451
|
Nakamura T, Xi G, Hua Y, Schallert T, Hoff JT, Keep RF. Intracerebral hemorrhage in mice: model characterization and application for genetically modified mice. J Cereb Blood Flow Metab 2004; 24:487-94. [PMID: 15129180 DOI: 10.1097/00004647-200405000-00002] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Gene knockout or transgenic animals may assist in elucidating the mechanisms of brain injury after intracerebral hemorrhage (ICH). However, almost all commercially available transgenic or knockout animals are mice. The purpose of this study was to develop an ICH model in mice and to investigate the influence of gender and complement C5 genetic differences on outcome after ICH. Male and female C57BL/6 mice and C5-deficient and -sufficient control mice were anesthetized and then received an injection of 30 microL autologous whole blood into the right basal ganglia. Brain water content was studied at 1 and 3 days after ICH. Behavioral tests (fore-limb use asymmetry and corner turn test) were performed at 1, 3, 7, 14, 21, or 28 days after ICH. In male mice, brain water content was significantly increased in the ipsilateral basal ganglia 1 and 3 days after ICH, compared with saline injection controls (P < 0.01). There were marked neurological deficits 1 and 3 days after ICH, with progressive recovery over 28 days. In contrast, although brain edema and behavioral deficits were similar at 1 day after ICH in female and male mice, female mice showed reduced edema at 3 days and a faster recovery of behavioral deficits after ICH. 17 beta-estradiol treatment in male mice markedly reduced ICH-induced edema (P < 0.01). Brain water content was significantly increased in C5-deficient mice compared with C5-sufficient at 3 days after ICH (P < 0.05). These findings suggest that the mouse ICH model is a reproducible and feasible model. These results also suggest that gender and complement C5 are factors affecting brain injury after ICH.
Collapse
Affiliation(s)
- Takehiro Nakamura
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
452
|
Merchenthaler I, Dellovade TL, Shughrue PJ. Neuroprotection by estrogen in animal models of global and focal ischemia. Ann N Y Acad Sci 2004; 1007:89-100. [PMID: 14993043 DOI: 10.1196/annals.1286.009] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Estrogen has been demonstrated to protect against brain injury, neurodegeneration, and cognitive decline. Furthermore, estrogen seems to specifically protect cortical and hippocampal neurons from ischemic injury. Here our data evaluating the neuroprotective effects of estrogens, the selective estrogen receptor modulators (SERMs), and estrogen receptor alpha- and beta-selective ligands in animal models of ischemic injury are discussed. In rats and mice, the middle cerebral artery occlusion (MCAO) model was used as models representing cerebrovascular stroke, while in gerbils the two-vessel occlusion model, resenting acute heart attack, was used. Using focal ischemia in ovariectomized ERalphaKO, ERbetaKO, and wild-type mice, we clearly established that the ERalpha subtype is the critical ER-mediating neuroprotection in mouse focal ischemia. Because of the characteristic blood supply of the gerbil, the gerbil global ischemia model was used to evaluate the neuroprotective effects of estrogen, SERMs, and ERalpha- and ERbeta-selective compounds in the hippocampus. Analysis of neurogranin mRNA, a marker of viability of hippocampal neurons, with in situ hybridization, revealed that estrogen treatment resulted in a complete protection in the CA1 regions not only when administered before, but also when given 1 hour after occlusion. Our in vivo binding studies with (125)I-estrogen in gerbils revealed the presence of nuclear estrogen binding sites primarily in CA1 neurons, but not in the CA3 region, as we saw in rats and mice. Together, these observations demonstrate that estrogen protects from ischemic injury in both the focal and global ischemia models by acting primarily via classical nuclear receptors.
Collapse
Affiliation(s)
- Istvan Merchenthaler
- Women's Health Research Institute, Wyeth Research, Collegeville, Pennsylvania 19526, USA.
| | | | | |
Collapse
|
453
|
Yang SH, Liu R, Wu SS, Simpkins JW. The use of estrogens and related compounds in the treatment of damage from cerebral ischemia. Ann N Y Acad Sci 2004; 1007:101-7. [PMID: 14993044 DOI: 10.1196/annals.1286.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
There are 750,000 new cases of stroke each year in the United States, and brain damage from stroke leads to high health care costs and disabilities. Needed, but currently not available, are therapies that can be administered prior to, during, or after cerebral ischemia that reduce or eliminate neuronal damage from stroke. To address this issue, we began to assess the neuroprotective effects of estrogens and related compounds in stroke neuroprotection to determine whether these compounds had potential for clinical application. First, we demonstrated that 17 beta-estradiol (E2) pretreatment exerted potent neuroprotection of the cerebral cortex over a wide dose range and pretreatment interval. Thereafter, we assessed the ability of a variety of non-feminizing estrogens to protect brain tissue from stroke. We observed that pretreatment with 17 alpha-estradiol, the complete enantiomer of E2 (ENT-E2), 2-adamantylestrone, and the enantiomer of 17-desoxyestradiol, were as effective as E2 in pretreatment protection from stroke damage. These data suggest that non-estrogen receptor mechanisms are involved in brain neuroprotection under our treatment conditions. We then determined whether the observed E2 protection could be extended to times after the onset of the cerebral ischemic event. Using a formulation of E2 that rapidly delivers the steroid, a necessary condition for acute therapy of an ongoing stroke, we demonstrated that 100 mg E2/kg could protect brain tissue for up to 3 h after the onset of the stroke. To determine whether this therapeutic window could be extended with higher doses of the steroid, we conducted a dose-response assessment of E2 when administered at 6 h after the onset of the ischemic event. While the effectiveness of the 100 micro g E2/kg was reduced at this time interval, higher doses of E2 were effective. E2, at doses of 500 and 1000 micro g/kg, reduced infarct volume by more than 50%, even with this 6-h delay in treatment. Collectively, these data indicate that estrogens could prove to be useful therapies in preventing brain damage from strokes.
Collapse
Affiliation(s)
- Shao-Hua Yang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | | | | | | |
Collapse
|
454
|
Morrison WE, Arbelaez JJ, Fackler JC, De Maio A, Paidas CN. Gender and age effects on outcome after pediatric traumatic brain injury. Pediatr Crit Care Med 2004; 5:145-51. [PMID: 14987344 DOI: 10.1097/01.pcc.0000112373.71645.2a] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To evaluate whether girls have better outcomes after traumatic brain injury than boys. DESIGN Retrospective cohort study. SETTING University hospital. PATIENTS A 16,586 patient subset of the National Pediatric Trauma Registry with nonpenetrating traumatic brain injury. INTERVENTIONS Retrospective review. MEASUREMENTS AND MAIN RESULTS The patients were subdivided by age into prepubertal (0-7 yrs), indeterminate pubertal (8-12 yrs), and probable pubertal (13-19 yrs). All analyses were adjusted for injury severity using the Injury Severity Score. Outcome variables were in-hospital death rate, intensive care unit length of stay, total length of stay, discharge to home vs. rehabilitation, and functional status at discharge. Overall, 6.1% of girls and 5.3% of boys died. A higher proportion of girls were injured in motor vehicle crashes. Gender did not have a significant effect on in-hospital mortality rate after adjustment for age, Injury Severity Score, and motor vehicle crashes. Boys had a shorter intensive care unit length of stay (p =.027). There were no statistically significant differences between boys and girls in total hospital length of stay, functional outcome, and discharge location, although for every outcome there was a trend toward girls doing worse. CONCLUSIONS There is evidence from this large study that girls do not have a better outcome after pediatric traumatic brain injury than boys, with a suggestion that girls may do worse.
Collapse
|
455
|
Carswell HV, Bingham D, Wallace K, Nilsen M, Graham DI, Dominiczak AF, Macrae IM. Differential effects of 17beta-estradiol upon stroke damage in stroke prone and normotensive rats. J Cereb Blood Flow Metab 2004; 24:298-304. [PMID: 15091110 DOI: 10.1097/01.wcb.0000112322.75217.fd] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We previously reported that during pro-estrus (high endogenous estrogen levels), brain damage after middle cerebral artery occlusion (MCAO) was reduced in stroke-prone spontaneously hypertensive rats (SHRSP) but not in normotensive Wistar Kyoto rat (WKY). In the present study, we examined the effect of exogenous estrogen on brain damage after MCAO in SHRSP and WKY. A 17beta-estradiol (0.025 mg or 0.25 mg, 21 day release) or matching placebo pellet was implanted into ovariectomized WKY and SHRSP (3 to 4 months old) who then underwent distal diathermy-induced MCAO 2 weeks later. Plasma 17beta-estradiol levels for placebo and 17beta-estradiol groups were as follows: WKY 0.025 mg 16.4 +/- 8.5 (pg/mL, mean +/- SD) and 25.85 +/- 12.6; WKY 0.25 mg 18.2 +/- 9.0 and 69.8 +/- 27.4; SHRSP 0.25 mg 20.7 +/- 8.8 and 81.0 +/- 16.9. In SHRSP, infarct volumes at 24 hours after MCAO were similar in placebo and 17beta-estradiol groups: SHRSP 0.025 mg 126.7 +/- 15.3 mm (n = 6) and 114.0 +/- 14.1 mm (n = 8) (not significant); SHRSP 0.25 mg 113.5 +/- 22.3 mm (n = 8) and 129.7 +/- 26.2 mm (n = 7) (not significant), respectively. In WKY, 17beta-estradiol significantly increased infarct volume by 65% with 0.025 mg dose [36.1 +/- 20.7 mm (n = 8) and 59.7 +/- 19.3 mm (n = 8) (P = 0.033, unpaired t-test)] and by 96% with 0.25 mg dose [55.9 +/- 36.4 mm (n = 8) and 109.7 +/- 6.7 mm (n = 4) (P = 0.017)]. Thus, 17beta-estradiol increased stroke damage in normotensive rats with no significant effect in stroke-prone rats. Despite being contrary to our hypothesis, our findings add substance to the recently reported negative effects of 17beta-estradiol in clinical studies.
Collapse
Affiliation(s)
- Hilary V Carswell
- Wellcome Surgical Institute, Division of Clinical Neuroscience, University of Glasgow, Glasgow, Scotland, UK.
| | | | | | | | | | | | | |
Collapse
|
456
|
Yune TY, Kim SJ, Lee SM, Lee YK, Oh YJ, Kim YC, Markelonis GJ, Oh TH. Systemic Administration of 17β-Estradiol Reduces Apoptotic Cell Death and Improves Functional Recovery following Traumatic Spinal Cord Injury in Rats. J Neurotrauma 2004; 21:293-306. [PMID: 15115604 DOI: 10.1089/089771504322972086] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent evidence indicates that estrogen exerts neuroprotective effects in both brain injury and neurodegenerative diseases. We examined the protective effect of estrogen on functional recovery after spinal cord injury (SCI) in rats. 17beta-estradiol (3, 100, or 300 microg/kg) was administered intravenously 1-2 h prior to injury (pre-treatment), and animals were then subjected to a mild, weight-drop spinal cord contusion injury. Estradiol treatment significantly improved hind limb motor function as determined by the Basso-Beattie-Bresnahan (BBB) locomotor open field behavioral rating test. Fifteen to 30 days after SCI, BBB scores were significantly higher in estradiol-treated (100 microg/kg) rats when compared to vehicle-treated rats. Morphological analysis showed that lesion sizes increased progressively in either vehicle-treated or 17beta-estradiol-treated spinal cords. However, in response to treatment with 17beta-estradiol, the lesion size was significantly reduced 18-28 days after SCI when compared to vehicle-treated controls. Terminal deoxynucleotidyl transferase-mediated UTP nickend labeling (TUNEL) staining and DNA gel electrophoresis revealed that apoptotic cell death peaked 24-48 h after injury. Also, SCI induced a marked increase in activated caspase-3 in the spinal cord, evident by 4 h after injury. However, administration of 17beta-estradiol significantly reduced the SCI-induced increase in apoptotic cell death and caspase-3 activity after SCI. Furthermore, 17beta-estradiol significantly increased expression of the anti-apoptotic genes, bcl-2 and bcl-x, after SCI while expression of the pro-apoptotic genes, bad and bax, was not affected by drug treatment. Finally, intravenous administration of 17beta-estradiol (100 microg/kg) immediately after injury (post-treatment) also significantly improved hind limb motor function 19-30 days after SCI compared to vehicle-treated controls. These data suggest that after SCI, 17 beta-estradiol treatment improved functional recovery in the injured rat, in part, by reducing apoptotic cell death.
Collapse
Affiliation(s)
- Tae Y Yune
- Biomedical Research Center, Korea Institute of Science & Technology, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
457
|
Lacerda ALT, Keshavan MS, Hardan AY, Yorbik O, Brambilla P, Sassi RB, Nicoletti M, Mallinger AG, Frank E, Kupfer DJ, Soares JC. Anatomic evaluation of the orbitofrontal cortex in major depressive disorder. Biol Psychiatry 2004; 55:353-8. [PMID: 14960287 DOI: 10.1016/j.biopsych.2003.08.021] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2003] [Revised: 08/12/2003] [Accepted: 08/27/2003] [Indexed: 11/26/2022]
Abstract
BACKGROUND The orbitofrontal cortex (OFC) plays a major role in neuropsychologic functioning including exteroceptive and interoceptive information coding, reward-guided behavior, impulse control, and mood regulation. This study examined the OFC and its subdivisions in patients with MDD and matched healthy control subjects. METHODS Magnetic resonance imaging (MRI) was performed on 31 unmedicated MDD and 34 control subjects matched for age, gender, and race. Gray matter volumes of the OFC and its lateral and medial subdivisions were measured blindly. RESULTS The MDD patients had smaller gray matter volumes in right medial [two-way analysis of covariance F(1,60) = 4.285; p =.043] and left lateral OFC [F(1,60) = 4.252; p =.044]. Left lateral OFC volume correlated negatively with age in patients but not in control subjects. Male, but not female patients exhibited smaller left and right medial OFC volumes compared with healthy control subjects of the same gender. CONCLUSIONS These findings suggest that patients with MDD have reduced OFC gray matter volumes. Although this reduction might be important in understanding the pathophysiology of MDD, its functional and psychopathologic consequences are as yet unclear. Future studies examining the relationship between specific symptomatic dimensions of MDD and OFC volumes could be especially informative.
Collapse
Affiliation(s)
- Acioly L T Lacerda
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
458
|
Fan T, Yang SH, Johnson E, Osteen B, Hayes R, Day AL, Simpkins JW. 17beta-Estradiol extends ischemic thresholds and exerts neuroprotective effects in cerebral subcortex against transient focal cerebral ischemia in rats. Brain Res 2004; 993:10-7. [PMID: 14642826 DOI: 10.1016/j.brainres.2003.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neuroprotective effects of estrogens are demonstrated consistently in the cerebral cortex, but not in subcortical areas. In the present study, transient middle cerebral artery occlusions (MCAO) were induced for various duration, and protective effects of estrogen treatment on the cerebral cortex and subcortex were evaluated. MCAO was induced for 30, 40 or 60 min in ovariectomized rats. Animals were treated with 17beta-estradiol (E2) or vehicle (OVX) 2 h before MCAO and sacrificed 24 h after the indicated duration of MCAO. Ischemic lesion was evaluated by 2,3,5-triphenyltetrazolium chloride staining, hematoxylin and eosin staining, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. E2 treatment reduced the magnitude and delayed the appearance of the total ischemic lesion area and largely prevented TUNEL staining in the cortex. In the subcortex, E2 treatment prevented the ischemic lesion in the 30-min group, reduced lesion area in the 40-min group, but had no effect on ischemic lesion area in the 60-min group. E2 treatment significantly decreased apoptotic cell number in the subcortical area at 30 and 40 min, but not at 60 min of MCAO. This study demonstrated that estrogen treatment can protect the cerebral subcortex in a severity-dependent manner, suggesting that the lack of protective effects of estrogen treatment in the subcortex is not due to the lack of estrogen receptors. Further, this study indicates that estrogens could be used as a neuroprotectant to prolong the therapeutic window of thrombolysis and prolong the time of cerebral circulation intervention for neurosurgical procedure.
Collapse
Affiliation(s)
- Tao Fan
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | | | | | | | | | | | | |
Collapse
|
459
|
Abstract
Current knowledge regarding the pathophysiology of cerebral ischemia and brain trauma indicates that similar mechanisms contribute to loss of cellular integrity and tissue destruction. Mechanisms of cell damage include excitotoxicity, oxidative stress, free radical production, apoptosis and inflammation. Genetic and gender factors have also been shown to be important mediators of pathomechanisms present in both injury settings. However, the fact that these injuries arise from different types of primary insults leads to diverse cellular vulnerability patterns as well as a spectrum of injury processes. Blunt head trauma produces shear forces that result in primary membrane damage to neuronal cell bodies, white matter structures and vascular beds as well as secondary injury mechanisms. Severe cerebral ischemic insults lead to metabolic stress, ionic perturbations, and a complex cascade of biochemical and molecular events ultimately causing neuronal death. Similarities in the pathogenesis of these cerebral injuries may indicate that therapeutic strategies protective following ischemia may also be beneficial after trauma. This review summarizes and contrasts injury mechanisms after ischemia and trauma and discusses neuroprotective strategies that target both types of injuries.
Collapse
Affiliation(s)
- Helen M Bramlett
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Medical School, FL 33101, USA
| | | |
Collapse
|
460
|
McCullough L, Wu L, Haughey N, Liang X, Hand T, Wang Q, Breyer RM, Andreasson K. Neuroprotective function of the PGE2 EP2 receptor in cerebral ischemia. J Neurosci 2004; 24:257-68. [PMID: 14715958 PMCID: PMC6729582 DOI: 10.1523/jneurosci.4485-03.2004] [Citation(s) in RCA: 305] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The cyclooxygenases COX-1 and COX-2 catalyze the first committed step of prostaglandin synthesis from arachidonic acid. Previous studies in rodent stroke models have shown that the inducible COX-2 isoform promotes neuronal injury, and the administration of COX-2 inhibitors reduces infarct volume. We investigated the function of PGE2, a principal prostaglandin product of COX-2 enzymatic activity, in neuronal survival in cerebral ischemia. PGE2 exerts its downstream effects by signaling through a class of four distinct G-protein-coupled EP receptors (for E-prostanoid: EP1, EP2, EP3, and EP4) that have divergent effects on cAMP and phosphoinositol turnover and different anatomical distributions in brain. The EP2 receptor subtype is abundantly expressed in cerebral cortex, striatum, and hippocampus, and is positively coupled to cAMP production. In vitro studies of dispersed neurons and organotypic hippocampal cultures demonstrated that activation of the EP2 receptor was neuroprotective in paradigms of NMDA toxicity and oxygen glucose deprivation. Pharmacologic blockade of EP2 signaling by inhibition of protein kinase A activation reversed this protective effect, suggesting that EP2-mediated neuroprotection is dependent on cAMP signaling. In the middle cerebral artery occlusion-reperfusion model of transient forebrain ischemia, genetic deletion of the EP2 receptor significantly increased cerebral infarction in cerebral cortex and subcortical structures. These studies indicate that activation of the PGE2 EP2 receptor can protect against excitotoxic and anoxic injury in a cAMP-dependent manner. Taken together, these data suggest a novel mechanism of neuroprotection mediated by a dominant PGE2 receptor subtype in brain that may provide a target for therapeutic intervention.
Collapse
Affiliation(s)
- Louise McCullough
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
461
|
Abstract
Estradiol is a known neurotrophic and neuroprotective factor. Our previous work demonstrated that replacement with physiological concentrations of estradiol protects the cortex against middle cerebral artery occlusion (MCAO)-induced cell death. The cerebral cortex exhibits caspase-dependent programmed cell death (PCD) in many models of focal cerebral ischemia. We hypothesized that estradiol attenuates PCD during stroke injury. The current study explored the temporospatial pattern of markers of PCD, their relationship to the evolution of injury, and their modulation by estradiol. Rats were ovariectomized and treated with either estradiol or vehicle. One week later, rats underwent MCAO, and brains were collected at 1, 4, 8, 16, and 24 hr. We assessed the temporospatial evolution of infarction volume, DNA fragmentation, and levels of spectrin cleavage products in ischemic cortex. Estradiol led to a delay and attenuation of injury-mediated DNA fragmentation as early as 8 hr after MCAO. Estradiol also dramatically reduced the level of the 120 kDa caspase-mediated spectrin breakdown product (SBDP120) at 4 hr but not at 8 or 16 hr. The SBDP150, produced by caspase and calpain, showed peak levels at 16 hr but was not altered by estradiol. These results strongly suggest that estradiol protects the ischemic cortex by attenuating PCD, thereby reducing caspase activity, DNA fragmentation, and subsequently, overall cell death. These studies deepen our understanding of the mechanisms underlying estrogen-mediated neuroprotection.
Collapse
|
462
|
Kovacic U, Sketelj J, Bajrović FF. Sex-related difference in collateral sprouting of nociceptive axons after peripheral nerve injury in the rat. Exp Neurol 2004; 184:479-88. [PMID: 14637117 DOI: 10.1016/s0014-4886(03)00269-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Possible sex-related differences in the extent of collateral sprouting of noninjured nociceptive axons after peripheral nerve injury were examined. In the first experiment, peroneal, tibial, and saphenous nerves were transected and ligated in female and male rats. Eight weeks after nerve injury, skin pinch tests revealed that the nociceptive area of the noninjured sural nerve in the instep skin expanded faster in females; the final result was a 30% larger increase in females than in males. In the second experiment, the end-to-side nerve anastomosis was used as a model for axon sprouting. In addition to the previous procedure, the end of an excised peroneal nerve segment was sutured to the side of the intact sural nerve. Eight weeks later, collateral sprouting of nociceptive axons into the anastomosed peroneal nerve segment was assessed by the nerve pinch test and axon counting. There was no significant difference with respect to the percentages of male and female rats with a positive nerve pinch test. The number of myelinated axons in the anastomosed nerve segment was significantly larger in female (456 +/- 217) than in male (202 +/- 150) rats, but the numbers of unmyelinated axons were not significantly different. In normal sural nerves, the numbers of either all myelinated axons or thin myelinated axons did not significantly differ between the two sexes. Therefore, the more extensive collateral axon sprouting observed in female than in male rats is probably due to the higher sprouting capacity of thin myelinated sensory axons in females.
Collapse
Affiliation(s)
- U Kovacic
- Institute of Pathophysiology, Medical Faculty, University of Ljubljana, Zalosbreve;ka 4, 1000, Ljubljana, Slovenia
| | | | | |
Collapse
|
463
|
Bayir H, Marion DW, Puccio AM, Wisniewski SR, Janesko KL, Clark RSB, Kochanek PM. Marked Gender Effect on Lipid Peroxidation after Severe Traumatic Brain Injury in Adult Patients. J Neurotrauma 2004; 21:1-8. [PMID: 14987460 DOI: 10.1089/089771504772695896] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Striking gender differences have been reported in the pathophysiology and outcome of acute neurological injury. Greater neuroprotection in females versus males may be due, in part, to direct and indirect sex hormone-mediated antioxidant mechanisms. Progesterone administration decreases brain levels of F(2)-isoprostane, a marker of lipid peroxidation, after experimental traumatic brain injury (TBI) in male rats, and estrogen is neuroprotective in experimental neurological injury. In this study, we evaluated the effect of gender on lipid peroxidation, as assessed by cerebrospinal fluid (CSF) levels of F(2)-isoprostane, after severe TBI in humans. Lipid peroxidation was assessed in CSF from 68 adults enrolled in two randomized controlled trials evaluating the effect of therapeutic hypothermia after severe TBI (Glasgow coma scale [GCS] score </= 8). Patients treated with hypothermia (n = 41, 12 females, 29 males) were cooled to 32-33 degrees C (within approximately 6 h) for either 24 or 48 h and then re-warmed. F(2)-isoprostane levels were assessed by ELISA in ventricular CSF samples (n = 199) on day 1, 2, and 3. The association between age, GCS score, time, gender, treatment, duration of treatment, core temperature at the time of CSF sampling, secondary hypoxemia, and CSF F(2)-isoprostane level was assessed by multivariate and dichotomous analyses. F(2)-isoprostane was approximately 2-fold higher in males than females (145.8 +/- 39.6 versus 75.4 +/- 16.6 pg/mL, day 1 p = 0.018). An effect of time after injury (p = 0.007) was reflected by a marked early peak in F(2)-isoprostane (day 1). CSF F(2)-isoprostane was also associated with hypoxemia (p = 0.04). Hypothermia tended to decrease F(2)-isoprostane levels only in males on d1 after TBI. To our knowledge, this is the first study showing gender differences in lipid peroxidation after clinical TBI. Lipid peroxidation occurs early after severe TBI in adults and is more prominent in males vs females. These results established that gender is an important consideration in clinical trial design, particularly in the case of antioxidant strategies.
Collapse
Affiliation(s)
- Hülya Bayir
- Safar Center for Resuscitation Research, and Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260, USA
| | | | | | | | | | | | | |
Collapse
|
464
|
Djebaili M, Hoffman SW, Stein DG. Allopregnanolone and progesterone decrease cell death and cognitive deficits after a contusion of the rat pre-frontal cortex. Neuroscience 2004; 123:349-59. [PMID: 14698743 DOI: 10.1016/j.neuroscience.2003.09.023] [Citation(s) in RCA: 206] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We compared the effects of three different doses of allopregnanolone (4, 8 or 16 mg/kg), a metabolite of progesterone, to progesterone (16 mg/kg) in adult rats with controlled cortical impact to the pre-frontal cortex. Injections were given 1 h, 6 h and every day for 5 consecutive days after the injury. One day after injury, both progesterone-treated (16 mg/kg) and allopregnanolone (8 or 16 mg/kg)-treated rats showed less caspase-3 activity, and rats treated with allopregnanolone (16 mg/kg) showed less DNA fragmentation in the lesion area, indicating reduced apoptosis. Nineteen days after the injury, rats treated with progesterone and allopregnanolone (8 or 16 mg/kg) showed no difference in necrotic cavity size but had less cell loss in the medio-dorsal nucleus of the thalamus and less learning and memory impairments compared with the injured vehicle-treated rats. On that same day the injured rats treated with progesterone showed more weight gain compared with the injured rats treated with the vehicle. These results can be taken to show that progesterone and allopregnanolone have similar neuroprotective effects after traumatic brain injury, but allopregnanolone appears to be more potent than progesterone in facilitating CNS repair.
Collapse
Affiliation(s)
- M Djebaili
- Brain Research Laboratory, Department of Emergency Medicine, 1648 Pierce Drive, Cell Biology Building, Room 261, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
465
|
Abstract
Estrogen replacement therapy enhances mood, delays cognitive decline, and reduces the risk of neurodegeneration. Our laboratory has shown previously that pretreatment with low physiological levels of estradiol protects against middle cerebral artery occlusion (MCAO)-induced brain injury during late phases of neuronal cell death. Immediate early genes (IEGs) are induced by various forms of brain injury, and their induction is known to be a critical step in programmed cell death. The current study tested the hypothesis that the ability of estradiol to reduce MCAO-induced cell death involves attenuation of expression of one or more IEGs. We examined the effects of MCAO on the temporospatial pattern of IEG expression and the modulation of this pattern by estradiol replacement. Rats were ovariectomized and treated with either vehicle or low physiological concentrations of estradiol. One week later, rats underwent MCAO and brains were collected 1, 4, 8, 16, and 24 hr later. We assessed IEG mRNAs in discrete regions of brain by RT-PCR at 24 hr. We examined expression of c-Fos mRNA and protein in greater detail using in situ hybridization and immunohistochemistry to delineate the time course and specific regions of cortex in which estradiol influenced its expression. Our results reveal that c-fos, fosB, c-jun, and junB levels were upregulated at 24 hr. Furthermore, estradiol selectively affected the expression of c-Fos mRNA and protein by attenuating the injury-induced increase in a time- and region-specific manner. Our findings strongly suggest that the ability of estradiol to protect against MCAO-induced cell death involves attenuation of c-Fos induction.
Collapse
|
466
|
Krejza J, Siemkowicz J, Sawicka M, Szylak A, Kochanowicz J, Mariak Z, Lewko J, Spektor V, Babikian V, Bert R. Oscillations of cerebrovascular resistance throughout the menstrual cycle in healthy women. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2003; 22:627-632. [PMID: 14689537 DOI: 10.1002/uog.907] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
OBJECTIVES Increased concentration of endogenous estrogen during a typical menstrual cycle has been shown to correlate with augmentation of blood flow through the internal carotid arteries (ICAs), which may be related to changes in vascular resistance within the brain. In this study we investigated the effects of endogenous estrogen and progesterone on cerebrovascular impedance in young healthy women. METHODS The blood flow in the ICA and the common (CCA) and external (ECA) carotid arteries was studied with duplex Doppler sonography. The resistance index (RI) was determined and correlated with plasma 17beta-estradiol concentration in 14 young healthy women throughout their menstrual cycle. RESULTS The concentration of 17beta-estradiol increased in the follicular phase of the cycle and reached a peak on day 14, whereas concentration of progesterone remained low. Along with an increase in estrogen concentration, the ICA RI had decreased from its initial level on average by 9.2% on day 13 and by 6.7% on day 14 (P < 0.05). In contrast, the trend of the ECA RI was to increase during the peak of estrogen concentration. There were no significant changes in the CCA RI or in the systolic blood pressure, heart rate, hematocrit and hemoglobin concentration through the menstrual cycle. CONCLUSIONS Estrogen-related augmentation of blood flow through the ICA is caused mainly by decreased cerebrovascular impedance, as shown by a decrease in the ICA RI. These changes in RI suggest that estrogen influences cerebral impedance mainly by altering the resistance of cerebral microvasculature.
Collapse
Affiliation(s)
- J Krejza
- Department of Radiology, Bialystok Medical Academy, Bialystok, Poland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
467
|
Suzuki T, Bramlett HM, Dietrich WD. The importance of gender on the beneficial effects of posttraumatic hypothermia. Exp Neurol 2003; 184:1017-26. [PMID: 14769396 DOI: 10.1016/s0014-4886(03)00389-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2003] [Revised: 07/09/2003] [Accepted: 07/22/2003] [Indexed: 10/26/2022]
Abstract
The authors studied the importance of gender on the consequences of mild posttraumatic hypothermia following parasagittal fluid-percussion (F-P) brain injury in rats. After traumatic brain injury (TBI), brain temperature was maintained at normothermia (37 degrees C) or reduced to 33 degrees C for 4 h starting 30 min after the insult followed by a 1.5-h slow rewarming period. Animals (n = 48) were allowed to survive for 3 days before quantitative histopathological and immunocytochemical examination. As previously reported, contusion volume in normothermic animals (37 degrees C) was smaller (P < 0.05) in intact females compared to males. In addition, numbers of NeuN-positive cortical neurons were greater in females versus males after TBI. Posttraumatic hypothermia significantly reduced overall contusion volume in males (P < 0.05), while not significantly reducing contusion volume in females. Likewise, hypothermia protected against the loss of cortical neurons in males but had no effect in females. Ovariectomized females showed contusion volumes and neuronal cell counts comparable to those seen in males as well as a significant reduction in contusion volumes and greater neuronal counts following posttraumatic hypothermia. These data are the first to demonstrate that posttraumatic hypothermia (4 h) does not affect short-term histopathological outcomes in female rats. Potential mechanisms underlying this gender difference are discussed. Finally, these experimental findings may have important implications in terms of clinical trials using therapeutic hypothermia targeting patients with central nervous system (CNS) injury.
Collapse
Affiliation(s)
- Takamoto Suzuki
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Neurotrauma Research Center, University of Miami School of Medicine, Miami, FL 33136, USA
| | | | | |
Collapse
|
468
|
Abstract
Female animals are protected from many forms of neurological injury and degeneration relative to their male counterparts, in part attributable to their native estrogens. We hypothesized that estradiol aromatized from precursor androgens via the cytochrome P450 aromatase contributes to ischemic neuroprotection in the female. Female homozygous aromatase knock-out (ArKO) mice and randomly cycling, wild-type (WT) female littermates were treated with reversible middle cerebral artery occlusion (90 min; 22 hr reperfusion). Total and regional ischemic damage was greater in female ArKOs (total, 33.5 +/- 4.8%; cortical, 47.4 +/- 5.7%; striatal, 44.8 +/- 7.8%) compared with WT (total, 14.2 +/- 5%; cortical, 14.2 +/- 4.5%; striatal, 17.5 +/- 8%). Baseline blood pressure and intra-ischemic cortical perfusion were comparable in knock-outs and WT, suggesting that vascular factors do not explain ArKO ischemic sensitivity. Injury was smaller in ovariectomized WT than in ArKO, emphasizing that extragonadal local estradiol plays a critical role in females. Similar increases in cortical and striatal damage were observed in female WT mice chronically treated with the aromatase inhibitor fadrozole compared with vehicle-treated control mice. Restoration of plasma 17beta-estradiol to physiological levels completely reversed the ArKO female's susceptibility to injury. These findings indicate that the biosynthetic enzyme P450 aromatase is key to endogenous neuroprotection in females and suggest that enhancing local, nongonadal estrogen formation could have therapeutic implications is ischemic neuropathology.
Collapse
|
469
|
Culver JP, Durduran T, Furuya D, Cheung C, Greenberg JH, Yodh AG. Diffuse optical tomography of cerebral blood flow, oxygenation, and metabolism in rat during focal ischemia. J Cereb Blood Flow Metab 2003; 23:911-24. [PMID: 12902835 DOI: 10.1097/01.wcb.0000076703.71231.bb] [Citation(s) in RCA: 255] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Diffuse optical tomography (DOT) is an attractive approach for evaluating stroke physiology. It provides hemodynamic and metabolic imaging with unique potential for continuous noninvasive bedside imaging in humans. To date there have been few quantitative spatial-temporal studies of stroke pathophysiology based on diffuse optical signatures. The authors report DOT images of hemodynamic and metabolic contrasts using a rat middle cerebral artery occlusion (MCAO) stroke model. This study used a novel DOT device that concurrently obtains coregistered images of relative cerebral blood volume (rCBV), tissue-averaged hemoglobin oxygen saturation (Sto(2)), and relative cerebral blood flow (rCBF). The authors demonstrate how these hemodynamic measures can be synthesized to calculate an index of the oxygen extraction fraction (OEF) and the cerebral metabolic rate of oxygen consumption (CMRo(2)). Temporary (60-minute) MCAO was performed on five rats. Ischemic changes, averaged over the 60 minutes of occlusion, were as follows: rCBF = 0.42 +/- 0.04, rCBV = 1.02 +/- 0.04, DeltaSto(2) = -11 +/- 2%, rOEF = 1.39 +/- 0.06 and rCMRo(2) = 0.59 +/- 0.07. Although rOEF increased in response to decreased blood flow, rCMRo(2) decreased. The sensitivity of this method of DOT analysis is discussed in terms of assumptions about baseline physiology, and the diffuse optical results are compared with positron emission tomography, magnetic resonance imaging, and histology observations in the literature.
Collapse
Affiliation(s)
- Joseph P Culver
- Department of Physics and Astronomy, Cerebrovascular Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A.
| | | | | | | | | | | |
Collapse
|
470
|
Stier CT, Chander PN, Rosenfeld L, Powers CA. Estrogen promotes microvascular pathology in female stroke-prone spontaneously hypertensive rats. Am J Physiol Endocrinol Metab 2003; 285:E232-9. [PMID: 12670833 DOI: 10.1152/ajpendo.00029.2003] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Estrogen produces both beneficial and adverse effects on cardiovascular health via mechanisms that remain unclear. Stroke-prone spontaneously hypertensive rats (SHRSP) maintained on Stroke-Prone Rodent Diet and 1% NaCl drinking water (starting at 8 wk of age) rapidly develop stroke and malignant nephrosclerosis that can be prevented, despite continued hypertension, by drugs targeting angiotensin II and aldosterone actions. This study evaluated estrogen's effects in the SHRSP model. Female SHRSP that were sham operated (SHAM), ovariectomized (OVX) at 4 wk of age, or OVX and treated with estradiol benzoate (E2,30 microg x kg-1 x wk-1) were studied. In a survival protocol, OVX rats lived significantly longer (15.1 +/- 0.3 wk) compared with SHAM (13.6 +/- 0.2 wk) or OVX+E2 rats (12.4 +/- 0.2 wk). In a protocol in which animals were matched for age, at 11.5 wk, terminal systolic blood pressure and urine protein excretion were elevated in SHAM and OVX+E2 rats compared with OVX rats; blood urea nitrogen, renal microvascular and glomerular lesions, and plasma renin concentration were elevated in OVX+E2 relative to SHAM or OVX rats. In a survival protocol using intact female SHRSP, treatment with an antiestrogen (tamoxifen, 7 mg.kg-1.wk-1) prolonged survival by >2 wk compared with controls (P < 0.01). The data indicate that estrogen promotes microangiopathy in the kidney and stroke in saline-drinking SHRSP.
Collapse
Affiliation(s)
- Charles T Stier
- Department of Pharmacology, New York Medical College, Valhalla 10595, USA.
| | | | | | | |
Collapse
|
471
|
Murray HE, Pillai AV, McArthur SR, Razvi N, Datla KP, Dexter DT, Gillies GE. Dose- and sex-dependent effects of the neurotoxin 6-hydroxydopamine on the nigrostriatal dopaminergic pathway of adult rats: differential actions of estrogen in males and females. Neuroscience 2003; 116:213-22. [PMID: 12535954 DOI: 10.1016/s0306-4522(02)00578-x] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Epidemiological and clinical studies provide growing evidence for marked sex differences in the incidence of certain neurological disorders that are largely attributed to the neuroprotective effects of estrogen. Thus there is a keen interest in the clinical potential of estrogen-related compounds to act as novel therapeutic agents in conditions of neuronal injury and neurodegeneration such as Parkinson's disease. Studies employing animal models of neurodegeneration in ovariectomised female rats treated with estrogen support this hypothesis, yet experimental evidence for sex differences in the CNS response to direct neurotoxic insult is limited and, as yet, few studies have addressed the role played by endogenously produced hormones in neuroprotection. Therefore, in this study we aimed to determine (1) whether the prevailing levels of sex steroid hormones in the intact rat provide a degree of protection against neuronal assault in females compared with males and (2) whether sex differences depend solely on male/female differences in circulating estrogen levels or whether androgens could also play a role. Using the selective, centrally administered neurotoxin 6-hydroxydopamine, which induces a lesion in the nigrostriatal dopaminergic pathway similar to that seen in Parkinson's disease, we have demonstrated a sexually dimorphic (male-dominant), dose-dependent susceptibility in rats. Furthermore, following gonadectomy, dopamine depletion resulting from a submaximal dose of 6-hydroxydopamine (1 microg) was reduced in male rats, whereas in females, ovariectomy enhanced dopamine depletion. Administration of the nonaromatizable androgen dihydrotestosterone to gonadectomized animals had no significant effect on 6-hydroxydopamine toxicity in either males or females, whereas treatment of gonadectomized males and females with physiological levels of estrogen restored the extent of striatal dopamine loss to that seen in intact rats, viz, estrogen therapy reduced lesion size in females but increased it in males. Taken together, our findings strongly suggest that there are sex differences in the mechanisms whereby nigrostriatal dopaminergic neurones respond to injury. They also reveal that the reported clinically beneficial effects of estrogen in females may not be universally adopted for males. While the reasons for this gender-determined difference in response to the activational action of estrogen are unknown, we hypothesize that they may well be related to the early organizational events mediated by sex steroid hormones, which ultimately result in the sexual differentiation of the brain.
Collapse
Affiliation(s)
- H E Murray
- Department of Neuroendocrinology, Division of Neuroscience and Psychological Medicine, Faculty of Medicine, Imperial College, Hammersmith Hospital Campus, London W12 0NN, UK.
| | | | | | | | | | | | | |
Collapse
|
472
|
Murphy S, McCullough L, Littleton-Kearney M, Hurn P. Estrogen and selective estrogen receptor modulators: neuroprotection in the Women's Health Initiative era. Endocrine 2003; 21:17-26. [PMID: 12777699 DOI: 10.1385/endo:21:1:17] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2003] [Revised: 02/04/2003] [Accepted: 02/11/2003] [Indexed: 11/11/2022]
Abstract
Estrogen has been comprehensively studied as a neuroprotective agent in women, animals, and a variety of in vitro models of neural injury and degeneration. Most data suggest that estrogen can benefit the ischemic brain and reduce cell death. However, recent data from the Women's Health Initiative have raised concerns about the utility and safety of chronic estrogen use in women. While estrogen is a potent and reproducible neuroprotectant in animals and in vitro, its current administration in women has had unanticipated and paradoxical effects. Nonetheless, estrogen's diverse actions make it an ideal prototype for developing new neuroprotectants such as selective estrogen receptor modulators (SERMs). SERMs represent a class of drugs with mixed estrogen agonistic and antagonistic activity. Experimental and clinical data suggest a neuroprotective role for SERMs in normal and injured brain. The discrepancy among observational studies, preclinical data, and clinical trials emphasizes the need for further study of the mechanisms leading to the increased incidence of stroke observed in postmenopausal women. Research is still needed to optimize combined or estrogen alone hormone replacement therapy options as well as the prevention/management of cerebrovascular/ central nervous system disorders. This review critiques estrogen and SERMs' neuroprotective potential in experimental and clinical studies of stroke and cerebrovascular disease.
Collapse
Affiliation(s)
- Stephanie Murphy
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
473
|
Shughrue PJ, Merchenthaler I. Estrogen prevents the loss of CA1 hippocampal neurons in gerbils after ischemic injury. Neuroscience 2003; 116:851-61. [PMID: 12573724 DOI: 10.1016/s0306-4522(02)00790-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Estrogen replacement therapy is thought to attenuate the incidence of Alzheimer's disease in women and enhance cognitive functions. In rodents, estrogen protects cerebral cortical neurons from ischemic injury and cultured neurons from a variety of perturbations. Because few nuclear estrogen receptors have been detected in the dorsal hippocampus, the present studies used a global ischemia model to evaluate the neuroprotective actions of estrogen in this region. Ovariectomized gerbils were treated with placebo, 0.5 mg or 1 mg pellets of estradiol for 13 days. On day 7, the common carotid arteries were occluded for 5 min and on day 13 the animals were killed. Analysis of neurogranin mRNA, a marker of hippocampal neurons, with in situ hybridization revealed a dramatic and selective loss of CA1 neurons in the placebo-treated ovariectomized gerbils, whereas both 0.5 mg and 1 mg pellets of 17beta-estradiol prevented cell loss. Subsequent studies showed that a variety of estrogens, including diethylstilbestrol, estrone and 17alpha-estradiol as well as vitamin E, also protected CA1 neurons from ischemic injury in ovariectomized gerbils, whereas treatment with the estrogen antagonist tamoxifen was ineffective. The results of in vivo binding studies with 17alpha-iodovinyl-11beta-methoxyestradiol revealed a concentration of nuclear estrogen binding sites in the CA1 region of the ovariectomized gerbil brain, whereas binding in other hippocampal regions was limited. Moreover, the binding studies revealed that the regional pattern of binding was not altered after ischemic injury, with the exception of the hippocampus, where the binding sites were attenuated in ovariectomized animals with time after ischemic injury. Together, these data demonstrate that a variety of steroidal and non-steroidal estrogens are potent neuroprotective agents in an animal model of global ischemia, agents that protect neurons critical for learning and memory and susceptible to neurodegenerative diseases.
Collapse
Affiliation(s)
- P J Shughrue
- The Women's Health Research Institute, Wyeth Research, Collegeville, PA 19426, USA.
| | | |
Collapse
|
474
|
Hilton GD, Nuñez JL, McCarthy MM. Sex differences in response to kainic acid and estradiol in the hippocampus of newborn rats. Neuroscience 2003; 116:383-91. [PMID: 12559094 DOI: 10.1016/s0306-4522(02)00716-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Premature and full-term human infants are at considerable risk of excitotoxic-mediated brain damage due to hypoxia-ischemia, infection or other trauma. Glutamate receptor activation is a major source of excitoxicity in the adult and developing brain, and the hippocampus is particularly vulnerable to damage. The seven-day-old rat is a widely used model of pediatric brain damage, in large part due to the relative insensitivity of the brain to exogenous glutamate treatment prior to this age. We have reexamined the possible role of glutamate in pediatric brain damage in the newborn rat using kainic acid treatment and attending to the sex of the animal as well as the effects of pretreatment with the gonadal steroid estradiol. Consistent with previous studies, we found no evidence of damage 7 days posttreatment in the CA1 region of the hippocampus in males or females. There was also little to no damage in the CA2/3 or dentate gyrus of males. In females, however, kainic-acid treatment induced substantial damage in the dentate gyrus and moderate damage in CA2/3, as assessed by neuron number and regional volume. Pretreatment with estradiol was protective against kainic acid-induced damage in females but was permissive for damage in the dentate gyrus of males. Estradiol treatment in the absence of kainic acid treatment was also neuroprotective in females in that it increased neuron number and volume throughout the hippocampal formation, suggesting that the basis of the sex difference observed in hippocampal volume was hormonally mediated. There was no effect of exogenous estradiol given to males in the absence of kainic acid. We conclude that the newborn female rat brain, but not the male, is sensitive to glutamate-mediated toxicity and that gonadal steroids play a complex role in both naturally occurring sex differences in hippocampal volume and response to injury.
Collapse
Affiliation(s)
- G D Hilton
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
475
|
Antezana DF, Clatterbuck RE, Alkayed NJ, Murphy SJ, Anderson LG, Frazier J, Hurn PD, Traystman RJ, Tamargo RJ. High-dose ibuprofen for reduction of striatal infarcts during middle cerebral artery occlusion in rats. J Neurosurg 2003; 98:860-6. [PMID: 12691413 DOI: 10.3171/jns.2003.98.4.0860] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Ibuprofen is an antiinflammatory drug that disrupts leukocyte-endothelial cell interactions by limiting expression of endothelial adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1), also known as CD54. The authors hypothesized that ibuprofen could reduce the size of the infarct associated with transient focal ischemia by inhibition of ICAM-1 expression, and they evaluated its effects in rats treated with middle cerebral artery (MCA) occlusion. Ibuprofen treatment was compared with mild systemic hypothermia, which is known to be neuroprotective and is commonly used during neurosurgical procedures. METHODS The maximum ibuprofen dose (240 mg/kg/day) that could be tolerated with no systemic toxicity was established in the initial experiments. In the efficacy experiment, rats were pretreated with vehicle, ibuprofen, or hypothermia (33 degrees C) prior to 2 hours of MCA occlusion; then their brains were harvested at 24 hours of reperfusion for histological studies. End-ischemic cerebral blood flow (CBF) was evaluated using [14C]iodoantipyrine autoradiography in additional cohorts. Expression of ICAM-1 within ischemic compared with nonischemic caudate nucleus and putamen (striatum) or cortex was evaluated using immunohistochemical studies. Compared with vehicle treatment, ibuprofen produced a 46.2% reduction (p = 0.01) in striatal infarcts, which was comparable to hypothermia (48.7% reduction, p = 0.02). Ibuprofen did not alter end-ischemic CBF in any region studied, and the ibuprofen treatment group had the lowest proportion of animals with marked ICAM-1 staining. CONCLUSIONS Ibuprofen given in maximum tolerated doses reduces the striatal infarct size after focal cerebral ischemia. The neuroprotective mechanism does not work through preservation of intraischemic CBF and is consistent with inhibition of ICAM-1 expression; however, at the doses used in this study, other effects of ibuprofen on platelet and endothelial function are possible.
Collapse
Affiliation(s)
- David F Antezana
- Department of Neurosurgery, The Johns Hopkins Hospital, Baltimore, Maryland 21287, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
476
|
Krupinski J, Slevin M, Marti E, Catena E, Rubio F, Gaffney J. Time-course phosphorylation of the mitogen activated protein (MAP) kinase group of signalling proteins and related molecules following middle cerebral artery occlusion (MCAO) in rats. Neuropathol Appl Neurobiol 2003; 29:144-58. [PMID: 12662322 DOI: 10.1046/j.1365-2990.2003.00454.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recovery from the debilitating effects of ischaemic stroke is variable and unpredictable. To maximize patient recovery, a greater understanding of the molecular mechanisms involved in regulating both apoptosis and the repair processes affecting neuronal protection, particularly in the penumbra region, is desirable. We have previously shown, in human subjects, the increased expression of several growth factors soon after stroke, together with appearance of tyrosine phosphorylated proteins, in particular mitogen activated protein (MAP) kinase (ERK1/2). In this paper, we demonstrate a relatively short-lasting (< 12 h), but substantial increase in expression of phosphorylated proteins, in particular, p-JNK (phosphorylated c-Jun N-terminal kinase) and p-ERK1/2 in both the grey matter penumbra and infarcted tissue of rats, following permanent middle cerebral artery occlusion. p-ERK1/2 was associated with neurones and endothelial cells in the vicinity of the infarct while p-JNK was mainly expressed in neurones. Expression of both p-MEK3/6 and p-p38 MAP kinase was also increased in neurones and astroglia, within 1 h of infarction, p-p38 remaining elevated and associated with neurones and in particular with astroglia in the penumbra region for > 4 days. Evidence suggests that short-term activation of these proteins may be detrimental to neuronal survival, while their transient nature makes them unlikely to support angiogenesis, revascularization and reperfusion over a period of days and weeks. On the other hand, short-medium-term up-regulation of neuronal p-JNK, p-c-Jun, p-Stat-1 and p-p38 might be a factor in the regulation of apoptosis. Therapeutic manipulation of phosphorylation/activation of these and other important signalling intermediates might form the basis of an appropriate treatment to maximize revascularization and neuronal protection after ischaemic stroke.
Collapse
Affiliation(s)
- J Krupinski
- Department of Neurology, Hospital Princeps d'Espanya, Ciutat Sanitaria I Universitaria de Belvitge, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
477
|
Kupina NC, Detloff MR, Bobrowski WF, Snyder BJ, Hall ED. Cytoskeletal protein degradation and neurodegeneration evolves differently in males and females following experimental head injury. Exp Neurol 2003; 180:55-73. [PMID: 12668149 DOI: 10.1016/s0014-4886(02)00048-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The resulting neuropathological degeneration that occurs following a traumatic brain injury (TBI) is a consequence of both immediate and secondary neurochemical sequelae. Proteolysis of cytoskeletal proteins, triggered by calcium-mediated events, is believed to be a particularly significant contributor to TBI-induced neuronal death. To date, efforts to associate cytoskeletal degradation and neurodegeneration in TBI have been primarily qualitative or semiquantitative. The objectives of this study were (1). to quantitatively describe, over a posttraumatic time course, the relationship and mechanisms of cytoskeletal degradation (Western blot) and neurodegeneration (silver staining) in male and female mice following a moderately severe weight-drop impact-acceleration head injury; (2). to evaluate gender differences in the response to TBI; and (3). to examine the potential therapeutic window for future pharmacological treatment strategies. In male and female mice, we report a close correlation in the time courses of neurofilament M protein degradation and alpha-spectrin breakdown products (SBDP 150 and 145) with the peak magnitude of neurodegeneration, as quantified by silver staining. Evidence from the increased patterns of SBDPs suggests that both calpain and caspase-3 are involved. In general, males incurred peak protein degradation and neurodegeneration within 3 days after injury, while in females this did not occur until 14 days. The neuroprotective effects of estrogen are believed to be key factors in the superior outcome of female vs male mice following TBI. In mice, the therapeutic window of opportunity for pharmacological intervention aimed at limiting cytoskeletal degradation might be as much as 24 h following injury. Evidence of a protracted time course of cytoskeletal degradation, especially in females, suggests a potential for an extended treatment-duration following TBI.
Collapse
Affiliation(s)
- Nancy C Kupina
- CNS Pharmacology, Pfizer Global Research and Development-Ann Arbor Laboratories, 2800 Plymouth Road, Ann Arbor, MI 48105, USA
| | | | | | | | | |
Collapse
|
478
|
Abstract
Over the past century, the average lifespan of women has increased from 50 to over 80 years, but the age of the menopause has remained fixed at 51 years. This "change of life" is marked by a dramatic and permanent decrease in circulating levels of ovarian estrogens. Therefore, more women will live a greater proportion of their lives in a chronic hypoestrogenic state. Ovarian steroid hormones are pleiotropic and have multiple, diverse, and possibly opposing actions in different contexts. In light of recent reports of the possible health risks of hormone replacement therapy (HRT) on several different physiological systems, the question of whether estrogens are protective or risk factors must be carefully re-evaluated.
Collapse
Affiliation(s)
- Phyllis M Wise
- Department of Neurobiology, Physiology, and Behavior, Division of Biological Sciences, University of California Davis, One Shields Avenue, Davis, CA 95616-8536, USA.
| |
Collapse
|
479
|
Jesmin S, Hattori Y, Sakuma I, Liu MY, Mowa CN, Kitabatake A. Estrogen deprivation and replacement modulate cerebral capillary density with vascular expression of angiogenic molecules in middle-aged female rats. J Cereb Blood Flow Metab 2003; 23:181-9. [PMID: 12571449 DOI: 10.1097/01.wcb.0000043341.09081.37] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The effect of postmenopausal estrogen replacement therapy (ERT) on the risk or severity of cerebrovascular disorders is as yet unclear, and the evidence for flow preservation being a mechanism of estrogen neuroprotection remains elusive. The authors examined whether estrogen-mediated flow-preserving neuroprotective mechanisms, if any, may involve its angiogenic action. This study was conducted using middle-aged (44 weeks) female rats because of the importance of aging in cerebrovascular disease in women. Middle-aged female rats were subjected to sham operation, ovariectomy, or ovariectomy with ERT. The anatomic cerebral capillary morphology showed a significant reduction in the total capillary density in the frontal cortex after ovariectomy. This was associated with marked decreases in protein and gene expression of vascular endothelial growth factor and its angiogenic receptors in cerebral vessels, as demonstrated by immunohistochemistry and hybridization. The expression levels of both estrogen receptor (ER) subtypes, ERalpha and ERbeta, in cerebral vessels were significantly reduced after ovariectomy, but ERbeta was more dramatically downregulated as assessed by the ERbeta/ERalpha ratio. These ovariectomy-induced changes were completely prevented by ERT. Vascular endothelial growth factor appears to be a critical regulatory molecule for physiologic cerebral angiogenesis in middle-aged female rats and may play an important role in the flow-preserving neuroprotective action of estrogen through its angiogenic and antiapoptotic properties.
Collapse
Affiliation(s)
- Subrina Jesmin
- Departments of Cardiobascular Medicine, Hakkaido University School of Medicine, Sapporo, Japan
| | | | | | | | | | | |
Collapse
|
480
|
Dhandapani KM, Brann DW. Protective effects of estrogen and selective estrogen receptor modulators in the brain. Biol Reprod 2002; 67:1379-85. [PMID: 12390866 DOI: 10.1095/biolreprod.102.003848] [Citation(s) in RCA: 173] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Within the last few years, there has been a growing interest in the neuroprotective effects of estrogen and the possible beneficial effects of estrogen in neurodegenerative diseases such as stroke, Alzheimer disease, and Parkinson disease. Here, we review the progress in this field, with a particular focus upon estrogen-induced protection from stroke-induced ischemic damage. The important issue of whether clinically relevant selective estrogen receptor modulators (SERMs) such as tamoxifen and raloxifene and estrogen replacement therapy can exert neuroprotection is also addressed. Although the mechanism of estrogen and SERM neuroprotection is not clearly resolved, we summarize the leading possibilities, including 1) a genomic estrogen receptor-mediated pathway that involves gene transcription, 2) a nongenomic signaling pathway involving activation of cell signalers such as mitogen-activated protein kinases and/or phosphatidylinositol-3-kinase /protein kinase B, and 3) a nonreceptor antioxidant free-radical scavenging pathway that is primarily observed with pharmacological doses of estrogen. The role of other potential mediatory factors such as growth factors and the possibility of an astrocyte role in neuroprotection is also discussed.
Collapse
Affiliation(s)
- Krishnan M Dhandapani
- Institute of Molecular Medicine and Genetics, Program in Neurobiology, and Department of Neurology, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | |
Collapse
|
481
|
Horsburgh K, Macrae IM, Carswell H. Estrogen is neuroprotective via an apolipoprotein E-dependent mechanism in a mouse model of global ischemia. J Cereb Blood Flow Metab 2002; 22:1189-95. [PMID: 12368657 DOI: 10.1097/01.wcb.0000037991.07114.4e] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Estrogen can ameliorate brain damage in experimental models of focal cerebral ischemia., estrogen increases levels of apolipoprotein E (apoE), which also has neuroprotective effects in brain injury. The authors tested the hypotheses that physiologically relevant levels of 17beta-estradiol are neuroprotective in global cerebral ischemia and that neuroprotection is mediated via apoE. In the first study, subcutaneous implants of 17beta-estradiol were tested in female C57Bl/6J mice (ovariectomized and nonovariectomized) and plasma levels measured by radioimmunoassay to validate that physiologically relevant levels could be achieved. In the second study, female C57Bl/6J and apoE-deficient mice were ovariectomized and implanted with 17beta-estradiol or placebo pellet. Two weeks later, transient global ischemia was induced by bilateral carotid artery occlusion and the mice killed after 72 hours. Ischemic and normal neurons were counted in the caudate nucleus and CA1 pyramidal cell layer and the percentage of neuronal damage was compared between the treated groups. In C57Bl/6J mice, there was less neuronal damage in the 17beta-estradiol-treated group compared with placebo group in the caudate nucleus (15 +/- 20% versus 39 +/- 27%, = 0.02) and in the CA1 pyramidal cell layer (1.8 +/- 2% versus 10 +/- 14%, = 0.08). In contrast, neuronal damage was not significantly different between the 17beta-estradiol and placebo groups in apoE-deficient mice in the caudate nucleus (47 +/- 35% versus 53 +/- 29%, = 0.7) or in the CA1 pyramidal cell layer (24 +/- 19% versus 24 +/- 19%, = 1.0). The data indicate a neuroprotective role for estrogen in global ischemia, the mechanism of which is apoE-dependent.
Collapse
Affiliation(s)
- Karen Horsburgh
- Wellcome Surgical Institute and Hugh Fraser Neuroscience Laboratories, Glasgow, Scotland, UK.
| | | | | |
Collapse
|
482
|
Estrogen Is Neuroprotective via an Apolipoprotein E???Dependent Mechanism in a Mouse Model of Global Ischemia. J Cereb Blood Flow Metab 2002. [DOI: 10.1097/00004647-200210000-00006] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
483
|
Murphy SJ, Littleton-Kearney MT, Hurn PD. Progesterone administration during reperfusion, but not preischemia alone, reduces injury in ovariectomized rats. J Cereb Blood Flow Metab 2002; 22:1181-8. [PMID: 12368656 DOI: 10.1097/01.wcb.0000037990.07114.07] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although progesterone is neuroprotective in traumatic brain injury, its efficacy in stroke is unclear. The authors determined whether there are infarction differences after middle cerebral artery occlusion (MCAO) in ovariectomized rats treated acutely with progesterone before MCAO or both pre- and postischemia. Rats received vehicle, 5 (P5), 10 (P10), or 20 (P20) mg/kg progesterone intraperitoneally 30 minutes before MCAO. In another cohort, animals received vehicle or 5 (P5R) mg/kg progesterone intraperitoneally 30 minutes before MCAO, at reperfusion initiation, and at 6-hour reperfusion. Animals underwent 2-hour MCAO by the intraluminal filament technique, followed by 22-hour reperfusion. Cortical (CTX) and caudate-putamen (CP) infarctions were determined by 2,3,5-triphenyltetrazolium chloride staining and digital image analysis. End-ischemic and early reperfusion regional cerebral blood flow (CBF) was measured by [ C]-iodoantipyrine quantitative autoradiography in vehicle- or progesterone (5 mg/kg)-treated rats. Cortical infarction (% contralateral CTX) was 31 +/- 30% (vehicle), 39 +/- 23% (P5), 41 +/- 14% (P10), and 28 +/- 20% (P20). Caudate-putamen infarction (% contralateral CP) was 45 +/- 37% (vehicle), 62 +/- 34% (P5), 75 +/- 17% (P10), and 52 +/- 30% (P20). In vehicle and P5R groups, CTX infarction was 37 +/- 20% and *20 +/- 17%, respectively (* < 0.05 from vehicle). In vehicle and P5R groups, CP infarction was 63 +/- 26% and 43 +/- 29%, respectively. End-ischemic regional CBF and CBF recovery during initial reperfusion was unaffected by progesterone treatment. These data suggest that progesterone administration both before MCAO and during reperfusion decreases ischemic brain injury.
Collapse
Affiliation(s)
- Stephanie J Murphy
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, USA.
| | | | | |
Collapse
|
484
|
Okamoto K, Takai S, Sasaki S, Miyazaki M. Trandolapril reduces infarction area after middle cerebral artery occlusion in rats. Hypertens Res 2002; 25:583-8. [PMID: 12358145 DOI: 10.1291/hypres.25.583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In this study, we investigated whether angiotensin-converting enzyme (ACE) is involved in the progression of cerebral infarct lesions after middle cerebral artery (MCA) occlusion in rats. After placebo or trandolapril was administered orally for 7 days, we infarcted in the territory of the right MCA by extracranial vascular occlusion and studied the effect of trandolapril on brain ACE activity and infarct size 7 days after MCA occlusion. In placebo-treated rats, brain ACE activity in the infarct side was increased by a significant 1.34-fold compared with that in the non-infarct side 7 days after MCA occlusion. Brain ACE activities in the infarct sides were suppressed to 39.8% by trandolapril treatment. The ratios of unilateral infarcts to the total coronal sectional areas in placebo- and trandolapril-treated rats were 48.1 +/- 3.3% and 37.4 +/- 2.3%, respectively, and the difference between these values was significant. These results demonstrate that inhibition of the increased brain ACE activity in infarct lesions can reduce the infarction area after MCA occlusion.
Collapse
Affiliation(s)
- Ken Okamoto
- Department of Pharmacology, Osaka Medical College, Takatsuki, Japan
| | | | | | | |
Collapse
|
485
|
Wilson ME, Liu Y, Wise PM. Estradiol enhances Akt activation in cortical explant cultures following neuronal injury. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 102:48-54. [PMID: 12191493 DOI: 10.1016/s0169-328x(02)00181-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We have previously demonstrated that estradiol reduces cell death in cortical explant cultures following injury induced by metabolic inhibition in a receptor-dependent fashion. In this study, we examined whether cell death involves apoptosis and assessed the potential mediators of estradiol's actions. Cortical explant cultures were generated from postnatal day 3 rat pups. On day 7 in vitro, explants were injured by exposure to 1 mM 2-DG/2 mM KCN for 2 h to model the metabolic inhibition observed during ischemia. Explants were fixed in 4% paraformaldehyde at 2, 6, 10 and 24 h following the injury period and 18-microm thick sections were cut on a cryostat and stained with cresyl violet to assess cell death. The same sections were also labeled by TUNEL to determine whether cell death occurred by apoptosis. Other sections were used for immunohistochemistry to determine whether cells that stained positive for activated caspase 3 were also immunopositive for NeuN, a neuronal marker, or GFAP, an astrocyte marker. Protein was extracted for Western blot analysis from a separate set of explants collected at 0, 0.5, 1, 2 and 4 h following the conclusion of the injury. Estradiol treatment significantly reduced the number of cells undergoing apoptotic cell death as indicated by nuclear condensation visualized by cresyl violet staining (P<0.05). TUNEL staining revealed that the majority of pyknotic and fragmented nuclei were also TUNEL positive. Furthermore, caspase 3 activation appeared to be restricted to neurons. To examine a possible mechanism by which estradiol prevents apoptosis, we examined the level of activation of Akt kinase, which mediates antiapoptotic signals. Potential activation was measured by phosphorylation of Akt at Ser473 by Western blot analysis. In the absence of estradiol, pAkt levels were significantly increased at 2 h following the termination of injury. Explants that were pretreated with estradiol exhibited elevated levels of pAkt at 1 h following injury. Treatment with ICI 182,780 prevented the effect of estradiol. These studies suggest that estradiol prevents injury-induced apoptosis and that Akt activation may mediate these protective effects.
Collapse
Affiliation(s)
- Melinda E Wilson
- Department of Physiology, College of Medicine, University of Kentucky, MS 508, 800 Rose Street, Lexington, KY 40536, USA.
| | | | | |
Collapse
|
486
|
Dhandapani KM, Brann DW. Estrogen-astrocyte interactions: implications for neuroprotection. BMC Neurosci 2002; 3:6. [PMID: 12067420 PMCID: PMC116596 DOI: 10.1186/1471-2202-3-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2002] [Accepted: 06/07/2002] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Recent work has suggested that the ovarian steroid 17beta-estradiol, at physiological concentrations, may exert protective effects in neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and acute ischemic stroke. While physiological concentrations of estrogen have consistently been shown to be protective in vivo, direct protection upon purified neurons is controversial, with many investigators unable to show a direct protection in highly purified primary neuronal cultures. These findings suggest that while direct protection may occur in some instances, an alternative or parallel pathway for protection may exist which could involve another cell type in the brain. PRESENTATION OF THE HYPOTHESIS A hypothetical indirect protective mechanism is proposed whereby physiological levels of estrogen stimulate the release of astrocyte-derived neuroprotective factors, which aid in the protection of neurons from cell death. This hypothesis is attractive as it provides a potential mechanism for protection of estrogen receptor (ER)-negative neurons through an astrocyte intermediate. It is envisioned that the indirect pathway could act in concert with the direct pathway to achieve a more widespread global protection of both ER+ and ER- neurons. TESTING THE HYPOTHESIS We hypothesize that targeted deletion of estrogen receptors in astrocytes will significantly attenuate the neuroprotective effects of estrogen. IMPLICATIONS OF THE HYPOTHESIS If true, the hypothesis would significantly advance our understanding of endocrine-glia-neuron interactions. It may also help explain, at least in part, the reported beneficial effects of estrogen in neurodegenerative disorders. Finally, it also sets the stage for potential extension of the hypothetical mechanism to other important estrogen actions in the brain such as neurotropism, neurosecretion, and synaptic plasticity.
Collapse
Affiliation(s)
- Krishnan M Dhandapani
- Institute of Molecular Medicine and Genetics, Neurobiology Program and Department of Neurology, 1120 15 Street, Medical College of Georgia, Augusta, GA 30912
| | - Darrell W Brann
- Institute of Molecular Medicine and Genetics, Neurobiology Program and Department of Neurology, 1120 15 Street, Medical College of Georgia, Augusta, GA 30912
| |
Collapse
|
487
|
Abstract
In addition to its role as a sex hormone, oestrogen affects the structure and function of the nervous system. Oestrogen receptors are expressed in brain regions that are involved in sex differentiation and maturation. But in addition to its well-known effects, oestrogen also has important neuroprotective actions that are both dependent and independent of a nuclear oestrogen-receptor activity. Furthermore, oestrogen can interact with neuroprotective intracellular signalling pathways and is itself a neuroprotective antioxidant. Understanding the mechanisms of oestrogen action will be crucial to determine its potential as a therapeutic agent, particularly in the elderly.
Collapse
Affiliation(s)
- Christian Behl
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| |
Collapse
|
488
|
DeGiorgio LA, Attardi B, Shimizu Y, Ogata M, Volpe BT. 17 beta-estradiol treatment retards excitotoxic delayed degeneration in substantia nigra reticulata neurons. Brain Res 2002; 936:15-20. [PMID: 11988225 DOI: 10.1016/s0006-8993(02)02482-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Estrogen treatment offers neuro-protection in animal experiments in which excitotoxic mechanisms destroy neurons. In a model of delayed neuronal degeneration that depends on excitotoxicity, we tested whether females had an altered susceptibility, and whether physiologic doses of estrogen administered after the brain insult would protect susceptible neurons. Females were ovariectomized, exposed to striatal-pallidal ibotenic acid injury that caused delayed degeneration of substantia nigra neurons, and treated with 17 beta -estradiol (30 microg, subcutaneously every other day, beginning 2 days after the striatal injury) or vehicle. At 6 and 8 days post lesion, the 17beta-estradiol treatment group maintained over 87 and 70% of control nigral neuron number, respectively. Physiologic levels of estrogen delivered days after the excitotoxic stress completely protected neurons in the substantia nigra reticulata 6 days post lesion and slowed degeneration 8 days post lesion.
Collapse
Affiliation(s)
- Lorraine A DeGiorgio
- Burke Medical Research Institute, Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 785 Mamaroneck Avenue, White Plains, NY 10605, USA.
| | | | | | | | | |
Collapse
|
489
|
Derdeyn CP, Cross DT, Moran CJ, Brown GW, Pilgram TK, Diringer MN, Grubb RL, Rich KM, Chicoine MR, Dacey RG. Postprocedure ischemic events after treatment of intracranial aneurysms with Guglielmi detachable coils. J Neurosurg 2002; 96:837-43. [PMID: 12008697 DOI: 10.3171/jns.2002.96.5.0837] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Ischemic stroke or transient ischemic attack (TIA) may occur after the treatment of intracranial aneurysms with Guglielmi detachable coils (GDCs). The purpose of the present study is to investigate possible risk factors for thromboembolic events and to determine their frequency and time course. METHODS The records of 178 consecutive patients with 193 treated intracranial saccular aneurysms were reviewed. A total of 159 GDC procedures were performed to treat 143 aneurysms in 133 of those patients who were in good neurological condition, allowing clinical detection of postprocedure ischemic events (TIA or stroke). The association of clinical, anatomical, and pharmacological factors with intraprocedure intraarterial thrombus and with postprocedure ischemic events was investigated by using uni- and multivariate analyses. Thrombus protruding into the parent artery was noted during six of 159 GDC procedures, resulting in a clinical deficit in one patient. No factor was associated with intraprocedure intraarterial thrombus. Ten postprocedure ischemic events occurred in nine patients. Seven events occurred within 24 hours, and three events occurred between 24 hours and 58 days. Aneurysm diameter and protruding coils were significant independent predictors of postprocedure ischemic events in multivariate analysis (both p = 0.02). The actuarial risk of stroke was 3.8%. CONCLUSIONS Larger aneurysm diameter and protruding loops of coils are associated with postprocedure ischemic events after GDC placement. It is unlikely that GDC-treated aneurysms retain thromboembolic potential beyond 2 months.
Collapse
Affiliation(s)
- Colin P Derdeyn
- Interventional Neuroradiology Service, Washington University School of Medicine, St Louis, MO, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
490
|
von Arnim CAF, Etrich SM, Timmler M, Riepe MW. Gender-dependent hypoxic tolerance mediated via gender-specific mechanisms. J Neurosci Res 2002; 68:84-8. [PMID: 11933052 DOI: 10.1002/jnr.10195] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Primary hypoxic tolerance and preconditioning are gender dependent and modulated in females during the estrus cycle. The underlying mechanisms, however, remain to be determined. mRNA of estrogen receptor-alpha (EAR), progesterone receptor (PR), and adenosine receptor subtypes A1 and A3 (A1R and A3R) were investigated with reverse transcriptase-PCR in hippocampi from control male and female mice and animals treated in vivo with a single i.p. injection of 20 mg/kg body weight 3-nitropropionate (3NP) 1 or 24 hr prior to preparation. Results were analyzed relative to expression in hippocampi from untreated males. mRNA levels of EAR and A1R were alike in males and females and unaltered by preconditioning with 3NP. In contrast, PR mRNA levels were alike in males and females during proestrus but lower during estrus and diestrus (85% +/- 15%, P < 0.05; and 80% +/- 10%, P < 0.05, respectively). Upon preconditioning, PR mRNA decreased to 67% +/- 19% (P < 0.05) and 56% +/- 13% (P < 0.05) during proestrus and diestrus, respectively, but was unaltered during estrus and in males. On preconditioning, A3R mRNA decreased from 115% +/- 16% to 86% +/- 29% (P < 0.05) during diestrus but remained at the control level during proestrus and estrus. With low-level expression of PRs, as achieved upon preconditioning, hypoxic tolerance is increased. Other than in males, adenosine A3 receptors are not up-regulated upon preconditioning in females. Thus, not only is net hypoxic tolerance gender dependent but mechanisms conferring hypoxic tolerance are gender specific.
Collapse
MESH Headings
- Adaptation, Physiological/drug effects
- Adaptation, Physiological/physiology
- Animals
- Convulsants/pharmacology
- Estrogen Receptor alpha
- Estrous Cycle/drug effects
- Estrous Cycle/metabolism
- Female
- Hippocampus/chemistry
- Hippocampus/drug effects
- Hippocampus/metabolism
- Hypoxia/physiopathology
- Injections, Intraperitoneal
- Male
- Mice
- Nitro Compounds
- Propionates/pharmacology
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/analysis
- Receptors, Progesterone/drug effects
- Receptors, Progesterone/metabolism
- Receptors, Purinergic P1/analysis
- Receptors, Purinergic P1/drug effects
- Receptors, Purinergic P1/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sex Characteristics
- Time Factors
Collapse
|
491
|
Estrogen protects against global ischemia-induced neuronal death and prevents activation of apoptotic signaling cascades in the hippocampal CA1. J Neurosci 2002. [PMID: 11896151 DOI: 10.1523/jneurosci.22-06-02115.2002] [Citation(s) in RCA: 204] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The importance of postmenopausal estrogen replacement therapy in affording protection against the selective and delayed neuronal death associated with cardiac arrest or cardiac surgery in women remains controversial. Here we report that exogenous estrogen at levels that are physiological for hormone replacement in postmenopausal women affords protection against global ischemia-induced neuronal death and prevents activation of apoptotic signaling cascades in the hippocampal CA1 of male gerbils. Global ischemia induced a marked increase in activated caspase-3 in CA1, evident at 6 hr after ischemia. Global ischemia induced a marked upregulation of the proapoptotic neurotrophin receptor p75(NTR) in CA1, evident at 48 hr. p75(NTR) expression was induced primarily in terminal deoxynucleotidyl transferase-mediated UTP nick-end labeling-positive cells, indicating expression in neurons undergoing apoptosis. Global ischemia also induced a marked downregulation of mRNA encoding the AMPA receptor GluR2 subunit in CA1. Caspase-3, p75(NTR), and GluR2 were not significantly changed in CA3 and dentate gyrus, indicating that the ischemia-induced changes in gene expression were cell specific. Exogenous estrogen attenuated the ischemia-induced increases in activated caspase-3 and blocked the increase in p75(NTR) in post-ischemic CA1 neurons but did not prevent ischemia-induced downregulation of GluR2. These findings demonstrate that long-term estrogen at physiological levels ameliorates ischemia-induced hippocampal injury and indicate that estrogen intervenes at the level of apoptotic signaling cascades to prevent onset of death in neurons otherwise "destined to die."
Collapse
|
492
|
Mayhan WG, Sun H, Irvine SD. Influence of gender on dilatation of the basilar artery during diabetes mellitus. Brain Res 2002; 930:182-90. [PMID: 11879808 DOI: 10.1016/s0006-8993(02)02245-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The first goal of the present study was to examine the influence of gender on reactivity of the basilar artery. The second goal of this study was to examine the effect of diabetes mellitus on reactivity of the basilar artery in male and female rats. We examined in vivo responses of the basilar artery in male and female nondiabetic and diabetic rats in response to a nitric oxide synthase (NOS)-dependent (acetylcholine) and -independent (nitroglycerin) agonist. In nondiabetic male and female rats, acetylcholine and nitroglycerin produced dose-related dilatation of the basilar artery. However, the magnitude of vasodilatation in response to acetylcholine and a high concentration of nitroglycerin was significantly greater in female than in male rats. Acetylcholine (1.0 microM) dilated the basilar artery by 11 +/- 2% in nondiabetic males versus 25 +/- 4% in nondiabetic females (P<0.05). Nitroglycerin (1.0 microM) dilated the basilar artery by 37 +/- 8% in nondiabetic males versus 62 +/- 5% in nondiabetic females (P<0.05). Thus, there is a significant effect of gender on reactivity of the basilar artery during physiologic conditions. Dilatation of the basilar artery in response to acetylcholine, but not nitroglycerin, was impaired in diabetic male and female rats compared to their nondiabetic counterparts. Acetylcholine (1.0 microM) dilated the basilar artery by only 5 +/- 1% in diabetic males and by only 4 +/- 1% in diabetic females. In summary, dilatation of the basilar artery in response to NOS-dependent agonist was significantly greater in nondiabetic female than in nondiabetic male rats. In addition, diabetes mellitus impaired NOS-dependent dilatation of the basilar artery not only in male rats, but also in female rats. We suggest that the results of these studies provide insight into the pathogenesis of cerebrovascular abnormalities observed in postmenopausal women.
Collapse
Affiliation(s)
- William G Mayhan
- Department of Physiology and Biophysics, University of Nebraska Medical Center, 984575 Nebraska Medical Center, Omaha, NE 68198-4575, USA.
| | | | | |
Collapse
|
493
|
Toung TJK, Hurn PD, Traystman RJ, Bhardwaj A. Global brain water increases after experimental focal cerebral ischemia: effect of hypertonic saline. Crit Care Med 2002; 30:644-9. [PMID: 11990928 DOI: 10.1097/00003246-200203000-00025] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Isolated experiments suggest that global cerebral edema is a sequela of large hemispheric ischemic lesions, presumably as an extension of the initial ischemic insult into areas of vital, noninjured tissue. Diuretics and osmotic agents are controversial and poorly defined therapeutic modalities after large infarction. By using a rat model of middle cerebral artery occlusion (MCAO), we tested the hypothesis that significant edema occurs in the contralateral uninjured hemisphere and that this postischemic complication can be manipulated by hypertonic saline therapy. DESIGN Prospective laboratory animal study. SETTING Research laboratory in a teaching hospital. SUBJECTS Halothane-anesthetized, male Wistar rats. INTERVENTIONS Under controlled conditions of normoxia, normocarbia, and normothermia, rats were subjected to 2 hrs of MCAO. MEASUREMENTS AND MAIN RESULTS Adequacy of MCAO and reperfusion was assessed by laser Doppler flowmetry. All animals except naive rats received continuous infusion of 0.9% saline at 0.5 mL/hr throughout the experiment. Brains were harvested, and tissue water content was estimated by comparing the wet-to-dry weight ratios of ipsilateral and contralateral cerebral hemispheres at 12 hrs, 24 hrs, or 2, 3, or 7 days postischemia. Naive and sham-operated rats served as control cohorts. In a second series of randomized experiments, wet-to-dry weight ratios were determined in rats treated with continuous intravenous infusion of 7.5% hypertonic saline (0.5 mL/hr; acetate/chloride, 50:50) and were compared with well-studied antiedema therapy: 20% mannitol (2.5 g/kg bolus every 6 hrs) or furosemide (2.5 mg/kg bolus every 6 hrs). Treatments were started at 24 hrs of reperfusion, and brain water was assessed at 2 days of reperfusion. In a third series of experiments, wet-to-dry ratios were determined in brains harvested at 2 days of reperfusion from rats that were subjected to 2 hrs of MCAO and did not receive any intravenous fluids. All values are mean +/- SEM. There were no differences between sham-operated and naive control cohorts. At 24 hrs of reperfusion, water content was higher in both ipsilateral ischemic (82.80 +/- 0.86%) and contralateral hemispheres (80.53 +/- 0.29%), compared with naive animals (ipsilateral, 79.62 +/- 0.12%; contralateral, 79.53 +/- 0.13%). Maximal cerebral edema was measured at 2 days in both hemispheres (ipsilateral, 83.94 +/- 0.47%; contralateral, 80.63 +/- 0.13%). Edema was present for up to 3 days in contralateral tissue (80.27 +/- 0.26%) and persisted to 7 days in the injured hemisphere (81.07 +/- 0.34%). Maximal edema (as assessed at 2 days postocclusion) was robustly attenuated with hypertonic saline therapy (ipsilateral, 81.59 +/- 0.52%; contralateral, 78.44 +/- 0.22%). The efficacy of hypertonic saline was equivalent to furosemide (ipsilateral, 82.09 +/- 0.50%; contralateral, 79.13 +/- 0.17%) but less robust than mannitol (ipsilateral, 79.89 +/- 0.36%; contralateral, 78.73 +/- 0.17%). CONCLUSIONS These data demonstrate that cerebral edema persists in both injured and contralateral hemispheres for days after MCAO. The global, maximal increase in brain water is responsive to continuous 7.5% hypertonic saline treatment begun at 24 hrs postischemia and to standard diuretic/osmotic agents. These results may have implications for diuretic and osmotic therapy in clinical ischemic stroke.
Collapse
Affiliation(s)
- Thomas J K Toung
- Department of Anesthesiology/Critcal Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
494
|
Wang MM, Klaus JA, Joh HD, Traystman RJ, Hurn PD. Postischemic angiogenic factor expression in stroke-prone rats. Exp Neurol 2002; 173:283-8. [PMID: 11822892 DOI: 10.1006/exnr.2001.7844] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Spontaneously hypertensive stroke-prone rats (SHRSP), a model for genetic stroke susceptibility, suffer spontaneous stroke and enhanced injury after experimental stroke, in part due to abnormal cerebrovascular development. We hypothesized that angiopoietin system genes in SHRSP may follow unique patterns of expression after experimentally induced stroke. SHRSP, hypertensive control rats (SHR), and normotensive controls (WKY) were subjected to experimental middle cerebral artery occlusion, and brain RNA was analyzed for expression of angiogenic genes. Expression of angiopoietin-2 increased after stroke in all rat strains and was significantly enhanced in SHRSP compared with control strains. In addition, expression of angiopoietin-1 and the angiopoietin receptor dropped markedly after stroke in SHRSP animals, but was not different after ischemia in SHR and WKY strains. Thus, the SHRSP brain elaborates a unique and specific pattern of angiopoietin system gene expression after stroke which may underlie stroke susceptibility of these rats.
Collapse
Affiliation(s)
- Michael M Wang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | | | | | |
Collapse
|
495
|
He Z, He YJ, Day AL, Simpkins JW. Proestrus levels of estradiol during transient global cerebral ischemia improves the histological outcome of the hippocampal CA1 region: perfusion-dependent and-independent mechanisms. J Neurol Sci 2002; 193:79-87. [PMID: 11790387 DOI: 10.1016/s0022-510x(01)00648-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We conducted this study to determine whether high physiological levels of estradiol (proestrus) could protect the hippocampal CA1 neurons following transient global ischemia. Ovariectomized or ovary-intact female rats were subjected to 20 min of ischemia and allowed to survive for 96 h. Estradiol was administered subcutaneously in a group of ovariectomized rats 24 h before ischemia induction. Ending serum estrogen levels were correlated to cerebral blood flow (CBF), histologic assessment and immunofluorescent caspase-3 active peptide (C-3AP) positive cell count. Estradiol administration significantly improved CBF in the hippocampus (compared with intact or ovariectomized rats) but not in the parietal cortex. No significant differences in CBF between intact or ovariectomized rats were noted. Estradiol administration maintained serum levels of the steroid in estradiol-treated rats-about 10 times that of intact animals and more than 20 times that of ovariectomized animals. Morphologically, live cell counts in estradiol-treated rats were significantly higher than in intact or ovariectomized rats. Live cell counts were also significantly higher in intact than ovariectomized rats. C-3AP positive cell counts were much higher in ovariectomized rats than in intact and estradiol-treated rats. In conclusion, proestrus levels of 17beta-estradiol protect hippocampal CA1 neurons against transient global ischemia, through mechanisms that appear to involve improvement of perfusion and inhibition of caspase-3 activity.
Collapse
Affiliation(s)
- Zhen He
- Department of Neurosurgery, College of Medicine, University of Florida, USA
| | | | | | | |
Collapse
|
496
|
Saleh TM, Saleh MC, Deacon CL, Chisholm A. 17beta-Estradiol release in the parabrachial nucleus of the rat evoked by visceral afferent activation. Mol Cell Endocrinol 2002; 186:101-10. [PMID: 11850126 DOI: 10.1016/s0303-7207(01)00665-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In the present investigation, in vivo microdialysis was used to measure the concentration of estrogen in the parabrachial nucleus (PBN) and plasma of male and ovariectomized female Sprague-Dawley rats supplemented with either estrogen (OVX-E(2)) or saline (OVX-S) following visceral afferent activation. Analysis of dialysate samples prior to vagal stimulation and in non-stimulated controls revealed a continuous concentration of estrogen in the PBN for all treatment groups (male, 38 +/-4 pg ml(-1); OVX-E(2), 38+/-5 pg ml(-1); OVX-S, 33 +/- 4 pg ml(-1)). This concentration of estrogen in the PBN was significantly increased during vagal stimulation in all groups (male, 64+/-4 pg ml(-1); OVX-E(2), 104+/-9 pg ml(-1); OVX-S, 80+/-6 pg ml(-1); P<0.05) and returned to pre-stimulation values within 2 h following termination of the stimulation. Immunohistochemical analysis revealed that estrogen receptor (ERalpha and ERbeta) density in males and ovariectomized saline-replaced female rats was significantly lower than that of estrogen-replaced female rats. These results suggest that estrogen is released into the PBN by an increase in visceral afferent traffic, however, alterations in estrogen receptor populations in the PBN may contribute to an attenuated physiological role of estrogen in the PBN of male and saline-replaced ovariectomized female rats.
Collapse
Affiliation(s)
- Tarek M Saleh
- Department of Anatomy and Physiology, Atlantic Veterinary College, University of Prince Edward Island, PEI, CIA 4P3, Charlottetown, Canada.
| | | | | | | |
Collapse
|
497
|
Ono H, Sasaki Y, Bamba E, Seki J, Giddings JC, Yamamoto J. Cerebral thrombosis and microcirculation of the rat during the oestrous cycle and after ovariectomy. Clin Exp Pharmacol Physiol 2002; 29:73-8. [PMID: 11906462 DOI: 10.1046/j.1440-1681.2002.03600.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
1. The effects of oestrogen on thrombogenesis and the cerebral microcirculation of the female rat were studied during the oestrous cycle and after ovariectomy. 2. Serum levels of oestradiol (E2) and plasma concentrations of nitric oxide (NO) metabolites were significantly greater at pro-oestrus than at dioestrus. Blood vessel diameter, mean red cell velocity, wall shear rate and blood flow at pro-oestrus were significantly higher than at dioestrus. Thrombotic tendency, assessed using a He-Ne laser-induced thrombosis model, was significantly decreased at pro-oestrus compared with dioestrus. 3. The long-term deprivation of oestrogen by ovariectomy significantly depressed serum levels of E2 and plasma concentrations of NO metabolites. Thrombotic tendency was significantly increased 4 weeks after ovariectomy. Vessel diameter, mean red cell velocity, wall shear rate and blood flow in pial arterioles were significantly reduced after ovariectomy. 4. Exogenous administration of oestrogen (17 beta-oestradiol) after surgery reversed the increased thrombotic tendency mediated by ovariectomy. 5. These results strongly indicate that oestrogen mediates beneficial effects on the cerebral microcirculation and moderates cerebral thrombotic mechanisms in the female rat.
Collapse
Affiliation(s)
- H Ono
- Laboratory of Physiology, Faculty of Nutrition, Kobe Gakuin University, Kobe, Japan
| | | | | | | | | | | |
Collapse
|
498
|
Yang SH, Perez E, Cutright J, Liu R, He Z, Day AL, Simpkins JW. Testosterone increases neurotoxicity of glutamate in vitro and ischemia-reperfusion injury in an animal model. J Appl Physiol (1985) 2002; 92:195-201. [PMID: 11744660 DOI: 10.1152/jappl.2002.92.1.195] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increasing evidence has demonstrated striking sex differences in the outcome of neurological injury. Whereas estrogens contribute to these differences by attenuating neurotoxicity and ischemia-reperfusion injury, the effects of testosterone are unclear. The present study was undertaken to determine the effects of testosterone on neuronal injury in both a cell-culture model and a rodent ischemia-reperfusion model. Glutamate-induced HT-22 cell-death model was used to evaluate the effects of testosterone on cell survival. Testosterone was shown to significantly increase the toxicity of glutamate at a 10 microM concentration, whereas 17beta-estradiol significantly attenuated the toxicity at the same concentration. In a rodent stroke model, ischemia-reperfusion injury was induced by temporal middle cerebral artery occlusion (MCAO) for 1 h and reperfusion for 24 h. To avoid the stress-related testosterone reduction, male rats were castrated and testosterone was replaced by testosterone pellet implantation. Testosterone pellets were removed at 1, 2, 4, or 6 h before MCAO to determine the duration of acute testosterone depletion effects on infarct volume. Ischemic lesion volume was significantly decreased from 239.6 +/- 25.9 mm(3) in control to 122.5 +/- 28.6 mm(3) when testosterone pellets were removed at 6 h before MCAO. Reduction of lesion volume was associated with amelioration of the hyperemia during reperfusion. Our in vitro and in vivo studies suggest that sex differences in response to brain injury are partly due to the consequence of damaging effects of testosterone.
Collapse
Affiliation(s)
- Shao-Hua Yang
- Department of Pharmacology and Neuroscience, Health Science Center at Fort Worth, University of North Texas, Fort Worth, Texas 76107, USA
| | | | | | | | | | | | | |
Collapse
|
499
|
Littleton-Kearney MT, Ostrowski NL, Cox DA, Rossberg MI, Hurn PD. Selective estrogen receptor modulators: tissue actions and potential for CNS protection. CNS DRUG REVIEWS 2002; 8:309-30. [PMID: 12353060 PMCID: PMC6741697 DOI: 10.1111/j.1527-3458.2002.tb00230.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Significant physiologic changes occur during menopause. Evidence exists to suggest that estrogen may be neuroprotective under specific conditions. However, there are limitations in the neuroprotection afforded by standard hormone therapy. Accordingly, alternative agents with selected estrogenic effects may hold even greater promise rather than conventional hormone replacement therapy for the prevention and treatment of CNS injury. Recently, a variety of selective estrogen receptor modulators (SERMs) have been developed to retain the favorable and minimize the adverse side effects of estrogens. This review focuses on the CNS and known neuroprotective effects of two specific SERMs, raloxifene and arzoxifene. Recent studies hint that raloxifene and arzoxifene are neuroprotective and may preserve some elements of cognitive function. However, the mechanism of action is not well described and it is unclear if the beneficial effects of SERMs rely on activation of estrogen receptors.
Collapse
|
500
|
Saleh TM, Cribb AE, Connell BJ. Reduction in infarct size by local estrogen does not prevent autonomic dysfunction after stroke. Am J Physiol Regul Integr Comp Physiol 2001; 281:R2088-95. [PMID: 11705796 DOI: 10.1152/ajpregu.2001.281.6.r2088] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Systemic estrogen administration in male rats has been shown to normalize the autonomic dysfunction and reduce the infarct size after permanent middle cerebral artery occlusion (MCAO). Therefore, the present investigation determined if local microinjection of estrogen at the site of the infarct also promoted recovery of autonomic function and reduction of the infarct size. Experiments were done in anesthetized (thiobutabarbitol sodium; 100 mg/kg) male Sprague-Dawley rats instrumented to record baseline and reflex changes in cardiovascular and autonomic parameters. The right middle cerebral artery was permanently occluded using bipolar coagulation. Local microinjection of estrogen into the insular cortex before MCAO significantly reduced the infarct size but did not attenuate the MCAO-induced autonomic dysfunction. Injection of ICI-182,780 alone significantly increased infarct area; however, the greater infarct area was not associated with enhanced autonomic dysfunction. These results suggest that within the insula, endogenous estrogen activity can affect the extent of MCAO-induced cell death, but extracortical central nervous system sites may be responsible for mediating the beneficial effects of estrogen on the autonomic disturbances.
Collapse
Affiliation(s)
- T M Saleh
- Department of Anatomy and Physiology, Atlantic Veterinary College, Univ. of Prince Edward Island, Charlottetown, Prince Edward Island, Canada C1A 4P3.
| | | | | |
Collapse
|