Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 57(6): 2727-2740, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32333254

RESUMO

Aging is accompanied by aberrant gene expression that ultimately affects brain plasticity and the capacity to form long-term memories. Immediate-early genes (IEGs) play an active role in these processes. Using a rat model of normal cognitive aging, we found that the expression of Egr1 and c-Fos was associated with chronological age, whereas Arc was more tightly linked to cognitive outcomes in aging. More specifically, constitutive Arc expression was significantly elevated in aged rats with memory impairment compared to cognitively intact aged rats and young adult animals. Since alterations in the neuroepigenetic mechanisms that gate hippocampal gene expression are also associated with cognitive outcome in aging, we narrowed our focus on examining potential epigenetic mechanisms that may lead to aberrant Arc expression. Employing a multilevel analytical approach using bisulfite sequencing, chromatin immunoprecipitations, and micrococcal nuclease digestion, we identified CpG sites in the Arc promoter that were coupled to poor cognitive outcomes in aging, histone marks that were similarly coupled to spatial memory deficits, and nucleosome positioning that also varied depending on cognitive status. Together, these findings paint a diverse and complex picture of the Arc epigenetic landscape in cognitive aging and bolster a body of work, indicating that dysfunctional epigenetic regulation is associated with memory impairment in the aged brain.


Assuntos
Envelhecimento Cognitivo/fisiologia , Proteínas do Citoesqueleto/genética , Aprendizagem em Labirinto/fisiologia , Proteínas do Tecido Nervoso/genética , Memória Espacial/fisiologia , Animais , Proteínas do Citoesqueleto/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Epigênese Genética , Hipocampo/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Long-Evans
2.
Hippocampus ; 29(12): 1165-1177, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31334577

RESUMO

Changes in neuronal network activity and increased interindividual variability in memory are among the most consistent features of growing older. Here, we examined the relationship between these hallmarks of aging. Young and aged rats were trained on a water maze task where aged individuals reliably display an increased range of spatial memory capacities relative to young. Two weeks later, neuronal activity was induced pharmacologically with a low dose of pilocarpine and control animals received vehicle. Activity levels were proxied by quantifying the immediate early gene products Arc and c-Fos. While no relationship was observed between basal, resting activity, and individual differences in spatial memory in any brain region, pilocarpine-induced marker expression was tightly coupled with memory in all areas of the prefrontal cortex (PFC) and hippocampus examined. The nature of this association, however, differed across regions and in relation to age-related cognitive outcome. Specifically, in the medial PFC, induced activity was greatest in aged rats with cognitive impairment and correlated with water maze performance across all subjects. In the hippocampus, the range of induced marker expression was comparable between groups and similarly coupled with memory in both impaired and unimpaired aged rats. Together the findings highlight that the dynamic range of neural network activity across multiple brain regions is a critical component of neurocognitive aging.


Assuntos
Envelhecimento Cognitivo/fisiologia , Hipocampo/metabolismo , Aprendizagem em Labirinto/fisiologia , Rede Nervosa/metabolismo , Córtex Pré-Frontal/metabolismo , Animais , Envelhecimento Cognitivo/psicologia , Proteínas do Citoesqueleto/biossíntese , Hipocampo/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Agonistas Muscarínicos/farmacologia , Rede Nervosa/efeitos dos fármacos , Proteínas do Tecido Nervoso/biossíntese , Pilocarpina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/biossíntese , Ratos , Ratos Long-Evans
3.
Neuroscience ; 394: 303-315, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30482274

RESUMO

Age- and menopause-related deficits in working memory can be partially restored with estradiol replacement in women and female nonhuman primates. Working memory is a cognitive function reliant on persistent firing of dorsolateral prefrontal cortex (dlPFC) neurons that requires the activation of GluN2B-containing glutamate NMDA receptors. We tested the hypothesis that the distribution of phospho-Tyr1472-GluN2B (pGluN2B), a predominant form of GluN2B seen at the synapse, is sensitive to aging or estradiol treatment and coupled to working memory performance. First, ovariectomized young and aged rhesus monkeys (Macaca mulatta) received long-term cyclic vehicle (V) or estradiol (E) treatment and were tested on the delayed response (DR) test of working memory. Then, serial section electron microscopic immunocytochemistry was performed to quantitatively assess the subcellular distribution of pGluN2B. While the densities of pGluN2B immunogold particles in dlPFC dendritic spines were not different across age or treatment groups, the percentage of gold particles located within the synaptic compartment was significantly lower in aged-E monkeys compared to young-E and aged-V monkeys. On the other hand, the percentage of pGluN2B gold particles in the spine cytoplasm was decreased with E treatment in young, but increased with E in aged monkeys. In aged monkeys, DR average accuracy inversely correlated with the percentage of synaptic pGluN2B, while it positively correlated with the percentage of cytoplasmic pGluN2B. Together, E replacement may promote cognitive health in aged monkeys, in part, by decreasing the relative representation of synaptic pGluN2B and potentially protecting the dlPFC from calcium toxicity.


Assuntos
Envelhecimento , Estrogênios/administração & dosagem , Memória de Curto Prazo/fisiologia , Córtex Pré-Frontal/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Sinapses/fisiologia , Animais , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/fisiologia , Espinhas Dendríticas/ultraestrutura , Feminino , Macaca mulatta , Memória de Curto Prazo/efeitos dos fármacos , Fosforilação , Densidade Pós-Sináptica/ultraestrutura , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/ultraestrutura , Receptores de N-Metil-D-Aspartato/ultraestrutura , Sinapses/efeitos dos fármacos , Sinapses/ultraestrutura
4.
Cereb Cortex ; 27(3): 2022-2033, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-26941383

RESUMO

Age- and menopause-related impairment in working memory mediated by the dorsolateral prefrontal cortex (dlPFC) occurs in humans and nonhuman primates. Long-term cyclic 17ß-estradiol treatment rescues cognitive deficits in aged ovariectomized rhesus monkeys while restoring highly plastic synapses. Here we tested whether distributions of G protein-coupled estrogen receptor 1 (GPER1) within monkey layer III dlPFC synapses are sensitive to age and estradiol, and coupled to cognitive function. Ovariectomized young and aged monkeys administered vehicle or estradiol were first tested on a delayed response test of working memory. Then, quantitative serial section immunoelectron microscopy was used to determine the distributions of synaptic GPER1. GPER1-containing nonperforated axospinous synapse density was reduced with age, and partially restored with estrogen treatment. The majority of synapses expressed GPER1, which was predominately localized to presynaptic cytoplasm and mitochondria. GPER1 was also abundant at plasmalemmas, and within cytoplasmic and postsynaptic density (PSD) domains of dendritic spines. GPER1 levels did not differ with age or treatment, and none of the variables examined were tightly associated with cognitive function. However, greater representation of GPER1 subjacent to the PSD accompanied higher synapse density. These data suggest that GPER1 is positioned to support diverse functions key to synaptic plasticity in monkey dlPFC.


Assuntos
Envelhecimento/metabolismo , Receptor alfa de Estrogênio/metabolismo , Menopausa/metabolismo , Córtex Pré-Frontal/metabolismo , Sinapses/metabolismo , Envelhecimento/patologia , Animais , Estradiol/administração & dosagem , Estrogênios/administração & dosagem , Feminino , Terapia de Reposição Hormonal , Imuno-Histoquímica , Macaca mulatta , Microscopia Imunoeletrônica , Plasticidade Neuronal/fisiologia , Ovariectomia , Córtex Pré-Frontal/patologia , Sinapses/patologia
5.
J Neurosci ; 36(3): 901-10, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26791219

RESUMO

Humans and nonhuman primates are vulnerable to age- and menopause- related decline in working memory, a cognitive function reliant on area 46 of the dorsolateral prefrontal cortex (dlPFC). We showed previously that presynaptic mitochondrial number and morphology in monkey dlPFC neurons correlate with working memory performance. The current study tested the hypothesis that the types of synaptic connections these boutons form are altered with aging and menopause in rhesus monkeys and that these metrics may be coupled with mitochondrial measures and working memory. Using serial section electron microscopy, we examined the frequencies and characteristics of nonsynaptic, single-synaptic, and multisynaptic boutons (MSBs) in the dlPFC. In contrast to our previous observations in the monkey hippocampal dentate gyrus, where MSBs comprised ∼40% of boutons, the vast majority of dlPFC boutons were single-synaptic, whereas MSBs constituted a mere 10%. The frequency of MSBs was not altered by normal aging, but decreased by over 50% with surgical menopause induced by ovariectomy in aged monkeys. Cyclic estradiol treatment in aged ovariectomized animals restored MSB frequencies to levels comparable to young and aged premenopausal monkeys. Notably, the frequency of MSBs positively correlated with working memory scores, as measured by the average accuracy on the delayed response (DR) test. Furthermore, MSB incidence positively correlated with the number of healthy straight mitochondria in dlPFC boutons and inversely correlated with the number of pathological donut-shaped mitochondria. Together, our data suggest that MSBs are coupled to cognitive function and mitochondrial health and are sensitive to estrogen. Significance statement: Many aged menopausal individuals experience deficits in working memory, an executive function reliant on recurrent firing of prefrontal cortex (PFC) neurons. However, little is known about the organization of presynaptic inputs to these neurons and how they may be altered with aging and menopause. Multisynaptic boutons (MSBs) were of particular interest, because they form multiple synapses and can enhance coupling between presynaptic and postsynaptic neurons. We found that higher MSB frequency correlated with better working memory performance in rhesus monkeys. Additionally, aged surgically menopausal monkeys experienced a 50% loss of MSBs that was restored with cyclic estradiol treatment. Together, our findings suggest that hormone replacement therapy benefits cognitive aging, in part by retaining complex synaptic organizations in the PFC.


Assuntos
Envelhecimento/metabolismo , Estrogênios/farmacologia , Memória de Curto Prazo/fisiologia , Córtex Pré-Frontal/metabolismo , Terminações Pré-Sinápticas/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Terapia de Reposição de Estrogênios/métodos , Feminino , Macaca mulatta , Memória de Curto Prazo/efeitos dos fármacos , Ovariectomia , Córtex Pré-Frontal/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
6.
FEBS Lett ; 588(17): 3233-9, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-25019573

RESUMO

Pancreatic polypeptide (PP) is a major agonist for neuropeptide Y4 receptors (NPY4R). While NPY4R has been identified in various tissues, the cells on which it is expressed and its function in those cells has not been clearly delineated. Here we report that NPY4R is present in all somatostatin-containing cells of tissues that we tested, including pancreatic islets, duodenum, hippocampus, and hypothalamus. Its agonism by PP decreases somatostatin secretion from human islets. Mouse embryonic hippocampal (mHippo E18) cells expressed NPY4Rs and their activation by PP consistently decreased somatostatin secretion. Furthermore, central injection of PP in mice induced c-Fos immunoreactivity in somatostatin-containing cells in the hippocampus compared with PBS-injected mice. In sum, our results identify PP as a pivotal modulator of somatostatin secretion.


Assuntos
Polipeptídeo Pancreático/farmacologia , Somatostatina/metabolismo , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/citologia , Humanos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Especificidade de Órgãos , Receptores de Neuropeptídeo Y/agonistas , Receptores de Neuropeptídeo Y/metabolismo
7.
J Exp Zool A Ecol Genet Physiol ; 321(7): 399-414, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24862737

RESUMO

Steroid hormone receptors are widely and heterogeneously expressed in the brain, and are regulated by age and gonadal hormones. Our goal was to quantify effects of aging, long-term estradiol (E2 ) treatment, and their interactions, on expression of G protein-coupled estrogen receptor (GPER), estrogen receptor α (ERα) and progesterone receptor (PR) immunoreactivity in two hypothalamic regions, the arcuate (ARC) and the periventricular area (PERI) of rhesus monkeys as a model of menopause and hormone replacement. Ovariectomized (OVX) rhesus macaques were young (∼ 11 years) or aged (∼ 25 years), given oil (vehicle) or E2 every 3 weeks for 2 years. Immunohistochemistry and stereologic analysis of ERα, PR, and GPER was performed. More effects were detected for GPER than the other two receptors. Specifically, GPER cell density in the ARC and PERI, and the percent of GPER-immunoreactive cells in the PERI, were greater in aged than in young monkeys. In addition, we mapped the qualitative distribution of GPER in the monkey hypothalamus and nearby regions. For ERα, E2 treated monkeys tended to have higher cell density than vehicle monkeys in the ARC. The percent of PR density in the PERI tended to be higher in E2 than vehicle monkeys of both ages. This study shows that the aged hypothalamus maintains expression of hormone receptors with age, and that long-term cyclic E2 treatment has few effects on their expression, although GPER was affected more than ERα or PR. This result is surprising in light of evidence for E2 regulation of the receptors studied here, and differences may be due to the selected regions, long-term nature of E2 treatment, among other possibilities.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Macaca mulatta , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Progesterona/metabolismo , Envelhecimento , Animais , Esquema de Medicação , Estradiol/administração & dosagem , Receptor alfa de Estrogênio/genética , Estrogênios/administração & dosagem , Estrogênios/farmacologia , Feminino , Imuno-Histoquímica , Receptores Acoplados a Proteínas G/genética , Receptores de Progesterona/genética
8.
Proc Natl Acad Sci U S A ; 111(1): 486-91, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24297907

RESUMO

Humans and nonhuman primates are vulnerable to age- and menopause-related decline in working memory, a cognitive function reliant on the energy-demanding recurrent excitation of neurons within Brodmann's Area 46 of the dorsolateral prefrontal cortex (dlPFC). Here, we tested the hypothesis that the number and morphology (straight, curved, or donut-shaped) of mitochondria in dlPFC presynaptic boutons are altered with aging and menopause in rhesus monkeys (Macaca mulatta) and that these metrics correlate with delayed response (DR) accuracy, a well-characterized measure of dlPFC-dependent working memory. Although presynaptic bouton density or size was not significantly different across groups distinguished by age or menses status, DR accuracy correlated positively with the number of total and straight mitochondria per dlPFC bouton. In contrast, DR accuracy correlated inversely with the frequency of boutons containing donut-shaped mitochondria, which exhibited smaller active zone areas and fewer docked synaptic vesicles than those with straight or curved mitochondria. We then examined the effects of estrogen administration to test whether a treatment known to improve working memory influences mitochondrial morphology. Aged ovariectomized monkeys treated with vehicle displayed significant working memory impairment and a concomitant 44% increase in presynaptic donut-shaped mitochondria, both of which were reversed with cyclic estradiol treatment. Together, our data suggest that hormone replacement therapy may benefit cognitive aging, in part by promoting mitochondrial and synaptic health in the dlPFC.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Memória de Curto Prazo/fisiologia , Mitocôndrias/metabolismo , Córtex Pré-Frontal/fisiologia , Terminações Pré-Sinápticas/fisiologia , Envelhecimento , Animais , Comportamento Animal , Mapeamento Encefálico/métodos , Cognição , Feminino , Haplorrinos , Imageamento Tridimensional , Macaca mulatta , Memória de Curto Prazo/efeitos dos fármacos , Ciclo Menstrual , Córtex Pré-Frontal/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , Reprodutibilidade dos Testes , Transmissão Sináptica , Fatores de Tempo
9.
Neurobiol Aging ; 34(7): 1882-90, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23369546

RESUMO

Preclinical studies in aged, surgically-menopausal rhesus monkeys have revealed powerful benefits of intermittent estrogen injections on prefrontal cortex-dependent working memory, together with corresponding effects on dendritic spine morphology in the prefrontal cortex. This contrasts with the inconsistent effects of hormone therapy (HT) reported in clinical studies in women. Factors contributing to this discrepancy could include differences in the formulation and sequence of HT regimens, resulting in different neurobiological outcomes. The current study evaluated, in aging surgically menopausal rhesus monkeys, the cognitive effects of 4 HT regimens modeled directly on human clinical practice, including continuous estrogen treatment opposed by progesterone. None of the regimens tested produced any cognitive effect, despite yielding physiologically relevant serum hormone levels, as intended. These findings have implications for the design of regimens that might optimize the benefits of hormone treatment for healthy aging, and suggest that common HT protocols used by women may fail to result in substantial cognitive benefit, at least via direct effects on the prefrontal cortex.


Assuntos
Envelhecimento/efeitos dos fármacos , Envelhecimento/psicologia , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/psicologia , Estradiol/administração & dosagem , Terapia de Reposição de Estrogênios/métodos , Ovariectomia , Envelhecimento/sangue , Animais , Transtornos Cognitivos/sangue , Implantes de Medicamento , Estradiol/sangue , Feminino , Humanos , Macaca mulatta
10.
J Neurosci ; 32(34): 11700-5, 2012 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-22915112

RESUMO

Preclinical animal models have provided strong evidence that estrogen (E) therapy (ET) enhances cognition and induces spinogenesis in neuronal circuits. However, clinical studies have been inconsistent, with some studies revealing adverse effects of ET, including an increased risk of dementia. In an effort to bridge this disconnect between the preclinical and clinical data, we have developed a nonhuman primate (NHP) model of ET combined with high-resolution dendritic spine analysis of dorsolateral prefrontal cortical (dlPFC) neurons. Previously, we reported cyclic ET in aged, ovariectomized NHPs increased spine density on dlPFC neurons. Here, we report that monkeys treated with cyclic E treatment paired with cyclic progesterone (P), continuous E combined with P (either cyclic or continuous), or unopposed continuous E failed to increase spines on dlPFC neurons. Given that the most prevalent form of ET prescribed to women is a combined and continuous E and P, these data bring into convergence the human neuropsychological findings and preclinical neurobiological evidence that standard hormone therapy in women is unlikely to yield the synaptic benefit presumed to underlie the cognitive enhancement reported in animal models.


Assuntos
Envelhecimento/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Estrogênios/farmacologia , Neurônios/citologia , Córtex Pré-Frontal/citologia , Progesterona/farmacologia , Envelhecimento/patologia , Análise de Variância , Animais , Estrogênios/sangue , Feminino , Macaca mulatta , Microscopia Confocal , Neurônios/efeitos dos fármacos , Ovariectomia , Córtex Pré-Frontal/efeitos dos fármacos , Progesterona/sangue
11.
Pharmacol Biochem Behav ; 100(1): 205-11, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21875615

RESUMO

Some patients experience enduring cognitive impairment after cancer treatment, a condition termed "chemofog". Animal models allow assessment of chemotherapy effects on learning and memory per se, independent of changes due to cancer itself or associated health consequences such as depression. The present study examined the long-term learning and memory effects of a chemotherapy cocktail used widely in the treatment of breast cancer, consisting of 5-fluorouracil (5FU) and cyclophosphamide (CYP). Eighty 5-month old male F344 rats received contextual and cued fear conditioning before treatment with saline, or a low or high dose drug cocktail (50mg/kg CYP and 75 mg/kg 5FU, or 75 mg/kg CYP and 120 mg/kg 5FU, i.p., respectively) every 30 days for 2 months. After a 2-month, no-drug recovery, both long-term retention and new task acquisition in the water maze and 14-unit T-maze were assessed. Neither dose of the CYP/5FU cocktail impaired retrograde fear memory despite marked toxicity documented by enduring weight loss and 50% mortality at the higher dose. Acquisition in the water maze and Stone maze was also normal relative to controls in rats treated with CYP/5FU. The results contribute to a growing literature suggesting that learning and memory mediated by the hippocampus can be relatively resistant to chemotherapy. Future investigation may need to focus on assessments of processing speed, executive function and attention, and the possible interactive contribution of cancer itself and aging to the post-treatment development of cognitive impairment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Memória/efeitos dos fármacos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Transtornos Cognitivos/induzido quimicamente , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Fluoruracila/administração & dosagem , Fluoruracila/efeitos adversos , Aprendizagem/efeitos dos fármacos , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Ratos , Ratos Endogâmicos F344 , Resultado do Tratamento
12.
J Neurosci ; 30(38): 12770-6, 2010 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-20861381

RESUMO

In rat hippocampus, estrogen receptor-α (ER-α) can initiate nongenomic signaling mechanisms that modulate synaptic plasticity in response to either circulating or locally synthesized estradiol (E). Here we report quantitative electron microscopic data demonstrating that ER-α is present within excitatory synapses in dorsolateral prefrontal cortex (dlPFC) of young and aged ovariectomized female rhesus monkeys with and without E treatment. There were no treatment or age effects on the percentage of excitatory synapses containing ER-α, nor were there any group differences in distribution of ER-α within the synapse. However, the mean size of synapses containing ER-α was larger than that of unlabeled excitatory synapses. All monkeys were tested on delayed response (DR), a cognitive test of working memory that requires dlPFC. In young ovariectomized monkeys without E treatment, presynaptic ER-α correlated with DR accuracy across memory delays. In aged monkeys that received E treatment, ER-α within the postsynaptic density (30-60 nm from the synaptic membrane) positively correlated with DR performance. Thus, although the lack of group effects suggests that ER-α is primarily in synapses that are stable across age and treatment, synaptic abundance of ER-α is correlated with individual performance in two key age/treatment groups. These data have important implications for individual differences in the cognitive outcome among menopausal women and promote a focus on cortical estrogen receptors for therapeutic efficacy with respect to cognition.


Assuntos
Cognição/fisiologia , Receptor alfa de Estrogênio/metabolismo , Córtex Pré-Frontal/metabolismo , Sinapses/metabolismo , Análise de Variância , Animais , Cognição/efeitos dos fármacos , Estradiol/metabolismo , Estradiol/farmacologia , Feminino , Macaca mulatta , Memória de Curto Prazo/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Microscopia Imunoeletrônica , Testes Neuropsicológicos , Ovariectomia , Córtex Pré-Frontal/efeitos dos fármacos , Sinapses/efeitos dos fármacos
13.
Ann N Y Acad Sci ; 1204: 104-12, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20738280

RESUMO

The surprising discovery in 1990 that estrogen modulates hippocampal structural plasticity launched a whole new field of scientific inquiry. Over the past two decades, estrogen-induced spinogenesis has been described in several brain areas involved in cognition in a number of species, in both sexes and on multiple time scales. Exploration into the interaction between estrogen and aging has illuminated some of the hormone's neuroprotective effects, most notably on age-related cognitive decline in nonhuman primates. Although there is still much to be learned about the mechanisms by which estrogen exerts its actions, key components of the signal transduction pathways are beginning to be elucidated and nongenomic actions via membrane bound estrogen receptors are of particular interest. Future studies are focused on identifying the most clinically relevant hormone treatment, as well as the potential identification of new therapeutics that can prevent or reverse age-related cognitive impairment by intercepting specific signal transduction pathways initiated by estrogen.


Assuntos
Envelhecimento/fisiologia , Encéfalo/fisiologia , Estrogênios/metabolismo , Animais , Encéfalo/metabolismo , Feminino , Hipocampo/metabolismo , Hipocampo/fisiologia , Humanos , Masculino , Modelos Animais , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiologia
14.
Proc Natl Acad Sci U S A ; 104(27): 11465-70, 2007 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-17592140

RESUMO

We previously reported that long-term cyclic estrogen (E) treatment reverses age-related impairment of cognitive function mediated by the dorsolateral prefrontal cortex (dlPFC) in ovariectomized (OVX) female rhesus monkeys, and that E induces a corresponding increase in spine density in layer III dlPFC pyramidal neurons. We have now investigated the effects of the same E treatment in young adult females. In contrast to the results for aged monkeys, E treatment failed to enhance dlPFC-dependent task performance relative to vehicle control values (group young OVX+Veh) but nonetheless led to a robust increase in spine density. This response was accompanied by a decline in dendritic length, however, such that the total number of spines per neuron was equivalent in young OVX+Veh and OVX+E groups. Robust effects of chronological age, independent of ovarian hormone status, were also observed, comprising significant age-related declines in dendritic length and spine density, with a preferential decrease in small spines in the aged groups. Notably, the spine effects were partially reversed by cyclic E administration, although young OVX+Veh monkeys still had a higher complement of small spines than did aged E treated monkeys. In summary, layer III pyramidal neurons in the dlPFC are sensitive to ovarian hormone status in both young and aged monkeys, but these effects are not entirely equivalent across age groups. The results also suggest that the cognitive benefit of E treatment in aged monkeys is mediated by enabling synaptic plasticity through a cyclical increase in small, highly plastic dendritic spines in the primate dlPFC.


Assuntos
Envelhecimento/fisiologia , Cognição/fisiologia , Estradiol/fisiologia , Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia , Animais , Feminino , Macaca mulatta , Córtex Pré-Frontal/citologia , Tempo de Reação/fisiologia
15.
J Neurosci ; 26(9): 2571-8, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16510735

RESUMO

Long-term cyclic treatment with 17beta-estradiol reverses age-related impairment in ovariectomized rhesus monkeys on a test of cognitive function mediated by the prefrontal cortex (PFC). Here, we examined potential neurobiological substrates of this effect using intracellular loading and morphometric analyses to test the possibility that the cognitive benefits of hormone treatment are associated with structural plasticity in layer III pyramidal cells in PFC area 46. 17beta-Estradiol did not affect several parameters such as total dendritic length and branching. In contrast, 17beta-estradiol administration increased apical and basal dendritic spine density, and induced a shift toward smaller spines, a response linked to increased spine motility, NMDA receptor-mediated activity, and learning. These results document that, although the aged primate PFC is vulnerable in the absence of factors such as circulating estrogens, it remains responsive to long-term cyclic 17beta-estradiol treatment, and that increased dendritic spine density and altered spine morphology may contribute to the cognitive benefits of such treatment.


Assuntos
Tamanho Celular/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Estradiol/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/citologia , Envelhecimento/fisiologia , Animais , Comportamento Animal , Feminino , Imageamento Tridimensional/métodos , Isoquinolinas/metabolismo , Macaca mulatta , Ovariectomia/métodos , Córtex Pré-Frontal/efeitos dos fármacos
16.
Cereb Cortex ; 14(2): 215-23, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14704219

RESUMO

While studies have shown that estrogen affects hippocampal spine density and function, behavioral studies in humans and nonhuman primates have also implicated the prefrontal cortex in the effects of estrogen on cognition. However, the potential for similar estrogen-induced increases in spines and synapses in the prefrontal cortex has not been investigated in primates. Moreover, it is not known if such an estrogen effect would be manifested throughout the neocortex or primarily in the regions involved in cognition. Therefore, we investigated the effects of estrogen on dendritic spines in the prefrontal and primary visual cortices of young rhesus monkeys. Young female monkeys were ovariectomized and administered either estradiol cypionate or vehicle by intramuscular injection. Using an antibody against the spine-associated protein, spinophilin, spine numbers were estimated in layer I of area 46 and in layer I of the opercular portion of area V1 (V1o). Spine numbers in layer I of area 46 were significantly increased (55%) in the ovariectomy + estrogen group compared to the ovariectomy + vehicle group, yet spine numbers in layer I of area V1o were equivalent across the two groups. The present results suggest that estrogen's effects on synaptic organization influence select neocortical layers and regions in a primate model, and provide a morphological basis for enhanced prefrontal cortical functions following estrogen replacement.


Assuntos
Dendritos/química , Dendritos/efeitos dos fármacos , Estradiol/farmacologia , Terapia de Reposição de Estrogênios/métodos , Proteínas dos Microfilamentos/análise , Proteínas do Tecido Nervoso/análise , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/efeitos dos fármacos , Animais , Contagem de Células/métodos , Feminino , Imuno-Histoquímica , Macaca mulatta , Ovariectomia , Córtex Pré-Frontal/química
17.
J Comp Neurol ; 465(4): 540-50, 2003 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-12975814

RESUMO

It is well documented that estrogen increases dendritic spine density in CA1 pyramidal cells of young female rats. However, this effect is attenuated in aged rats. We report here a quantitative analysis of estrogen effects on hippocampal spine number as visualized with antispinophilin in young (6-8 years old) and aged (19-23 years old) female rhesus monkeys, a species with a pattern of female endocrine senescence comparable to that of humans. Monkeys were ovariectomized and administered either vehicle or estradiol cypionate 3 months postovariectomy, followed by an additional dose 3 weeks later, with perfusion 24 hours after the last estrogen treatment. Immunolocalization of spinophilin, a spine-associated protein, was used for quantitative stereologic analyses of total spinophilin-immunoreactive spine numbers in CA1 stratum radiatum and the inner and outer molecular layers of dentate gyrus. In both young and aged female monkeys, the estrogen-treated groups had an increase in spinophilin-immunoreactive spines (37% in young, P <.005; 35% in aged, P <.05) compared with the untreated groups that amounted to more than 1 billion additional immunoreactive spines. The young group also showed a trend toward an estrogen-induced increase in immunoreactive spines in the dentate gyrus outer molecular layer, but this effect was not statistically significant (P =.097). We conclude that spine number in the rhesus monkey hippocampus is highly responsive to estrogen, yet, unlike the female rat, aged female rhesus monkeys retain the capacity for spine induction in response to estrogen. These data have important implications for cognitive effects of estrogen replacement in postmenopausal women and demonstrate that an estrogen replacement protocol that mimics normal physiological cycles with timed, intermittent peaks can have profound neurobiological effects.


Assuntos
Dendritos/efeitos dos fármacos , Estrogênios/farmacologia , Hipocampo/efeitos dos fármacos , Macaca mulatta/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fatores Etários , Animais , Biomarcadores , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/fisiopatologia , Dendritos/metabolismo , Dendritos/ultraestrutura , Giro Denteado/citologia , Giro Denteado/efeitos dos fármacos , Giro Denteado/metabolismo , Esquema de Medicação , Terapia de Reposição de Estrogênios , Estrogênios/metabolismo , Estrogênios/uso terapêutico , Ciclo Estral/efeitos dos fármacos , Ciclo Estral/fisiologia , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Imuno-Histoquímica , Macaca mulatta/anatomia & histologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/metabolismo , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ovariectomia , Pós-Menopausa/metabolismo
18.
J Neurosci ; 23(13): 5708-14, 2003 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-12843274

RESUMO

Among the identified risks and benefits of hormone-replacement therapy, the effects of treatment on cognitive function in postmenopausal women have proved difficult to define. Here we conducted a controlled, prospective analysis in a nonhuman primate model to test whether surgical menopause and estrogen replacement influence the cognitive outcome of normal aging. Sixteen aged rhesus monkeys were ovariectomized, and throughout the course of subsequent neuropsychological assessment, half received a regimen of low-dose, cyclic estradiol replacement. Hormone treatment substantially reversed the marked age-related impairment vehicle-injected monkeys exhibited on a delayed response test of spatial working memory. Modest improvement was also observed on a delayed nonmatching-to-sample recognition memory task. In contrast, ovariectomy exacerbated age-related deficits in object discrimination learning; the magnitude of this effect was equivalent among vehicle- and estrogen-treated monkeys. Together, these results demonstrate that ovarian hormone status can broadly influence normal cognitive aging in monkeys, affecting capacities mediated by multiple brain regions, including the prefrontal cortex and the medial temporal lobe memory system. The animal model established here should enable progress toward defining the neurobiological mechanisms that mediate the beneficial effects of estrogen on age-related cognitive decline in primates.


Assuntos
Cognição/efeitos dos fármacos , Terapia de Reposição de Estrogênios , Macaca mulatta/fisiologia , Periodicidade , Envelhecimento/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Comportamento de Escolha/efeitos dos fármacos , Comportamento de Escolha/fisiologia , Cognição/fisiologia , Aprendizagem por Discriminação/efeitos dos fármacos , Esquema de Medicação , Estradiol/administração & dosagem , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Memória de Curto Prazo/efeitos dos fármacos , Modelos Animais , Testes Neuropsicológicos , Ovariectomia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Lobo Temporal/efeitos dos fármacos , Lobo Temporal/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA