese , Neurônios/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteína Reelina , Transdução de Sinais
16.
eNeuro ; 5(5)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30406178

RESUMO

HCN1 compartmentalization in CA1 pyramidal cells, essential for hippocampal information processing, is believed to be controlled by the extracellular matrix protein Reelin. Expression of Reelin, in turn, is stimulated by 17ß-estradiol (E2). In this study, we therefore tested whether E2 regulates the compartmentalization of HCN1 in CA1 via Reelin. In organotypic entorhino-hippocampal cultures, we found that E2 promotes HCN1 distal dendritic enrichment via the G protein-coupled estrogen receptor GPER1, but apparently independent of Reelin, because GST-RAP, known to reduce Reelin signaling, did not prevent E2-induced HCN1 enrichment in distal CA1. We therefore re-examined the role of Reelin for the regulation of HCN1 compartmentalization and could not detect effects of reduced Reelin signaling on HCN1 distribution in CA1, either in the (developmental) slice culture model or in tamoxifen-inducible conditional reelin knockout mice during adulthood. We conclude that for HCN1 channel compartmentalization in CA1 pyramidal cells, Reelin is not as essential as previously proposed, and E2 effects on HCN1 distribution in CA1 are mediated by mechanisms that do not involve Reelin. Because HCN1 localization was not altered at different phases of the estrous cycle, gonadally derived estradiol is unlikely to regulate HCN1 channel compartmentalization, while the pattern of immunoreactivity of aromatase, the final enzyme of estradiol synthesis, argues for a role of local hippocampal E2 synthesis.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Dendritos/efeitos dos fármacos , Estrogênios/farmacologia , Proteínas da Matriz Extracelular/metabolismo , Hipocampo/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Serina Endopeptidases/metabolismo , Animais , Dendritos/metabolismo , Estradiol/metabolismo , Estradiol/farmacologia , Estrogênios/metabolismo , Hipocampo/metabolismo , Neurônios/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Células Piramidais/metabolismo , Ratos Wistar , Proteína Reelina
17.
Psychopharmacology (Berl) ; 235(12): 3465-3477, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30306229

RESUMO

17-Beta-estradiol (E2) stimulates neural plasticity and dopaminergic transmission in the prefrontal cortex, which is critically involved in attentional control, working memory, and other executive functions. Studies investigating E2's actions on prefrontally mediated behavior in the course of the menstrual cycle or during hormone replacement therapy are inconclusive, with numerous null findings as well as beneficial and detrimental effects. The current study focused on the effect of E2 on attentional performance, as animal studies indicate that supraphysiological doses (i.e., above estrous cycle levels) of E2 have beneficial effects on measures of attention in female rodents. To translate these findings to humans, we administered 12 mg E2-valerate or placebo orally to 34 naturally cycling women in the low-hormone early follicular phase using a randomized, double-blinded, pre-post design. Behavioral performance was tested twice during baseline and E2 peak, where E2 levels reached mildly supraphysiological levels in the E2 group. Aside from mainly prefrontally mediated tasks of attention, working memory, and other executive functions, we employed tasks of affectively modulated attention, emotion recognition, and verbal memory. E2 administration had a significant, but subtle negative impact on general processing speed and working memory performance. These effects could be related to an overstimulation of dopaminergic transmission. The negative effect of supraphysiological E2 on working memory connects well to animal literature. There were no effects on attentional performance or any other measure. This could be explained by different E2 levels being optimal for changing behavioral performance in specific tasks, which likely depends on the brain regions involved.


Assuntos
Estradiol/administração & dosagem , Estrogênios/administração & dosagem , Memória de Curto Prazo/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Administração Oral , Adulto , Animais , Método Duplo-Cego , Emoções/efeitos dos fármacos , Emoções/fisiologia , Ciclo Estral/efeitos dos fármacos , Ciclo Estral/fisiologia , Função Executiva/efeitos dos fármacos , Função Executiva/fisiologia , Feminino , Humanos , Memória de Curto Prazo/fisiologia , Ciclo Menstrual/efeitos dos fármacos , Ciclo Menstrual/fisiologia , Ciclo Menstrual/psicologia , Córtex Pré-Frontal/fisiologia , Desempenho Psicomotor/fisiologia , Adulto Jovem
18.
Sci Rep ; 8(1): 8722, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29880879

RESUMO

Reelin plays an important role in cerebral cortex development and synaptogenesis. In the hippocampus, the neurosteroid estrogen affects reelin expression. In this study we tested a potential crosstalk between estradiol and reelin, thus the possibility of a reelin-induced activation of the estradiol synthesizing enzyme aromatase. As a model system, we used ovaries, which express reelin and are a major source of estradiol. We found that in wild-type mice, reelin and aromatase are expressed in granulosa cells of growing follicles. The expression of reelin varies with the estrus cycle and is highest shortly before ovulation, when estradiol serum levels are at their maximum. In ovaries of reelin-deficient reeler mice, aromatase mRNA and protein are significantly reduced, as evidenced by real-time PCR, western blot analysis, and quantitative immunohistochemistry in granulosa cells of preovulatory follicles. In line with reduced estradiol synthesis, ovarian estrus cycle length is prolonged in reeler mice. Most importantly, treating cultured granulosa cells with recombinant reelin results in significant upregulation of aromatase mRNA and protein and increased secretion of estradiol into the supernatant. Our data provide evidence of a local increase of aromatase expression by reelin. Regarding reproduction, this crosstalk may contribute to follicular stability and counteract luteinization in ovaries.


Assuntos
Aromatase/biossíntese , Moléculas de Adesão Celular Neuronais/biossíntese , Ciclo Estral/fisiologia , Proteínas da Matriz Extracelular/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Células da Granulosa/metabolismo , Luteinização/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Serina Endopeptidases/biossíntese , Animais , Feminino , Células da Granulosa/citologia , Camundongos , Ratos Wistar , Proteína Reelina
19.
Brain Pathol ; 28(1): 14-27, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27880990

RESUMO

Nowadays, amyotrophic lateral sclerosis (ALS) is considered as a multisystem disorder, characterized by a primary degeneration of motor neurons as well as neuropathological changes in non-motor regions. Neurodegeneration in subcortical areas, such as the thalamus, are believed to contribute to cognitive and behavioral abnormalities in ALS patients. In the present study, we investigated neurodegenerative changes including neuronal loss and glia pathology in the anterodorsal thalamic nucleus (AD) of SOD1(G93A) mice, a widely used animal model for ALS. We detected massive dendrite swelling and neuronal loss in SOD1(G93A) animals, which was accompanied by a mild gliosis. Furthermore, misfolded SOD1 protein and autophagy markers were accumulating in the AD. Since innate immunity and activation inflammasomes seem to play a crucial role in ALS, we examined protein expression of Nod-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase-1 recruitment domain (ASC) and the cytokine interleukin 1 beta (IL1ß) in AD glial cells and neurons. NLRP3 and ASC were significantly up-regulated in the AD of SOD1(G93A) mice. Finally, co-localization studies revealed expression of NLRP3, ASC and IL1ß in neurons. Our study yielded two main findings: (i) neurodegenerative changes already occur at an early symptomatic stage in the AD and (ii) increased inflammasome expression may contribute to neuronal cell death. In conclusion, neurodegeneration in the anterior thalamus may critically account for cognitive changes in ALS pathology.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Núcleos Anteriores do Tálamo/patologia , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Degeneração Neural/patologia , Neurônios/patologia , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Núcleos Anteriores do Tálamo/fisiopatologia , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Morte Celular/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Interleucina-1beta/metabolismo , Masculino , Camundongos Transgênicos , Degeneração Neural/fisiopatologia , Neuroglia/patologia , Neuroglia/fisiologia , Neurônios/fisiologia , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...