Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Brain Behav Immun ; 88: 699-710, 2020 08.
Article in English | MEDLINE | ID: mdl-32387511

ABSTRACT

OBJECTIVE: Exercise is one of the most potent strategies available to support cognitive health with age, yet substantial variability exists. Sexual dimorphism is evident for brain and immune functioning, the latter being implicated as important pathway for exercise. We examined the moderating role of sex on the relationship between physical activity and systemic inflammatory and brain health outcomes in support of more personalized approaches to behavioral interventions. METHODS: Our discovery cohort included 45 typically aging women matched on age (±5y) and education (±2y) to 45 men (mean age = 72.5; Clinical Dementia Rating = 0) who completed self-reported current physical activity (Physical Activity Scale for Elderly), blood draw, neuropsychological evaluation, and brain MRI. An independent sample of 45 typically aging women and 36 men who completed the same measures comprised a replication cohort. Plasma was analyzed for 11 proinflammatory cytokine and chemokine markers via MesoScale Discovery. RESULTS: Discovery cohort: Reported physical activity did not differ between sexes (150 vs. 157, p = 0.72). There was a significant interaction between sex and physical activity on chemokine markers MDC, MIP-1b, MCP-4, and eotaxin-3 (ps < 0.03), with a similar trend for MCP-1 and INFγ (ps < 0.09). Men who reported greater activity demonstrated lower inflammatory markers, an effect attenuated-to-absent in women. An interaction between sex and physical activity was also observed for parahippocampal volumes (p = 0.02) and cognition (processing speed and visual memory; ps < 0.04). Again, the beneficial effect of physical activity on outcomes was present in men, but not women. Replication cohort analyses conferred a consistent effect of sex on the relationship between physical activity and immune markers; models examining neurobehavioral outcomes did not strongly replicate. Across cohorts, post-hoc models demonstrated an interaction between sex and activity-related inflammatory markers on total gray matter volume and visual memory. Men with higher inflammatory markers demonstrated poorer brain structure and function, whereas inflammatory markers did not strongly relate to neurobehavioral outcomes in women. CONCLUSIONS: Greater physical activity was associated with lower markers of inflammation in clinically normal older men, but not women - an effect consistently replicated across cohorts. Additionally, men appeared disproportionately vulnerable to the adverse effects of peripheral inflammatory markers on brain structure and function compared to women. Immune activation may be a male-specific pathway through which exercise confers neurobehavioral benefit.


Subject(s)
Cognitive Aging , Exercise , Sex Characteristics , Aged , Aging , Brain/diagnostic imaging , Female , Humans , Male
2.
Transl Psychiatry ; 5: e585, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26080320

ABSTRACT

Chronic psychological stress is associated with accelerated aging and premature morbidity and mortality; however, the biology linking chronic psychological stress and its maladaptive effects remains largely unknown. Klotho is a pleiotropic hormone that regulates the aging process and promotes better brain and body health. Whether klotho is linked to psychosocial stress or its negative impact in humans has not been investigated. To address this gap, we recruited 178 healthy women who were either chronically high-stress maternal caregivers for a child with autism spectrum disorder (n = 90) or low-stress control mothers of a typically developing child (n = 88). We found that women under high chronic stress displayed significantly lower levels of the longevity hormone klotho compared with low-stress controls (t(176) = 2.92, P = 0.004; d = 0.44), and the decrease among those under high stress was age-dependent. In addition, high-stress caregivers who reported more depressive symptoms displayed even lower klotho levels compared with low-stress participants. These findings provide the first evidence that klotho levels are sensitive to psychosocial stressors and raise the possibility that klotho may serve as a novel biological link connecting stress, depression and risk for accelerated disease development. Furthermore, these findings have important implications for understanding the plasticity of the aging process and may represent a therapeutic target for mitigating the deleterious effects of chronic psychological stress on health and well-being.


Subject(s)
Depression/blood , Glucuronidase/blood , Longevity , Mothers/psychology , Stress, Psychological/blood , Adult , Age Factors , Antidepressive Agents/therapeutic use , Autism Spectrum Disorder , Cohort Studies , Depression/drug therapy , Depression/psychology , Female , Humans , Klotho Proteins , Middle Aged , Stress, Psychological/psychology
3.
J Neuroendocrinol ; 18(2): 97-103, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16420278

ABSTRACT

Activins are members of the transforming growth factor-beta superfamily that exert neurotrophic and neuroprotective effects on various neuronal populations. To determine the possible function of activin in stroke injury, we assessed which components of the activin signalling pathway were modulated in response to middle cerebral artery occlusion (MCAO). Furthermore, because oestradiol replacement protects against MCAO-induced cell death, we explored whether oestradiol replacement influences activin gene expression. Female Sprague-Dawley rats underwent permanent MCAO and the expression of activins and their corresponding receptors was determined by semiquantitative reverse transcriptase-polymerase chain reaction at 24 h after onset of ischaemia. We observed up-regulation of activin betaA and activin type I receptor A mRNA in response to injury. Dual-label immunocytochemistry followed by confocal z-stack analysis showed that the activin A expressing cells comprised neurones. Next, we monitored the time course of activin betaA mRNA expression in oestradiol- or vehicle-treated rats at 4, 8, 16 and 24 h after MCAO via in situ hybridisation. Starting at 4 h after injury, activin betaA mRNA was up-regulated in cortical and striatal areas in the ipsilateral hemisphere. Activin betaA mRNA levels in the cortex increased dramatically with time and were highest at 24 h after the insult, and oestradiol replacement did not influence this increase.


Subject(s)
Activin Receptors, Type I/metabolism , Estradiol/physiology , Gene Expression Regulation , Infarction, Middle Cerebral Artery/metabolism , Inhibin-beta Subunits/genetics , Neurons/metabolism , Activin Receptors, Type I/genetics , Analysis of Variance , Animals , Brain Damage, Chronic/genetics , Brain Damage, Chronic/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Estradiol/administration & dosage , Female , Gene Expression Regulation/physiology , Infarction, Middle Cerebral Artery/genetics , Inhibin-beta Subunits/metabolism , Neostriatum/metabolism , Neostriatum/pathology , Neurons/pathology , RNA, Messenger/analysis , Rats , Signal Transduction/physiology , Tissue Distribution
4.
Brain Res Brain Res Rev ; 37(1-3): 313-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11744096

ABSTRACT

We have shown that 17beta-estradiol exerts profound protective effects against stroke-like ischemic injury in female rats. These effects are evident using physiological levels of estradiol replacement in ovariectomized rats and require hormone treatment prior to the time of injury. The protective actions of estradiol appear to be most prominent in the cerebral cortex, where cell death is not apparent until at least 4 h after the initiation of ischemic injury and where cell death is thought to be apoptotic in nature. Middle-aged rats remain equally responsive to the protective actions of estradiol. The maintenance of responsiveness of the cerebral cortex to the neuroprotective actions of estradiol was unexpected since responsiveness of the hypothalamus to estradiol decreases dramatically by the time animals are middle-aged. We believe that the protective actions of estradiol require the estrogen receptor-alpha, since estradiol does not protect in estrogen receptor-alpha knockout mice. We have also implemented a method of culturing cerebral cortical explants to assess the protective effects of estradiol in vitro. This model exhibits remarkable parallelisms with our in vivo model of brain injury. We have found that 17beta-estradiol decreases the extent of cell death and that this protective effect requires hormone pretreatment. Finally, 17alpha-estradiol, which does not interact effectively with the estrogen receptor, does not protect; and addition of ICI 182,780, an estrogen receptor antagonist, blocks the protective actions of estradiol. We have begun to explore the molecular and cellular mechanisms of estradiol-mediated protection. In summary, our findings demonstrate that estradiol exerts powerful protective effects both in vivo and in vitro and suggest that these actions are mediated by estrogen receptors.


Subject(s)
Brain/metabolism , Cytoprotection/physiology , Estradiol/metabolism , Neuroprotective Agents/metabolism , Aging/metabolism , Animals , Brain/cytology , Brain/drug effects , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Cells, Cultured , Cytoprotection/drug effects , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha , Female , Hypoglycemia/drug therapy , Hypoglycemia/metabolism , Neuroprotective Agents/pharmacology , Rats , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism
5.
Endocrinology ; 142(3): 969-73, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181507

ABSTRACT

An accumulating body of evidence clearly establishes that estradiol is a potent neuroprotective and neurotrophic factor in the adult: it influences memory and cognition, decreases the risk and delays the onset of neurological diseases such as Alzheimer's disease, and attenuates the extent of cell death that results from brain injuries such as cerebrovascular stroke and neurotrauma. Thus, estradiol appears to act at two levels: 1) it decreases the risk of disease or injury; and/or 2) it decreases the extent of injury incurred by suppressing the neurotoxic stimulus itself or increasing the resilience of the brain to a given injury. During the past century, the average life span of women has increased dramatically, whereas the time of the menopause has remained essentially constant. Thus, more women will live a larger fraction of their lives in a postmenopausal, hypoestrogenic state than ever before. Clearly, it is critical for us understand the circumstances under which estradiol exerts protective actions and the cellular and molecular mechanisms that underlie these novel, nonreproductive actions.


Subject(s)
Brain/drug effects , Estradiol/pharmacology , Neuroprotective Agents/pharmacology , Animals , Brain/metabolism , Humans , Receptors, Estrogen/physiology
6.
Proc Natl Acad Sci U S A ; 98(4): 1952-7, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11172057

ABSTRACT

Estradiol protects against brain injury, neurodegeneration, and cognitive decline. Our previous work demonstrates that physiological levels of estradiol protect against stroke injury and that this protection may be mediated through receptor-dependent alterations of gene expression. In this report, we tested the hypothesis that estrogen receptors play a pivotal role in mediating neuroprotective actions of estradiol and dissected the potential biological roles of each estrogen receptor (ER) subtype, ER alpha and ER beta, in the injured brain. To investigate and delineate these mechanisms, we used ER alpha-knockout (ER alpha KO) and ER beta-knockout (ER beta KO) mice in an animal model of stroke. We performed our studies by using a controlled endocrine paradigm, because endogenous levels of estradiol differ dramatically among ER alpha KO, ER beta KO, and wild-type mice. We ovariectomized ER alpha KO, ER beta KO, and the respective wild-type mice and implanted them with capsules filled with oil (vehicle) or a dose of 17 beta-estradiol that produces physiological hormone levels in serum. One week later, mice underwent ischemia. Our results demonstrate that deletion of ER alpha completely abolishes the protective actions of estradiol in all regions of the brain; whereas the ability of estradiol to protect against brain injury is totally preserved in the absence of ER beta. Thus, our results clearly establish that the ER alpha subtype is a critical mechanistic link in mediating the protective effects of physiological levels of estradiol in brain injury. Our discovery that ER alpha mediates protection of the brain carries far-reaching implications for the selective targeting of ERs in the treatment and prevention of neural dysfunction associated with normal aging or brain injury.


Subject(s)
Brain Injuries/prevention & control , Estradiol/physiology , Receptors, Estrogen/physiology , Animals , Brain/blood supply , Brain/pathology , Brain Injuries/pathology , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Cerebrovascular Circulation , Estradiol/blood , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Hormone Replacement Therapy , Mice , Mice, Inbred C57BL , Stroke/pathology , Stroke/prevention & control
7.
Front Neuroendocrinol ; 22(1): 33-66, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11141318

ABSTRACT

Our appreciation that estrogens are important neurotrophic and neuroprotective factors has grown rapidly. Although a thorough understanding of the molecular and cellular mechanisms that underlie this effect requires further investigation, significant progress has been made due to the availability of animal models in which we can test potential candidates. It appears that estradiol can act via mechanisms that require classical intracellular receptors (estrogen receptor alpha or beta) that affect transcription, via mechanisms that include cross-talk between estrogen receptors and second messenger pathways, and/or via mechanisms that may involve membrane receptors or channels. This area of research demands attention since estradiol may be an important therapeutic agent in the maintenance of normal neural function during aging and after injury.


Subject(s)
Brain/physiology , Estradiol/physiology , Nerve Growth Factors/physiology , Neuroprotective Agents/metabolism , Animals , Apoptosis/genetics , Humans , Learning/physiology , Memory/physiology , Receptors, Estrogen/physiology , Second Messenger Systems/physiology , Transcription, Genetic/physiology
8.
Endocrinology ; 142(1): 43-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145565

ABSTRACT

Estrogen replacement therapy in postmenopausal women ameliorates cognitive dysfunction and decreases the risk and/or severity of neurodegenerative conditions such as Alzheimer's disease and stroke. Furthermore, estradiol exerts neuroprotective effects in a variety of in vitro and in vivo models of brain injury. We have previously shown that physiological levels of estradiol attenuate ischemic brain injury in young female rats. However, neurodegenerative events occur more frequently in elderly women who are chronically hypoestrogenic. Therefore, we investigated whether aging rats remain responsive to the neuroprotective actions of estradiol. Young (3-4 months) and middle-aged (9-12 months) rats were ovariectomized and treated for 1 week with estradiol before middle cerebral artery occlusion (MCAO). Regional cerebral blood flow was monitored in some animals at the time of injury. Brains were collected 24 h after MCAO and infarct volume was analyzed. Our data demonstrate that in both young and aging rats, low and high physiological doses of estradiol decrease ischemic injury by almost 50%, compared with oil-treated controls. Additionally, our data suggest that estradiol acts in both age groups via blood flow-independent mechanisms, as basal and postinjury blood flow was equivalent between estradiol- and oil-treated young and aging rats. These data demonstrate that replacement with physiological levels of estradiol protects against stroke-related injury in young and aging female rats and strongly suggest that older animals remain responsive to the protective actions of estradiol.


Subject(s)
Aging/physiology , Brain/pathology , Cerebrovascular Circulation/physiology , Estradiol/pharmacology , Ischemic Attack, Transient/physiopathology , Myocardial Infarction/prevention & control , Myocardial Infarction/physiopathology , Neuroprotective Agents , Animals , Brain/drug effects , Brain/growth & development , Cerebrovascular Circulation/drug effects , Estrogen Replacement Therapy , Female , Humans , Ischemic Attack, Transient/prevention & control , Models, Animal , Myocardial Infarction/pathology , Ovariectomy , Postmenopause , Rats , Rats, Sprague-Dawley
9.
Biol Reprod ; 63(4): 982-5, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10993817

ABSTRACT

Several clinical studies suggest that estradiol acts as a potent growth and protective factor in the adult brain. Postmenopausal women experience permanent hypoestrogenicity and suffer from increased risk of brain injury associated with neurodegenerative diseases such as stroke and Alzheimer's disease. Estrogen replacement therapy appears to decrease the risk and severity of these neurodegenerative conditions. Studies using animal models have shown that estradiol exerts similar effects in rodents and can enhance cell survival and induce synaptic plasticity. Therefore, we undertook studies to assess whether estradiol treatment can decrease brain injury and cell death induced by an experimental model of ischemia and whether aging animals remain responsive to the protective effects of estradiol. We will review results from recent studies that demonstrate that 1) in young animals, estrogens exert profound protective effects against ischemic brain injury induced by cerebral artery occlusion and 2) the response of aging animals has been tested with varying results. We will discuss and compare our experimental findings that utilize a permanent cerebral artery occlusion model and physiological levels of estradiol replacement therapy in young and middle-aged rats with those of previous studies. These observations provide important insights into the potential protective actions of estrogen replacement therapy on age- and disease-related processes in the brain.


Subject(s)
Aging/physiology , Brain Ischemia/prevention & control , Brain Ischemia/physiopathology , Estradiol/physiology , Animals , Estradiol/pharmacology , Female , Ovariectomy , Rats , Rats, Sprague-Dawley
10.
Brain Res ; 873(2): 235-42, 2000 Aug 11.
Article in English | MEDLINE | ID: mdl-10930549

ABSTRACT

Estradiol has been shown to exert trophic and protective actions in the brain. Our laboratory has shown that in vivo, low physiological levels of estradiol protect the female rat brain against ischemic injury. In the present study, we used organotypic cortical explant cultures to begin to decipher the mechanisms of estradiol's actions. Injury was induced by exposure to kainic acid or potassium cyanide/2-deoxyglucose (KCN/2-DG) for varying lengths of time, and cell death was monitored by LDH release at 2, 6, 12, 24, 48, 72 and 96 h after injury. We found that exposure to 1 mM KCN/2 mM 2-DG for 2 h produced consistent delayed cell death that was detectable by 24 h. The presence of 17beta-estradiol (E2) during the 7 days prior to injury significantly reduced the extent of cell death; whereas, administration of E2 at the time of injury did not protect. The protective effects of estradiol were dose dependent. Low doses of E2 (1, 10, and 30 nM) significantly reduced cell death; however, higher concentrations of E2 (>60 nM) had no protective effect. The observations that low levels of E2 protect against cell death, and that pretreatment is required suggest that the protective actions of estradiol may involve estrogen receptors. Therefore, we examined the ability of 17alpha-estradiol, which does not efficiently activate the estrogen receptor, and the addition of the estrogen receptor antagonist, ICI 182,780, to influence the extent of cell death induced by KCN/2-DG. 17alpha-Estradiol failed to protect, and ICI 182,780 prevented E2 from protecting against cell death. Furthermore, E2 pretreatment is required for more than 24 h to be neuroprotective. Our results clearly show that in cortical explant cultures, estradiol protects cells against ischemic injury, and suggest that these protective actions involve estrogen receptors.


Subject(s)
Cell Death/drug effects , Cerebral Cortex/drug effects , Estradiol/pharmacology , Neuroprotective Agents/pharmacology , Receptors, Estrogen/drug effects , Animals , Animals, Newborn , Biomarkers , Cell Death/physiology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Deoxyglucose/pharmacology , Drug Combinations , Estradiol/metabolism , Female , Kainic Acid/pharmacology , Neuroprotective Agents/metabolism , Neurotoxins/pharmacology , Organ Culture Techniques , Potassium Cyanide/pharmacology , Pregnancy , Propidium , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism
11.
J Neurocytol ; 29(5-6): 401-10, 2000.
Article in English | MEDLINE | ID: mdl-11424956

ABSTRACT

Many clinical studies suggest that estrogen enhances memory and cognition and protects against neurodegenerative diseases and injury associated with stroke or stress. These results are strongly supported by experiments performed in animal models using both in vivo and in vitro methods. We present here data from our lab that establishes that physiological levels of estradiol exert profound protective actions against ischemic injury. Further we will present evidence that these effects may be mediated through estrogen receptors that may influence the bcl-2 family of genes.


Subject(s)
Brain Injuries/metabolism , Estradiol/metabolism , Neuroprotective Agents/metabolism , Animals , Disease Models, Animal , In Vitro Techniques
12.
J Neurosci ; 19(15): 6385-93, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10414967

ABSTRACT

We have shown that physiological levels of estradiol exert profound protective effects on the cerebral cortex in ischemia induced by permanent middle cerebral artery occlusion. The major goal of this study was to begin to elucidate potential mechanisms of estradiol action in injury. Bcl-2 is a proto-oncogene that promotes cell survival in a variety of tissues including the brain. Because estradiol is known to promote cell survival via Bcl-2 in non-neural tissues, we tested the hypothesis that estradiol decreases cell death by influencing bcl-2 expression in ischemic brain injury. Furthermore, because estradiol may protect the brain through estrogen receptor-mediated mechanisms, we examined expression of both receptor subtypes ERalpha and ERbeta in the normal and injured brain. We analyzed gene expression by RT-PCR in microdissected regions of the cerebral cortex obtained from injured and sham female rats treated with estradiol or oil. We found that estradiol prevented the injury-induced downregulation of bcl-2 expression. This effect was specific to bcl-2, as expression of other members of the bcl-2 family (bax, bcl-x(L), bcl-x(S), and bad) was unaffected by estradiol treatment. We also found that estrogen receptors were differentially modulated in injury, with ERbeta expression paralleling bcl-2 expression. Finally, we provide the first evidence of functional ERbeta protein that is capable of binding ligand within the region of the cortex where estradiol-mediated neuroprotection was observed in cerebral ischemia. These findings indicate that estradiol modulates the expression of bcl-2 in ischemic injury. Furthermore, our data suggest that estrogen receptors may be involved in hormone-mediated neuroprotection.


Subject(s)
Brain Ischemia/metabolism , Estradiol/pharmacology , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Estrogen/physiology , Animals , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Ischemia/genetics , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Death/drug effects , Female , Gene Expression/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Sprague-Dawley
13.
Endocr Rev ; 20(3): 243-8, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10368769

ABSTRACT

In summary, the evidence that both the ovary and the brain are key pacemakers in the menopause is compelling. Our appreciation that estrogens are important neurotrophic and neuroprotective factors has grown rapidly. Future studies will allow us to better understand the ensemble of factors that interact to maintain regular reproductive cyclicity and how this precise dynamic balance changes with age. Furthermore, understanding how estrogen exerts trophic and protective actions should lead to its use as an important therapeutic agent in the maintenance of normal neural function during aging and after injury.


Subject(s)
Menopause/physiology , Neurosecretory Systems/physiology , Aging , Brain/physiology , Female , Hormones/physiology , Humans , Middle Aged , Ovary/physiology
14.
J Alzheimers Dis ; 1(4-5): 265-74, 1999 Nov.
Article in English | MEDLINE | ID: mdl-12214124

ABSTRACT

In recent years our appreciation that estradiol is truly a pleiotropic hormone has grown dramatically. We will review the findings that suggest that estrogens may exert important non-reproductive actions on the brain. These studies provide important insights into the clinical effects of estrogen replacement therapy on age- and disease-related processes in the brain. We will also discuss the multiple cellular and molecular mechanisms that may underlie estradiol's neurotrophic and neuroprotective effects.

15.
J Cereb Blood Flow Metab ; 18(11): 1253-8, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9809515

ABSTRACT

Clinical studies demonstrate that estrogen replacement therapy in postmenopausal women may enhance cognitive function and reduce neurodegeneration associated with Alzheimer's disease and stroke. This study assesses whether physiologic levels of estradiol prevent brain injury in an in vivo model of permanent focal ischemia. Sprague-Dawley rats were ovariectomized; they then were implanted, immediately or at the onset of ischemia, with capsules that produced physiologically low or physiologically high 17beta-estradiol levels in serum (10 or 60 pg/mL, respectively). One week after ovariectomy, ischemia was induced. Estradiol pretreatment significantly reduced overall infarct volume compared with oil-pretreated controls (mean+/-SD: oil = 241+/-88; low = 139+/-91; high = 132+/-88 mm3); this protective effect was regionally specific to the cortex, since no protection was observed in the striatum. Baseline and ischemic regional CBF did not differ between oil and estradiol pretreated rats, as measured by laser Doppler flowmetry. Acute estradiol treatment did not protect against ischemic injury. Our finding that estradiol pretreatment reduces injury demonstrates that physiologic levels of estradiol can protect against neurodegeneration.


Subject(s)
Brain Ischemia/physiopathology , Cerebral Infarction/prevention & control , Cerebrovascular Circulation/physiology , Estradiol/therapeutic use , Ischemic Attack, Transient/prevention & control , Ischemic Attack, Transient/physiopathology , Animals , Brain/blood supply , Brain Ischemia/prevention & control , Cerebral Infarction/physiopathology , Cerebrovascular Circulation/drug effects , Drug Implants , Estradiol/administration & dosage , Estradiol/blood , Estrogen Replacement Therapy , Female , Humans , Laser-Doppler Flowmetry , Organ Specificity , Ovariectomy , Postmenopause , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...