Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 15: 1353151, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38348415

RESUMO

Reproduction in mammals is controlled by hypothalamic gonadotropin-releasing hormone (GnRH) neurons. Recent studies from our laboratory established that the basal ganglia of the human brain contain additional large populations of GnRH synthesizing neurons which are absent in adult mice. Such extrahypothalamic GnRH neurons mostly occur in the putamen where they correspond to subsets of the striatal cholinergic interneurons (ChINs) and express GnRHR autoreceptors. In an effort to establish a mouse model for functional studies of striatal GnRH/GnRHR signaling, we carried out electrophysiological experiments on acute brain slices from male transgenic mice. Using PN4-7 neonatal mice, half of striatal ChINs responded with transient hyperpolarization and decreased firing rate to 1.2 µM GnRH, whereas medium spiny projection neurons remained unaffected. GnRH acted on its specific receptor because no response was observed in the presence of the GnRHR antagonist Antide. Addition of the membrane-impermeable G protein-coupled receptor inhibitor GDP-ß-S to the internal electrode solution eliminated the effect of GnRH. Further, GnRH was able to inhibit ChINs in presence of tetrodotoxin which blocked action potential mediated events. Collectively, these data indicated that the receptor underlying the effects of GnRH in neonatal mice is localized within ChINs. GnRH responsiveness of ChINs was transient and entirely disappeared in adult mice. These results raise the possibility to use neonatal transgenic mice as a functional model to investigate the role of GnRH/GnRHR signaling discovered earlier in adult human ChINs.


Assuntos
Hormônio Liberador de Gonadotropina , Receptores LHRH , Animais , Masculino , Camundongos , Neurônios Colinérgicos , Hormônio Liberador de Gonadotropina/farmacologia , Mamíferos , Camundongos Transgênicos , Transdução de Sinais
2.
J Biol Chem ; 299(9): 105121, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37536628

RESUMO

Single-cell transcriptomics are powerful tools to define neuronal cell types based on co-expressed gene clusters. Limited RNA input in these technologies necessarily compromises transcriptome coverage and accuracy of differential expression analysis. We propose that bulk RNA-Seq of neuronal pools defined by spatial position offers an alternative strategy to overcome these technical limitations. We report a laser-capture microdissection (LCM)-Seq method which allows deep transcriptome profiling of fluorescently tagged neuron populations isolated with LCM from histological sections of transgenic mice. Mild formaldehyde fixation of ZsGreen marker protein, LCM sampling of ∼300 pooled neurons, followed by RNA isolation, library preparation and RNA-Seq with methods optimized for nanogram amounts of moderately degraded RNA enabled us to detect ∼15,000 different transcripts in fluorescently labeled cholinergic neuron populations. The LCM-Seq approach showed excellent accuracy in quantitative studies, allowing us to detect 2891 transcripts expressed differentially between the spatially defined and clinically relevant cholinergic neuron populations of the dorsal caudate-putamen and medial septum. In summary, the LCM-Seq method we report in this study is a versatile, sensitive, and accurate bulk sequencing approach to study the transcriptome profile and differential gene expression of fluorescently tagged neuronal populations isolated from transgenic mice with high spatial precision.

3.
Front Endocrinol (Lausanne) ; 13: 960769, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36093104

RESUMO

Kisspeptin neurons residing in the rostral periventricular area of the third ventricle (KPRP3V) and the arcuate nucleus (KPARC) mediate positive and negative estrogen feedback, respectively. Here, we aim to compare transcriptional responses of KPRP3V and KPARC neurons to estrogen. Transgenic mice were ovariectomized and supplemented with either 17ß-estradiol (E2) or vehicle. Fluorescently tagged KPRP3V neurons collected by laser-capture microdissection were subjected to RNA-seq. Bioinformatics identified 222 E2-dependent genes. Four genes encoding neuropeptide precursors (Nmb, Kiss1, Nts, Penk) were robustly, and Cartpt was subsignificantly upregulated, suggesting putative contribution of multiple neuropeptides to estrogen feedback mechanisms. Using overrepresentation analysis, the most affected KEGG pathways were neuroactive ligand-receptor interaction and dopaminergic synapse. Next, we re-analyzed our previously obtained KPARC neuron RNA-seq data from the same animals using identical bioinformatic criteria. The identified 1583 E2-induced changes included suppression of many neuropeptide precursors, granins, protein processing enzymes, and other genes related to the secretory pathway. In addition to distinct regulatory responses, KPRP3V and KPARC neurons exhibited sixty-two common changes in genes encoding three hormone receptors (Ghsr, Pgr, Npr2), GAD-65 (Gad2), calmodulin and its regulator (Calm1, Pcp4), among others. Thirty-four oppositely regulated genes (Kiss1, Vgf, Chrna7, Tmem35a) were also identified. The strikingly different transcriptional responses in the two neuron populations prompted us to explore the transcriptional mechanism further. We identified ten E2-dependent transcription factors in KPRP3V and seventy in KPARC neurons. While none of the ten transcription factors interacted with estrogen receptor-α, eight of the seventy did. We propose that an intricate, multi-layered transcriptional mechanism exists in KPARC neurons and a less complex one in KPRP3V neurons. These results shed new light on the complexity of estrogen-dependent regulatory mechanisms acting in the two functionally distinct kisspeptin neuron populations and implicate additional neuropeptides and mechanisms in estrogen feedback.


Assuntos
Núcleo Arqueado do Hipotálamo , Kisspeptinas , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Estrogênios/metabolismo , Estrogênios/farmacologia , Kisspeptinas/genética , Kisspeptinas/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Fatores de Transcrição/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(27): e2113749119, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35763574

RESUMO

Kisspeptin neurons in the mediobasal hypothalamus (MBH) are critical targets of ovarian estrogen feedback regulating mammalian fertility. To reveal molecular mechanisms underlying this signaling, we thoroughly characterized the estrogen-regulated transcriptome of kisspeptin cells from ovariectomized transgenic mice substituted with 17ß-estradiol or vehicle. MBH kisspeptin neurons were harvested using laser-capture microdissection, pooled, and subjected to RNA sequencing. Estrogen treatment significantly (p.adj. < 0.05) up-regulated 1,190 and down-regulated 1,139 transcripts, including transcription factors, neuropeptides, ribosomal and mitochondrial proteins, ion channels, transporters, receptors, and regulatory RNAs. Reduced expression of the excitatory serotonin receptor-4 transcript (Htr4) diminished kisspeptin neuron responsiveness to serotonergic stimulation. Many estrogen-regulated transcripts have been implicated in puberty/fertility disorders. Patients (n = 337) with congenital hypogonadotropic hypogonadism (CHH) showed enrichment of rare variants in putative CHH-candidate genes (e.g., LRP1B, CACNA1G, FNDC3A). Comprehensive characterization of the estrogen-dependent kisspeptin neuron transcriptome sheds light on the molecular mechanisms of ovary-brain communication and informs genetic research on human fertility disorders.


Assuntos
Núcleo Arqueado do Hipotálamo , Estrogênios , Fertilidade , Kisspeptinas , Neurônios , Ovário , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Estrogênios/metabolismo , Feminino , Fertilidade/genética , Perfilação da Expressão Gênica , Humanos , Hipogonadismo/congênito , Hipogonadismo/genética , Kisspeptinas/genética , Kisspeptinas/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Ovário/metabolismo
5.
Elife ; 102021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34128468

RESUMO

Human reproduction is controlled by ~2000 hypothalamic gonadotropin-releasing hormone (GnRH) neurons. Here, we report the discovery and characterization of additional ~150,000-200,000 GnRH-synthesizing cells in the human basal ganglia and basal forebrain. Nearly all extrahypothalamic GnRH neurons expressed the cholinergic marker enzyme choline acetyltransferase. Similarly, hypothalamic GnRH neurons were also cholinergic both in embryonic and adult human brains. Whole-transcriptome analysis of cholinergic interneurons and medium spiny projection neurons laser-microdissected from the human putamen showed selective expression of GNRH1 and GNRHR1 autoreceptors in the cholinergic cell population and uncovered the detailed transcriptome profile and molecular connectome of these two cell types. Higher-order non-reproductive functions regulated by GnRH under physiological conditions in the human basal ganglia and basal forebrain require clarification. The role and changes of GnRH/GnRHR1 signaling in neurodegenerative disorders affecting cholinergic neurocircuitries, including Parkinson's and Alzheimer's diseases, need to be explored.


Assuntos
Gânglios da Base , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios , Adulto , Prosencéfalo Basal/citologia , Gânglios da Base/citologia , Gânglios da Base/metabolismo , Gânglios da Base/fisiologia , Células Cultivadas , Colina O-Acetiltransferase , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/citologia , Neurônios/metabolismo , Neurônios/fisiologia , Putamen/citologia , Transcriptoma
6.
J Steroid Biochem Mol Biol ; 188: 185-194, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30685384

RESUMO

Gonadal hormones including 17ß-estradiol exert important protective functions in skeletal homeostasis. However, numerous details of ovarian hormone deficiency in the common bone marrow microenvironment have not yet been revealed and little information is available on the tissue-specific acts either, especially those via estrogen receptor beta (ERß). The aim of the present study was therefore to examine the bone-related gene expression changes after ovariectomy (OVX) and long-term ERß agonist diarylpropionitrile (DPN) administration. We found that OVX produced strong and widespread changes of gene expression in both femoral bone and bone marrow. In the bone out of 22 genes, 20 genes were up- and 2 were downregulated after OVX. It is noteworthy that DPN restored mRNA expression of 10 OVX-induced changes (Aldh2, Col1a1, Daam1, Fgf12, Igf1, Il6r, Nfkb1, Notch1, Notch2 and Psen1) suggesting a modulatory role of ERß in bone physiology. In bone marrow, out of 37 categorized genes, transcription of 25 genes were up- and 12 were downregulated indicating that the marrow is highly responsive to gonadal hormones. DPN modestly affected transcription, only expression of two genes (Nfatc1 and Tgfb1) was restored by DPN action. The PI3K/Akt signaling pathway was the most affected gene cluster following the interventions in bone and bone marrow, as demonstrated by canonical variates analysis (CVA). We suggested that our results contribute to a deeper understanding of alterations in gene expression of bone and bone marrow niche elicited by ERß and selective ERß analogs.


Assuntos
Medula Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Receptor beta de Estrogênio/agonistas , Nitrilas/farmacologia , Propionatos/farmacologia , Transcriptoma/efeitos dos fármacos , Animais , Medula Óssea/metabolismo , Osso e Ossos/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Ovariectomia , Ratos , Ratos Wistar
7.
Neuroscience ; 405: 35-46, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29522854

RESUMO

Microglia are instrumental for recognition and elimination of amyloid ß1-42 oligomers (AßOs), but the long-term consequences of AßO-induced inflammatory changes in the brain are unclear. Here, we explored microglial responses and transciptome-level inflammatory signatures in the rat hippocampus after chronic AßO challenge. Middle-aged Long Evans rats received intracerebroventricular infusion of AßO or vehicle for 4 weeks, followed by treatment with artificial CSF or MCC950 for the subsequent 4 weeks. AßO infusion evoked a sustained inflammatory response including activation of NF-κB, triggered microglia activation and increased the expression of pattern recognition and phagocytic receptors. Aß1-42 plaques were not detectable likely due to microglial elimination of infused oligomers. In addition, we found upregulation of neuronal inhibitory ligands and their cognate microglial receptors, while downregulation of Esr1 and Scn1a, encoding estrogen receptor alpha and voltage-gated sodium-channel Na(v)1.1, respectively, was observed. These changes were associated with impaired hippocampus-dependent spatial memory and resembled early neurological changes seen in Alzheimer's disease. To investigate the role of inflammatory actions in memory deterioration, we performed MCC950 infusion, which specifically blocks the NLRP3 inflammasome. MCC950 attenuated AßO-evoked microglia reactivity, restored expression of neuronal inhibitory ligands, reversed downregulation of ERα, and abolished memory impairments. Furthermore, MCC950 abrogated AßO-invoked reduction of serum IL-10. These findings provide evidence that in response to AßO infusion microglia change their phenotype, but the resulting inflammatory changes are sustained for at least one month after the end of AßO challenge. Lasting NLRP3-driven inflammatory alterations and altered hippocampal gene expression contribute to spatial memory decline.


Assuntos
Peptídeos beta-Amiloides/administração & dosagem , Hipocampo/efeitos dos fármacos , Microglia/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Peptídeos beta-Amiloides/toxicidade , Animais , Comunicação Celular/efeitos dos fármacos , Citocinas/sangue , Citocinas/metabolismo , Furanos , Compostos Heterocíclicos de 4 ou mais Anéis , Hipocampo/metabolismo , Hipocampo/patologia , Indenos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Infusões Intraventriculares , Masculino , Aprendizagem em Labirinto , Microglia/metabolismo , Microglia/patologia , Modelos Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/toxicidade , Ratos , Ratos Long-Evans , Memória Espacial/efeitos dos fármacos , Sulfonamidas , Sulfonas
8.
Endocrinology ; 158(1): 69-83, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27805868

RESUMO

Ovarian hormones regulate the transcriptome of the hippocampus and modulate its functions. During menopause this complex signaling declines, leading to impaired learning and memory. This study was undertaken to clarify the effects of long-term, surgical ovariectomy (OVX) on the rat hippocampal transcriptome. At age of 13 months, intact control and ovariectomized groups were formed. All animals were killed 5 weeks after gonadectomy; hippocampal formations were dissected and processed for transcriptome analysis. Microarray and polymerase chain reaction studies identified 252 and 61 genes, respectively, whose expression was altered in the lack of ovarian hormones. Pathway analysis revealed impact on neuroactive ligand-receptor interaction, endocannabinoid, and estrogen signaling, among others. Network and interaction analyses of proteins encoded by OVX-regulated genes revealed upregulation of growth/troph/transcription factor signaling assembly (Mdk, Fgf1, Igf2, Ngf, Ngfr, Ntf3, Ntrk1, Otx2, Hif1a, Esr1, Nr4a3), peptides/peptide receptors (Cartpt, Kl, Ttr, Gnrhr), neurotransmission (Grm1, Gria4, Gls, Slc18a2, Kcnj6), and genes serving immune functions (C3, Ccl2, Itgam, Il1b). Downregulated clusters included neuropeptides and their receptors (Adcyap1, Cbln2, Cck, Cckbr, Crhr1 and 2, Oprd1, Nts, Penk, Sstr1, Vip), neurotransmitter signaling (Htr2c, Chrna3, Chrm4, Grm8, Hrh3, Slc17a6), and potassium channels (Kcnk9, Kcnj9, Kcnma1, Kcnc2). Several transcription factors (Rxra, Thrb), solute carriers and defense molecules (Apitd1, Bcl2, C1ql3, Ilr3a, Sod1, Sncb) also underwent downregulation. The findings indicate that surgical gonadectomy carried out at middle-age robustly changes the hippocampal transcriptome that alters neurogenesis, synaptic plasticity, immune modulation, causing cognitive dysfunctions.


Assuntos
Hipocampo/metabolismo , Menopausa/metabolismo , Transcriptoma , Animais , Feminino , Perfilação da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos , Ovariectomia , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
9.
Front Cell Neurosci ; 10: 149, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27375434

RESUMO

Estradiol (E2) robustly activates transcription of a broad array of genes in the hippocampal formation of middle-aged ovariectomized rats via estrogen receptors (ERα, ERß, and G protein-coupled ER). Selective ERß agonists also influence hippocampal functions, although their downstream molecular targets and mechanisms are not known. In this study, we explored the effects of long-term treatment with ERß agonist diarylpropionitrile (DPN, 0.05 mg/kg/day, sc.) on the hippocampal transcriptome in ovariectomized, middle-aged (13 month) rats. Isolated hippocampal formations were analyzed by Affymetrix oligonucleotide microarray and quantitative real-time PCR. Four hundred ninety-seven genes fulfilled the absolute fold change higher than 2 (FC > 2) selection criterion. Among them 370 genes were activated. Pathway analysis identified terms including glutamatergic and cholinergic synapse, RNA transport, endocytosis, thyroid hormone signaling, RNA degradation, retrograde endocannabinoid signaling, and mRNA surveillance. PCR studies showed transcriptional regulation of 58 genes encoding growth factors (Igf2, Igfb2, Igf1r, Fgf1, Mdk, Ntf3, Bdnf), transcription factors (Otx2, Msx1), potassium channels (Kcne2), neuropeptides (Cck, Pdyn), peptide receptors (Crhr2, Oprm1, Gnrhr, Galr2, Sstr1, Sstr3), neurotransmitter receptors (Htr1a, Htr2c, Htr2a, Gria2, Gria3, Grm5, Gabra1, Chrm5, Adrb1), and vesicular neurotransmitter transporters (Slc32a1, Slc17a7). Protein-protein interaction analysis revealed networking of clusters associated with the regulation of growth/troph factor signaling, transcription, translation, neurotransmitter and neurohormone signaling mechanisms and potassium channels. Collectively, the results reveal the contribution of ERß-mediated processes to the regulation of transcription, translation, neurogenesis, neuromodulation, and neuroprotection in the hippocampal formation of ovariectomized, middle-aged rats and elucidate regulatory channels responsible for DPN-altered functional patterns. These findings support the notion that selective activation of ERß may be a viable approach for treating the neural symptoms of E2 deficiency in menopause.

10.
Neuroendocrinology ; 103(3-4): 369-82, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26338351

RESUMO

Neuropeptides of the hypothalamic arcuate nucleus (ARC) regulate important homeostatic and endocrine functions and also play critical roles in pubertal development. The altered peptidergic and aminoacidergic neurotransmission accompanying pubertal maturation of the ARC is not fully understood. Here we studied the developmental shift in the gene expression profile of the ARC of male mice. RNA samples for quantitative RT-PCR studies were isolated from the ARC of 14-day-old infantile and 60-day-old adult male mice with laser capture microdissection. The expression of 18 neuropeptide, 15 neuropeptide receptor, 4 sex steroid receptor and 6 classic neurotransmitter marker mRNAs was compared between the two time points. The adult animals showed increased mRNA levels encoding cocaine- and amphetamine-regulated transcripts, galanin-like peptide, dynorphin, kisspeptin, proopiomelanocortin, proenkephalin and galanin and a reduced expression of mRNAs for pituitary adenylate cyclase-activating peptide, calcitonin gene-related peptide, neuropeptide Y, substance P, agouti-related protein, neurotensin and growth hormone-releasing hormone. From the neuropeptide receptors tested, melanocortin receptor-4 showed the most striking increase (5-fold). Melanocortin receptor-3 and the Y1 and Y5 neuropeptide Y receptors increased 1.5- to 1.8-fold, whereas δ-opioid receptor and neurotensin receptor-1 transcripts were reduced by 27 and 21%, respectively. Androgen receptor, progesterone receptor and α-estrogen receptor transcripts increased by 54-72%. The mRNAs of glutamic acid decarboxylases-65 and -67, vesicular GABA transporter and choline acetyltransferase remained unchanged. Tyrosine hydroxylase mRNA increased by 44%, whereas type-2 vesicular glutamate transporter mRNA decreased by 43% by adulthood. Many of the developmental changes we revealed in this study suggest a reduced inhibitory and/or enhanced excitatory neuropeptidergic drive on fertility in adult animals.


Assuntos
Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Fatores Etários , Animais , Masculino , Camundongos , Neuropeptídeos/genética , RNA Mensageiro/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Transmissão Sináptica/genética
11.
Neuroendocrinology ; 102(1-2): 44-59, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25925152

RESUMO

BACKGROUND: Gonadotropin-releasing hormone (GnRH) neurons play a pivotal role in the regulation of the hypothalamic-pituitary gonadal axis in a sex-specific manner. We hypothesized that the differences seen in reproductive functions of males and females are associated with a sexually dimorphic gene expression profile of GnRH neurons. METHODS AND RESULTS: We compared the transcriptome of GnRH neurons obtained from intact metestrous female and male GnRH-green fluorescent protein transgenic mice. About 1,500 individual GnRH neurons from each sex were sampled with laser capture microdissection followed by whole-transcriptome amplification for gene expression profiling. Under stringent selection criteria (fold change >1.6, adjusted p value 0.01), Affymetrix Mouse Genome 430 PM array analysis identified 543 differentially expressed genes. Sexual dimorphism was most apparent in gene clusters associated with synaptic communication, signal transduction, cell adhesion, vesicular transport and cell metabolism. To validate microarray results, 57 genes were selected, and 91% of their differential expression was confirmed by real-time PCR. Similarly, 88% of microarray results were confirmed with PCR from independent samples obtained by patch pipette harvesting and pooling of 30 GnRH neurons from each sex. We found significant differences in the expression of genes involved in vesicle priming and docking (Syt1, Cplx1), GABAergic (Gabra3, Gabrb3, Gabrg2) and glutamatergic (Gria1, Grin1, Slc17a6) neurotransmission, peptide signaling (Sstr3, Npr2, Cxcr4) and the regulation of intracellular ion homeostasis (Cacna1, Cacnb1, Cacng5, Kcnq2, Kcnc1). CONCLUSION: The striking sexual dimorphism of the GnRH neuron transcriptome we report here contributes to a better understanding of the differences in cellular mechanisms of GnRH neurons in the two sexes.


Assuntos
Encéfalo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Caracteres Sexuais , Transcriptoma , Animais , Feminino , Proteínas de Fluorescência Verde , Masculino , Metestro/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
12.
Endocrinology ; 156(7): 2632-45, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25924104

RESUMO

In the hippocampus, estrogens are powerful modulators of neurotransmission, synaptic plasticity and neurogenesis. In women, menopause is associated with increased risk of memory disturbances, which can be attenuated by timely estrogen therapy. In animal models of menopause, 17ß-estradiol (E2) replacement improves hippocampus-dependent spatial memory. Here, we explored the effect of E2 replacement on hippocampal gene expression in a rat menopause model. Middle-aged ovariectomized female rats were treated continuously for 29 days with E2, and then, the hippocampal transcriptome was investigated with Affymetrix expression arrays. Microarray data were analyzed by Bioconductor packages and web-based softwares, and verified with quantitative PCR. At standard fold change selection criterion, 156 genes responded to E2. All alterations but 4 were transcriptional activation. Robust activation (fold change > 10) occurred in the case of transthyretin, klotho, claudin 2, prolactin receptor, ectodin, coagulation factor V, Igf2, Igfbp2, and sodium/sulfate symporter. Classification of the 156 genes revealed major groups, including signaling (35 genes), metabolism (31 genes), extracellular matrix (17 genes), and transcription (16 genes). We selected 33 genes for further studies, and all changes were confirmed by real-time PCR. The results suggest that E2 promotes retinoid, growth factor, homeoprotein, neurohormone, and neurotransmitter signaling, changes metabolism, extracellular matrix composition, and transcription, and induces protective mechanisms via genomic effects. We propose that these mechanisms contribute to effects of E2 on neurogenesis, neural plasticity, and memory functions. Our findings provide further support for the rationale to develop safe estrogen receptor ligands for the maintenance of cognitive performance in postmenopausal women.


Assuntos
Estradiol/farmacologia , Terapia de Reposição de Estrogênios , Estrogênios/farmacologia , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Menopausa/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Animais , Proteínas de Transporte de Cátions/efeitos dos fármacos , Proteínas de Transporte de Cátions/genética , Claudinas/efeitos dos fármacos , Claudinas/genética , Fator V/efeitos dos fármacos , Fator V/genética , Feminino , Glucuronidase/efeitos dos fármacos , Glucuronidase/genética , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/efeitos dos fármacos , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Fator de Crescimento Insulin-Like II/efeitos dos fármacos , Fator de Crescimento Insulin-Like II/genética , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Klotho , Modelos Animais , Pré-Albumina/efeitos dos fármacos , Pré-Albumina/genética , Proteínas/efeitos dos fármacos , Proteínas/genética , RNA Mensageiro/metabolismo , Ratos , Receptores da Prolactina/efeitos dos fármacos , Receptores da Prolactina/genética , Cotransportador de Sódio-Sulfato , Simportadores/efeitos dos fármacos , Simportadores/genética
13.
Brain Res ; 1583: 1-11, 2014 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-24976584

RESUMO

The mesocortical dopaminergic pathway projecting from the ventral tegmental area (VTA) to the prefrontal cortex (PFC) contributes to the processing of reward signals. This pathway is regulated by gonadal steroids including estradiol. To address the putative role of estradiol and isotype-selective estrogen receptor (ER) agonists in the regulation of the rodent mesocortical system, we combined fMRI, HPLC-MS and qRT-PCR techniques. In fMRI experiments adult, chronically ovariectomized rats, treated with either vehicle, estradiol, ERα agonist 16α-lactone-estradiol (LE2) or ERß agonist diarylpropionitrile (DPN), received a single dose of d-amphetamine-sulphate (10mg/kg, i.p.) and BOLD responses were monitored in the VTA and the PFC. Ovariectomized rats showed no significant response to amphetamine. In contrast, the VTA of ER agonist-substituted ovariectomized rats showed robust amphetamine-evoked BOLD increases. The PFC of estradiol-replaced animals was also responsive to amphetamine. Mass spectroscopic analysis of dopamine and its metabolites revealed a two-fold increase in both dopamine and 3,4-dihydroxyphenylacetic acid content of the PFC in estradiol-replaced animals compared to ovariectomized controls. qRT-PCR studies revealed upregulation of dopamine transporter and dopamine receptor in the VTA and PFC, respectively, of ER agonist-treated ovariectomized animals. Collectively, the results indicate that E2 and isotype-selective ER agonists can powerfully modulate the responsiveness of the mesocortical dopaminergic system, increase the expression of key genes related to dopaminergic neurotransmission and augment the dopamine content of the PFC. In a broader sense, the findings support the concept that the manifestation of reward signals in the PFC is dependent on the actual estrogen milieu of the brain.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Dextroanfetamina/farmacologia , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/metabolismo , Feminino , Lactonas/farmacologia , Nitrilas/farmacologia , Ovariectomia , Oxigênio/sangue , Córtex Pré-Frontal/fisiologia , Propionatos/farmacologia , Ratos Wistar , Receptores Dopaminérgicos/metabolismo , Área Tegmentar Ventral/fisiologia
14.
PLoS One ; 9(5): e97651, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24830778

RESUMO

The orexigenic gut-brain peptide, ghrelin and its G-protein coupled receptor, the growth hormone secretagogue receptor 1a (GHS-R1A) are pivotal regulators of hypothalamic feeding centers and reward processing neuronal circuits of the brain. These systems operate in a cooperative manner and receive a wide array of neuronal hormone/transmitter messages and metabolic signals. Functional magnetic resonance imaging was employed in the current study to map BOLD responses to ghrelin in different brain regions with special reference on homeostatic and hedonic regulatory centers of energy balance. Experimental groups involved male, ovariectomized female and ovariectomized estradiol-replaced rats. Putative modulation of ghrelin signaling by endocannabinoids was also studied. Ghrelin-evoked effects were calculated as mean of the BOLD responses 30 minutes after administration. In the male rat, ghrelin evoked a slowly decreasing BOLD response in all studied regions of interest (ROI) within the limbic system. This effect was antagonized by pretreatment with GHS-R1A antagonist JMV2959. The comparison of ghrelin effects in the presence or absence of JMV2959 in individual ROIs revealed significant changes in the prefrontal cortex, nucleus accumbens of the telencephalon, and also within hypothalamic centers like the lateral hypothalamus, ventromedial nucleus, paraventricular nucleus and suprachiasmatic nucleus. In the female rat, the ghrelin effects were almost identical to those observed in males. Ovariectomy and chronic estradiol replacement had no effect on the BOLD response. Inhibition of the endocannabinoid signaling by rimonabant significantly attenuated the response of the nucleus accumbens and septum. In summary, ghrelin can modulate hypothalamic and mesolimbic structures controlling energy balance in both sexes. The endocannabinoid signaling system contributes to the manifestation of ghrelin's BOLD effect in a region specific manner. In females, the estradiol milieu does not influence the BOLD response to ghrelin.


Assuntos
Encéfalo/patologia , Regulação da Expressão Gênica , Grelina/metabolismo , Homeostase , Imageamento por Ressonância Magnética , Animais , Encéfalo/metabolismo , Estradiol/metabolismo , Comportamento Alimentar , Feminino , Hipotálamo/metabolismo , Hipotálamo/patologia , Sistema Límbico/fisiologia , Masculino , Núcleo Accumbens/patologia , Núcleo Hipotalâmico Paraventricular/patologia , Córtex Pré-Frontal/patologia , Ratos , Ratos Wistar , Receptor CB1 de Canabinoide/metabolismo , Recompensa , Transdução de Sinais , Núcleo Supraquiasmático/patologia , Fatores de Tempo
15.
PLoS One ; 9(2): e88540, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24551115

RESUMO

The innate immune system including microglia has a major contribution to maintenance of the physiological functions of the hippocampus by permanent monitoring of the neural milieu and elimination of tissue-damaging threats. The hippocampus is vulnerable to age-related changes ranging from gene expression to network connectivity. The risk of hippocampal deterioration increases with the decline of gonadal hormone supply. To explore the impact of hormone milieu on the function of the innate immune system in middle-aged female rats, we compared mRNA expression in the hippocampus after gonadal hormone withdrawal, with or without subsequent estrogen replacement using estradiol and isotype-selective estrogen receptor (ER) agonists. Targeted profiling assessed the status of the innate immune system (macrophage-associated receptors, complement, inhibitory neuronal ligands), local estradiol synthesis (P450 aromatase) and estrogen reception (ER). Results established upregulation of macrophage-associated (Cd45, Iba1, Cd68, Cd11b, Cd18, Fcgr1a, Fcgr2b) and complement (C3, factor B, properdin) genes in response to ovariectomy. Ovariectomy upregulated Cd22 and downregulated semaphorin3A (Sema3a) expression, indicating altered neuronal regulation of microglia. Ovariectomy also led to downregulation of aromatase and upregulation of ERα gene. Of note, analogous changes were observed in the hippocampus of postmenopausal women. In ovariectomized rats, estradiol replacement attenuated Iba1, Cd11b, Fcgr1a, C3, increased mannose receptor Mrc1, Cd163 and reversed Sema3a expression. In contrast, reduced expression of aromatase was not reversed by estradiol. While the effects of ERα agonist closely resembled those of estradiol, ERß agonist was also capable of attenuating the expression of several macrophage-associated and complement genes. These data together indicate that the innate immune system of the aging hippocampus is highly responsive to the gonadal hormone milieu. In ovariectomized female rats, estradiol replacement exerts potent immunomodulatory effects including attenuation of microglia sensitization, initiation of M2-like activation and modulation of complement expression by targeting hippocampal neurons and glial cells through ERα and ERß.


Assuntos
Envelhecimento/metabolismo , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Hipocampo/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Ovariectomia , Envelhecimento/genética , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Aromatase/genética , Aromatase/imunologia , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/imunologia , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/imunologia , Terapia de Reposição de Estrogênios , Feminino , Expressão Gênica , Hipocampo/citologia , Hipocampo/imunologia , Humanos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Microglia/citologia , Microglia/efeitos dos fármacos , Pessoa de Meia-Idade , Nitrilas/farmacologia , Pós-Menopausa , Propionatos/farmacologia , Ratos , Receptores de IgG/genética , Receptores de IgG/imunologia , Semaforina-3A/genética , Semaforina-3A/imunologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia
16.
PLoS One ; 8(10): e78178, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24124622

RESUMO

The orexigenic peptide, ghrelin is known to influence function of GnRH neurons, however, the direct effects of the hormone upon these neurons have not been explored, yet. The present study was undertaken to reveal expression of growth hormone secretagogue receptor (GHS-R) in GnRH neurons and elucidate the mechanisms of ghrelin actions upon them. Ca(2+)-imaging revealed a ghrelin-triggered increase of the Ca(2+)-content in GT1-7 neurons kept in a steroid-free medium, which was abolished by GHS-R-antagonist JMV2959 (10 µM) suggesting direct action of ghrelin. Estradiol (1nM) eliminated the ghrelin-evoked rise of Ca(2+)-content, indicating the estradiol dependency of the process. Expression of GHS-R mRNA was then confirmed in GnRH-GFP neurons of transgenic mice by single cell RT-PCR. Firing rate and burst frequency of GnRH-GFP neurons were lower in metestrous than proestrous mice. Ghrelin (40 nM-4 µM) administration resulted in a decreased firing rate and burst frequency of GnRH neurons in metestrous, but not in proestrous mice. Ghrelin also decreased the firing rate of GnRH neurons in males. The ghrelin-evoked alterations of the firing parameters were prevented by JMV2959, supporting the receptor-specific actions of ghrelin on GnRH neurons. In metestrous mice, ghrelin decreased the frequency of GABAergic mPSCs in GnRH neurons. Effects of ghrelin were abolished by the cannabinoid receptor type-1 (CB1) antagonist AM251 (1µM) and the intracellularly applied DAG-lipase inhibitor THL (10 µM), indicating the involvement of retrograde endocannabinoid signaling. These findings demonstrate that ghrelin exerts direct regulatory effects on GnRH neurons via GHS-R, and modulates the firing of GnRH neurons in an ovarian-cycle and endocannabinoid dependent manner.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Endocanabinoides/metabolismo , Ciclo Estral/efeitos dos fármacos , Grelina/farmacologia , Hormônio Liberador de Gonadotropina/biossíntese , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Cálcio/metabolismo , Feminino , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/fisiologia , Expressão Gênica , Masculino , Metestro/efeitos dos fármacos , Camundongos , Proestro/efeitos dos fármacos , RNA Mensageiro/genética , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Potenciais Sinápticos/efeitos dos fármacos
17.
J Neuroinflammation ; 9: 264, 2012 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-23206327

RESUMO

BACKGROUND: The intricate interactions between the immune, endocrine and central nervous systems shape the innate immune response of the brain. We have previously shown that estradiol suppresses expression of immune genes in the frontal cortex of middle-aged ovariectomized rats, but not in young ones reflecting elevated expression of these genes in middle-aged, ovarian hormone deficient animals. Here, we explored the impact of menopause on the microglia phenotype capitalizing on the differential expression of macrophage-associated genes in quiescent and activated microglia. METHODS: We selected twenty-three genes encoding phagocytic and recognition receptors expressed primarily in microglia, and eleven proinflammatory genes and followed their expression in the rat frontal cortex by real-time PCR. We used young, middle-aged and middle-aged ovariectomized rats to reveal age- and ovariectomy-related alterations. We analyzed the expression of the same set of genes in the postcentral and superior frontal gyrus of pre- and postmenopausal women using raw microarray data from our previous study. RESULTS: Ovariectomy caused up-regulation of four classic microglia reactivity marker genes including Cd11b, Cd18, Cd45 and Cd86. The change was reversible since estradiol attenuated transcriptional activation of the four marker genes. Expression of genes encoding phagocytic and toll-like receptors such as Cd11b, Cd18, C3, Cd32, Msr2 and Tlr4 increased, whereas scavenger receptor Cd36 decreased following ovariectomy. Ovarian hormone deprivation altered the expression of major components of estrogen and neuronal inhibitory signaling which are involved in the control of microglia reactivity. Strikingly similar changes took place in the postcentral and superior frontal gyrus of postmenopausal women. CONCLUSIONS: Based on the overlapping results of rat and human studies we propose that the microglia phenotype shifts from the resting toward the reactive state which can be characterized by up-regulation of CD11b, CD14, CD18, CD45, CD74, CD86, TLR4, down-regulation of CD36 and unchanged CD40 expression. As a result of this shift, microglial cells have lower threshold for subsequent activation in the forebrain of postmenopausal women.


Assuntos
Envelhecimento/metabolismo , Antígenos CD/metabolismo , Lobo Frontal/metabolismo , Regulação da Expressão Gênica/fisiologia , Menopausa/metabolismo , Receptor 4 Toll-Like/metabolismo , Adulto , Fatores Etários , Idoso , Animais , Antígenos CD/genética , Citocinas/genética , Citocinas/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Antígenos de Histocompatibilidade/genética , Antígenos de Histocompatibilidade/metabolismo , Humanos , Menopausa/efeitos dos fármacos , Pessoa de Meia-Idade , Ovariectomia , Fagocitose/efeitos dos fármacos , Fagocitose/fisiologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptor 4 Toll-Like/genética , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
18.
Neurochem Int ; 60(6): 631-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22406418

RESUMO

Complement C5a is associated primarily with inflammation. The widespread expression of its receptors, C5aR and C5L2 in neuronal cells, however, suggests additional regulatory roles for C5a in the CNS. C5aR agonist (PL37-MAP) evokes Ca(2+)-influx in GT1-7 neuronal cell line and the Ca(2+)-influx is regulated by estradiol. In the present study, we examined further the mechanism of Ca(2+)-influx and the contribution of the two estrogen receptor (ER) isotypes, ERα and ERß, to estrogenic modulation of intracellular Ca(2+)-content. GT1-7 neurons were treated with isotype selective ER agonists for 24h then C5aR agonist evoked Ca(2+)-responses were measured by Ca(2+)-imaging. Transcriptional changes were followed by real-time PCR. We found that not only estradiol (100 pM), but the ERα selective agonist PPT (100 pM) enhanced the PL37-MAP-evoked Ca(2+)-influx (E2: 215%, PPT: 175%, compared to the PL37-MAP-evoked Ca(2+)-influx). In contrast, the ERß selective agonist DPN (100 pM) significantly reduced the Ca(2+)-influx (32%). Attenuated Ca(2+)-response (25%) was observed in Ca-free environment and depletion of the Ca(2+)-pool by CPA eliminated the remaining elevation in the Ca(2+)-content, demonstrating that the majority of Ca(2+) originated from the extracellular compartment. L-type voltage-gated Ca(2+)-channel (L-VGCC) blocker nifedipine abolished the Ca(2+)-influx, while R-type Ca(2+)-channel blocker SNX-482 had no effect, exemplifying the predominant role of L-VGCC in this process. Acute pre-treatments (8 min) with ER agonists did not affect the evoked Ca(2+)-influx, revealing that the observed effects of estrogens were genomic. Therefore, we checked estrogenic regulation of C5a receptors and L-VGCC subunits. ER agonists increased C5aR mRNA expression, whereas they differentially regulated C5L2. Estradiol decreased transcription of Ca(v)1.3 L-VGCC subunit. Based on these results we propose that estradiol may differentially modulate C5a-induced Ca(2+)-influx via L-VGCCs in neurons depending on the expression of the two ER isotypes.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Neurônios/metabolismo , Receptor da Anafilatoxina C5a/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/fisiologia , Sinalização do Cálcio/fisiologia , Linhagem Celular Transformada , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Receptor da Anafilatoxina C5a/agonistas , Receptor da Anafilatoxina C5a/genética
19.
J Neuroinflammation ; 8: 82, 2011 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-21774811

RESUMO

BACKGROUND: Estrogens exert anti-inflammatory and neuroprotective effects in the brain mainly via estrogen receptors α (ERα) and ß (ERß). These receptors are members of the nuclear receptor superfamily of ligand-dependent transcription factors. This study was aimed at the elucidation of the effects of ERα and ERß agonists on the expression of neuroinflammatory genes in the frontal cortex of aging female rats. METHODS: To identify estrogen-responsive immunity/inflammation genes, we treated middle-aged, ovariectomized rats with 17ß-estradiol (E2), ERα agonist 16α-lactone-estradiol (16α-LE2) and ERß agonist diarylpropionitrile (DPN), or vehicle by Alzet minipump delivery for 29 days. Then we compared the transcriptomes of the frontal cortex of estrogen-deprived versus ER agonist-treated animals using Affymetrix Rat230 2.0 expression arrays and TaqMan-based quantitative real-time PCR. Microarray and PCR data were evaluated by using Bioconductor packages and the RealTime StatMiner software, respectively. RESULTS: Microarray analysis revealed the transcriptional regulation of 21 immunity/inflammation genes by 16α-LE2. The subsequent comparative real-time PCR study analyzed the isotype specific effects of ER agonists on neuroinflammatory genes of primarily glial origin. E2 regulated the expression of sixteen genes, including down-regulation of complement C3 and C4b, Ccl2, Tgfb1, macrophage expressed gene Mpeg1, RT1-Aw2, Cx3cr1, Fcgr2b, Cd11b, Tlr4 and Tlr9, and up-regulation of defensin Np4 and RatNP-3b, IgG-2a, Il6 and ER gene Esr1. Similar to E2, both 16α-LE2 and DPN evoked up-regulation of defensins, IgG-2a and Il6, and down-regulation of C3 and its receptor Cd11b, Ccl2, RT1-Aw2 and Fcgr2b. CONCLUSIONS: These findings provide evidence that E2, 16α-LE2 and DPN modulate the expression of neuroinflammatory genes in the frontal cortex of middle-aged female rats via both ERα and ERß. We propose that ERß is a promising target to suppress regulatory functions of glial cells in the E2-deprived female brain and in various neuroinflammatory diseases.


Assuntos
Encefalite/fisiopatologia , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fatores Etários , Animais , Peso Corporal/efeitos dos fármacos , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Análise em Microsséries , Fármacos Neuroprotetores/farmacologia , Tamanho do Órgão , Ratos , Ratos Wistar , Útero/anatomia & histologia , Útero/efeitos dos fármacos
20.
Endocrinology ; 151(8): 3847-62, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20534718

RESUMO

Estradiol (E2) modulates a wide range of functions of the frontal cerebral cortex. From the onset of menopause, declining levels of E2 can cause cognitive disturbances and changes in behavior that can be counterbalanced by hormone replacement. To study the effect of E2 replacement on the cortical transcriptome in a rodent model with low serum E2 level, we treated middle-aged, ovariectomized rats with E2 or vehicle using osmotic minipumps for 4 wk. Six animals for each group were selected, and samples of their frontal cortex were subjected to expression profiling using oligonucleotide microarrays. The explored E2-regulated genes were related to neurotransmission (Adora2a, Cartpt, Drd1a, Drd2, Gjb2, Nts, and Tac1), immunity (C3, C4b, Cd74, Fcgr2b, Mpeg1, and RT1-Aw2), signal transduction (Igf2, Igfbp2, Igfbp6, Rgs9, and Sncg), transport (Abca1, Hba-a2, Slc13a3, and Slc22a8), extracellular matrix (Col1a2, Col3a1, Fmod, and Lum), and transcription (Irf7 and Nupr1). Seventy-four percent of the transcriptional changes identified by microarray were confirmed by quantitative real-time PCR. The genes identified by expression profiling indicated that chronic E2 replacement significantly altered the transcriptome of the frontal cortex. The genomic effects of E2 influenced dopaminergic and peptidergic neurotransmission, immune surveillance, adenosine and insulin-like growth factor signaling and transport processes, among other functions. Identification of these novel E2-regulated mechanisms highlights the wide range of genomic responses of the aging female frontal cerebral cortex subjected to hormone replacement. Some of the genomic effects identified in this study may underlie the beneficial effects of E2 on cognition, behavior, and neuroprotection.


Assuntos
Envelhecimento/efeitos dos fármacos , Estradiol/farmacologia , Lobo Frontal/efeitos dos fármacos , Vigilância Imunológica/genética , Transmissão Sináptica/genética , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Comportamento/efeitos dos fármacos , Cognição/efeitos dos fármacos , Cognição/fisiologia , Citoproteção/efeitos dos fármacos , Citoproteção/genética , Terapia de Reposição de Estrogênios , Feminino , Lobo Frontal/metabolismo , Lobo Frontal/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Genes/efeitos dos fármacos , Vigilância Imunológica/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Ovariectomia , Ratos , Ratos Wistar , Transmissão Sináptica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...