Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Endocrinology ; 165(7)2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38788194

RESUMO

Androgen excess is a hallmark feature of polycystic ovary syndrome (PCOS), the most common form of anovulatory infertility. Clinical and preclinical evidence links developmental or chronic exposure to hyperandrogenism with programming and evoking the reproductive and metabolic traits of PCOS. While critical androgen targets remain to be determined, central GABAergic neurons are postulated to be involved. Here, we tested the hypothesis that androgen signaling in GABAergic neurons is critical in PCOS pathogenesis in 2 well-characterized hyperandrogenic mouse models of PCOS. Using cre-lox transgenics, GABA-specific androgen receptor knockout (GABARKO) mice were generated and exposed to either acute prenatal androgen excess (PNA) or chronic peripubertal androgen excess (PPA). Females were phenotyped for reproductive and metabolic features associated with each model and brains of PNA mice were assessed for elevated GABAergic input to gonadotropin-releasing hormone (GnRH) neurons. Reproductive and metabolic dysfunction induced by PPA, including acyclicity, absence of corpora lutea, obesity, adipocyte hypertrophy, and impaired glucose homeostasis, was not different between GABARKO and wild-type (WT) mice. In PNA mice, acyclicity remained in GABARKO mice while ovarian morphology and luteinizing hormone secretion was not significantly impacted by PNA or genotype. However, PNA predictably increased the density of putative GABAergic synapses to GnRH neurons in adult WT mice, and this PNA-induced plasticity was absent in GABARKO mice. Together, these findings suggest that while direct androgen signaling in GABA neurons is largely not required for the development of PCOS-like traits in androgenized models of PCOS, developmental programming of GnRH neuron innervation is dependent upon androgen signaling in GABA neurons.


Assuntos
Modelos Animais de Doenças , Neurônios GABAérgicos , Hiperandrogenismo , Camundongos Knockout , Síndrome do Ovário Policístico , Receptores Androgênicos , Animais , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/genética , Feminino , Receptores Androgênicos/metabolismo , Receptores Androgênicos/genética , Camundongos , Neurônios GABAérgicos/metabolismo , Hiperandrogenismo/metabolismo , Hiperandrogenismo/genética , Ovário/metabolismo , Androgênios/metabolismo , Gravidez , Hormônio Liberador de Gonadotropina/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/genética
2.
Curr Opin Pharmacol ; 68: 102345, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36621270

RESUMO

Polycystic ovary syndrome is a prevalent endocrinopathy involving androgen excess, and anovulatory infertility. The disorder is also associated with many comorbidities such as obesity and hyperinsulinemia, and an increased risk of cardiovascular complications. Reproductive, endocrine, and metabolic symptoms are highly variable, with heterogenous phenotypes adding complexity to clinical management of symptoms. This review highlights recent findings regarding emerging therapies for treating polycystic ovary syndrome, including i) pharmacological agents to target androgen excess, ii) modulation of kisspeptin signalling to target central neuroendocrine dysregulation, and iii) novel insulin sensitisers to combat peripheral metabolic dysfunction.


Assuntos
Resistência à Insulina , Síndrome do Ovário Policístico , Feminino , Humanos , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/metabolismo , Androgênios , Comorbidade , Obesidade/tratamento farmacológico , Obesidade/complicações
3.
Psychoneuroendocrinology ; 96: 132-141, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29940426

RESUMO

Maternal obesity during pregnancy can impact long-term health, predisposition to disease, and risk of neurological disorders in offspring. This may arise from disruption to epigenetic processes during offspring brain development. Using a maternal high fat diet (mHFD) mouse model, we investigated the expression of genes encoding epigenetic regulators in the brains of gestational day (GD) 17.5 mHFD offspring. We found significant, regionally unique changes in expression of epigenetic regulators in the developing brain of mHFD offspring compared to controls, with Gadd45b downregulated in medial prefrontal cortex, Mecp2 downregulated in amygdala, and sex-specific downregulation of Crebbp, Dnmt3b, and Mecp2 in male mHFD hippocampus. Decreased Mecp2 in the amygdala was associated with significant upregulation of the Mecp2-repressed gene, Tbr1, and an increased number of TBR1+ glutamatergic neurons in the basomedial nucleus of the amygdala. Tbr1 upregulation in amygdala was also observed in postnatal day 8 (P8) mHFD offspring, and levels of glutamate receptor gene Grin2b, and Fos, a marker for neuronal activity, were increased. Indications of heightened excitatory drive in mHFD offspring amygdala were associated with an anxiety-like phenotype, with mHFD offspring displaying altered ultrasonic vocalization characteristics at P8, and adult female mHFD offspring spending decreased time on the open arm of the Elevated Plus Maze. Together, this data provides insight into sex-specific offspring vulnerability to perinatal mHFD programming of anxiety-like behaviors.


Assuntos
Tonsila do Cerebelo/metabolismo , Ansiedade/metabolismo , Epigênese Genética/fisiologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Transtornos de Ansiedade , Encéfalo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dieta Hiperlipídica/efeitos adversos , Fármacos Atuantes sobre Aminoácidos Excitatórios/metabolismo , Feminino , Hipocampo , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Proteínas com Domínio T
4.
Int J Dev Neurosci ; 53: 18-25, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27326907

RESUMO

Maternal obesity during pregnancy is associated with chronic maternal, placental, and fetal inflammation; and it elevates the risk for offspring obesity. Changes in the development of the hypothalamus, a brain region that regulates body weight and energy balance, are emerging as important determinants of offspring risk, but such changes are only beginning to be defined. Here we focused on the hypothesis that the pathological exposure of developing hypothalamic astrocytes to cytokines would alter their development. A maternal high-fat diet (mHFD) mouse model was used to investigate changes in hypothalamic astrocytes in the fetus during late gestation and in early neonates by using immunochemistry, confocal microscopy, and qPCR. The number of astrocytes and the proportion of proliferating astrocytes was significantly higher in the arcuate nucleus (ARC) and the supraoptic nucleus (SON) of the hypothalamus at both ages compared to control offspring from normal weight pregnancies. Supplemental to this we found that cultured fetal hypothalamic astrocytes proliferated significantly in response to IL6 (10ng/ml), one of the cytokines significantly elevated in fetuses of obese dams, via the JAK/STAT3 signaling pathway. Thus, maternal obesity during pregnancy stimulated the proliferation and thereby increased numbers of astrocytes in the fetal as well as early neonatal hypothalamus, which may be driven, during fetal life, by IL6.


Assuntos
Astrócitos/metabolismo , Desenvolvimento Fetal/fisiologia , Hipotálamo/embriologia , Hipotálamo/patologia , Neurônios/fisiologia , Obesidade , Animais , Astrócitos/efeitos dos fármacos , Peso Corporal , Bromodesoxiuridina/metabolismo , Proliferação de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Proteína Glial Fibrilar Ácida/metabolismo , Interleucina-6/farmacologia , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Proteína Oncogênica v-akt/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
5.
J Neurol Sci ; 351(1-2): 140-145, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25813273

RESUMO

Congenital mirror movements (CMM) is a disorder characterized by unintentional mirroring in homologous motor systems of voluntary movements on the opposite side, usually affecting the distal upper extremities. Genetic analyses have revealed involvement of three genes (DCC, RAD51, and DNAL4). We sought to distinguish whether different phenotypes of CMM exist, and if so, whether they might map to different causative genes. We studied 14 individuals across five families with dominantly-inherited CMM. We used accelerometer gloves to analyse the fine detail of index finger tapping movements, and applied standard genetic methodology to analyse DNA samples. Two forms of mirroring were distinguished: 'actual' in which the mirroring followed precisely the movements of the voluntary hand, and 'fractionated' in which the mirroring was saccadic. We found that actual mirroring was characteristic of individuals in a family with a RAD51 mutation, and fractionated more characteristic of a family with a DCC mutation. These findings are suggestive of specific genotype-phenotype correlations in CMM. Three heterozygous individuals (one RAD51; two DCC) showed no apparent mirroring on visual inspection, although mirroring was detectable with the accelerometer gloves. Thus, subclinical mirroring may be present even when undetectable on clinical observation.


Assuntos
Transtornos dos Movimentos/genética , Rad51 Recombinase/genética , Receptores de Superfície Celular/genética , Proteínas Supressoras de Tumor/genética , Adolescente , Adulto , Idoso , Dineínas do Axonema/genética , Receptor DCC , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Adulto Jovem
6.
Endocrinology ; 155(7): 2566-77, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24773340

RESUMO

Maternal obesity during pregnancy increases the risk of obesity in the offspring. Several observations have pointed to a causative role for the proinflammatory cytokine IL-6, but whether it is present in the fetal circulation and how it acts on the developing fetus are unclear. We first observed that postnatal day 0 offspring from obese mothers had significantly reduced neuropeptide Y (NPY) innervation of the paraventricular nucleus (PVN) compared with that for offspring of normal-weight controls. Thus, the growth of NPY neurites from the arcuate nucleus (ARC) was impaired in the fetal brain by maternal obesity. The neurite growth regulator, Netrin-1, was expressed in the ARC and PVN and along the pathway between the two at gestational day (GD) 17.5 in normal animals, making it likely to be involved in the development of NPY ARC-PVN projections. In addition, the expression of Dcc and Unc5d, receptors for Netrin-1, were altered in the GD17.5 ARC in obese but not normal weight pregnancies. Thus, this important developmental pathway is perturbed by maternal obesity and may explain the defect in NPY innervation of the PVN that occurs in fetuses developing in obese mothers. To investigate whether IL-6 may play a role in these developmental changes, we found first that IL-6 was significantly elevated in the fetal and maternal circulation in pregnancies of obese mice compared with those of normal-weight mice. In addition, treatment of GD17.5 ARC tissue with IL-6 in vitro significantly reduced ARC neurite outgrowth and altered developmental gene expression similar to maternal obesity in vivo. These findings demonstrate that maternal obesity may alter the way in which fetal ARC NPY neurons respond to key developmental signals that regulate normal prenatal neural connectivity and suggest a causative role for elevated IL-6 in these changes.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Interleucina-6/farmacologia , Neuritos/efeitos dos fármacos , Obesidade/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/embriologia , Núcleo Arqueado do Hipotálamo/metabolismo , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Relação Dose-Resposta a Droga , Feminino , Imuno-Histoquímica , Hibridização In Situ , Interleucina-6/sangue , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Netrina-1 , Neuritos/metabolismo , Neuritos/fisiologia , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Obesidade/etiologia , Obesidade/genética , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/embriologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Gravidez , Complicações na Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
7.
Cereb Cortex ; 18(1): 53-66, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17478416

RESUMO

Adult neocortical areas are characterized by marked differences in cytoarchitecture and connectivity that underlie their functional roles. The molecular determinants of these differences are largely unknown. We performed a microarray analysis to identify molecules that define the somatosensory and visual areas during the time when afferent and efferent projections are forming. We identified 122 molecules that are differentially expressed between the regions and confirmed by quantitative polymerase chain reaction 95% of the 20 genes tested. Two genes were chosen for further investigation: Bcl6 and Ten_m3. Bcl6 was highly expressed in the superficial cortical plate corresponding to developing layer IV of somatosensory cortex at postnatal day (P) 0. This had diminished by P3, but strong expression was found in layer V pyramidal cells by P7 and was maintained until adulthood. Retrograde tracing showed that Bcl6 is expressed in corticospinal neurons. Ten_m3 was expressed in a graded pattern within layer V of caudal cortex that corresponds well with visual cortex. Retrograde tracing and immunostaining showed that Ten_m3 is highly expressed along axonal tracts of projection neurons of the developing visual pathway. Overexpression demonstrated that Ten_m3 promotes homophilic adhesion and neurite outgrowth in vivo. This suggests an important role for Ten_m3 in the development of the visual pathway.


Assuntos
Padronização Corporal/fisiologia , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neocórtex/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Córtex Somatossensorial/fisiologia , Vias Visuais/fisiologia , Animais , Proteínas de Membrana , Camundongos , Neocórtex/citologia , Proteínas Proto-Oncogênicas c-bcl-6 , Córtex Somatossensorial/citologia , Distribuição Tecidual , Vias Visuais/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA