Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Minerva Endocrinol ; 35(2): 127-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20595940

ABSTRACT

Sex steroids are essential for reproduction and development in animals and humans, and sex steroids also play an important role in neuroprotection following brain injury. New data indicate that sex-specific responses to brain injury occur at the cellular and molecular levels. This review summarizes the current understanding of neuroprotection by sex steroids, particularly estrogen, androgen, and progesterone, based on both in vitro and in vivo studies. Better understanding of the role of sex steroids under physiological and pathological conditions will help us to develop novel effective therapeutic strategies for brain injury.


Subject(s)
Brain/metabolism , Gonadal Steroid Hormones/metabolism , Neuroprotective Agents/metabolism , Androgens/metabolism , Animals , Brain/pathology , Brain Injuries/drug therapy , Estrogens/metabolism , Evidence-Based Medicine , Gonadal Steroid Hormones/therapeutic use , Humans , Neuroprotective Agents/therapeutic use , Progesterone/metabolism , Wound Healing
2.
Neuroscience ; 169(2): 758-69, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20580788

ABSTRACT

Isoflurane preconditioning neuroprotection in experimental stroke is male-specific. The role of androgens in the ischemic sensitivity of isoflurane preconditioned male brain and whether androgen effects are androgen receptor dependent were assessed. Male C57BL/6 mice were implanted with flutamide (androgen receptor antagonist), or castrated and implanted with testosterone, dihydrotestosterone, flutamide, letrozole (aromatase inhibitor), or vehicle 7-13 days before preconditioning. P450 estrogen aromatase wild-type and knockout mice were also evaluated. All mice were preconditioned for 4 h with 0% (sham preconditioning) or 1% isoflurane (isoflurane preconditioning) and recovered for 24 h. Mice then underwent 2 h of middle cerebral artery occlusion and were evaluated 22 h later for infarct volume. For neurobehavioral outcomes, sham and isoflurane preconditioned castrated male+/-dihydrotestosterone groups underwent 1 h of middle cerebral artery occlusion followed by 9 days of reperfusion. Isoflurane preconditioning neuroprotection relative to infarct volume outcomes were testosterone and dihydrotestosterone dose-specific and androgen receptor-dependent. Relative to long-term neurobehavioral outcomes, front paw sensorimotor function improved in isoflurane preconditioned mice regardless of androgen status while androgen replacement independently improved sensorimotor function. In contrast, isoflurane preconditioning improved cognitive function in castrates lacking endogenous androgens, but this improvement was absent in androgen replaced mice. Our findings suggest that androgen availability during isoflurane preconditioning may influence infarct volume and neurobehavioral outcomes in male mice following experimental stroke.


Subject(s)
Androgens/physiology , Anesthetics, Inhalation/therapeutic use , Ischemic Attack, Transient/prevention & control , Isoflurane/therapeutic use , Neuroprotective Agents/therapeutic use , Androgens/blood , Androgens/pharmacology , Animals , Aromatase/genetics , Aromatase Inhibitors/pharmacology , Brain/metabolism , Brain/pathology , Dihydrotestosterone/blood , Dihydrotestosterone/pharmacology , Estradiol/blood , Exploratory Behavior/drug effects , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/prevention & control , Infarction, Middle Cerebral Artery/psychology , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/psychology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Orchiectomy , Receptors, Androgen/physiology , Recognition, Psychology/drug effects , Testosterone/blood , Testosterone/pharmacology
3.
Neuroscience ; 166(2): 476-81, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-20035840

ABSTRACT

Activation of poly (ADP-ribose) polymerases (PARP) contributes to ischemic damage by causing neuronal nicotinamide adenine dinucleotide (NAD(+)) depletion, release of apoptosis-inducing factor and consequent caspase-independent cell death. PARP-mediated cell death is sexually dimorphic, participating in ischemic damage in the male brain, but not the female brain. We tested the hypothesis that androgen signaling is required for this male-specific neuronal cell death pathway. We observed smaller damage following focal cerebral ischemia (MCAO) in male PARP-1 knockout mice compared to wild type (WT) as well as decreased damage in male mice treated with the PARP inhibitor PJ34. Protection from ischemic damage provided by PJ-34 in WT mice is lost after removal of testicular androgens (CAST) and rescued by androgen replacement. CAST PARP-1 KO mice exhibit increased damage compared to intact male KO mice, an effect reversed by androgen replacement in an androgen receptor-dependent manner. Lastly, we observed that ischemia causes an increase in PARP-1 expression that is diminished in the absence of testicular androgens. Our data indicate that PARP-mediated neuronal cell death in the male brain requires intact androgen-androgen receptor signaling.


Subject(s)
Androgens/metabolism , Brain Ischemia/metabolism , Brain/metabolism , Cell Death/physiology , Poly(ADP-ribose) Polymerases/metabolism , Sex Characteristics , Analysis of Variance , Animals , Brain/drug effects , Brain/pathology , Brain Ischemia/pathology , Cell Death/drug effects , Dihydrotestosterone/pharmacology , Female , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Orchiectomy , Poly(ADP-ribose) Polymerases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
4.
Neuroscience ; 158(3): 1098-111, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-18597949

ABSTRACT

The profound damage to the CNS caused by ischemic lesions has been well documented. Yet, relatively little is known about the contribution to and effects on the immune system during stroke. We have focused on both early and late events in the peripheral immune system during stroke in mice and have observed an early activation of splenocytes that conceivably could result in immune-mediated damage in the developing CNS lesion, followed by global immunosuppression that affects the spleen, thymus, lymph nodes and circulation. While this second immunosuppressive phase may not directly enhance infarction size, it without doubt leads to an inability to respond to antigenic challenges, thereby enhancing the risk for crippling systemic infection and septicemia in stroke survivors. These novel findings advocate the need to develop or effectively utilize agents that can block early neural splenic activation and modulate immune cells specific for brain antigens as a means to prevent mobilization of T and B cells carrying a cytokine death warrant to the brain. Equally important for the recovering stroke patient are approaches that can derail the second phase of immune dysfunction and restore the ability to mount a defense against systemic infectious insults.


Subject(s)
Brain Ischemia/immunology , Chemotaxis, Leukocyte/immunology , Cytokines/immunology , Immune System/immunology , Immune Tolerance/immunology , Stroke/immunology , Animals , Brain Ischemia/physiopathology , Cell Death/immunology , Disease Models, Animal , Humans , Immune System/physiopathology , Mice , Rats , Spleen/immunology , Spleen/pathology , Spleen/physiopathology , Stroke/physiopathology
5.
Eur J Neurosci ; 24(9): 2567-74, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17100844

ABSTRACT

The survival of rat Purkinje cell (PCs) cerebellar cultures was used to test the hypothesis that progesterone is protective against oxygen-glucose deprivation through potentiation of GABA(A) receptor activity. Electrophysiological recordings confirm that PCs develop robust excitatory and inhibitory synapses in culture. Exposure of cultured PCs to increasing concentrations of progesterone during oxygen-glucose deprivation revealed a concentration-dependent protection by progesterone, with significant protection observed at physiological concentrations, as low as 10 nm. The concurrent application of the GABA(A) receptor antagonist picrotoxin (100 microm) completely abolished the neuroprotection afforded by progesterone, indicating that progesterone is neuroprotective through activation of GABA(A) receptors. Progesterone potentiates GABA(A) receptor activity indirectly through its metabolites, such as allopregnanolone (ALLO). Therefore, ALLO was applied to PC cultures and was observed to produce significant protection at all concentrations tested, from 10 to 1000 nm. Finally, the inhibition of progesterone metabolism with finasteride abolished the protection afforded by progesterone without having any effect on the neuroprotection caused by ALLO. These data indicate that progesterone protects cerebellar PCs at physiological concentrations through a GABA-active metabolite.


Subject(s)
Cell Hypoxia/physiology , Cerebellum/metabolism , Progesterone/metabolism , Purkinje Cells/metabolism , Animals , Apoptosis/physiology , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Female , Glucose/deficiency , Hypoxia-Ischemia, Brain/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Male , Oxygen/metabolism , Patch-Clamp Techniques , Purkinje Cells/pathology , Rats , Rats, Sprague-Dawley
6.
Curr Drug Metab ; 5(3): 225-34, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15180492

ABSTRACT

Advances in a multitude of disciplines support an emerging role for cytochrome P450 enzymes and their metabolic substrates and end-products in the pathogenesis and treatment of central nervous system disorders, including acute cerebrovascular injury, such as stroke, chronic neurodegenerative disease, such as Alzheimer's and Parkinson's disease, as well as epilepsy, multiple sclerosis and psychiatric disorders, including anxiety and depression. The neural tissue contains its own unique set of P450 genes that are regulated in a manner that is distinct from their molecular regulation in peripheral tissue. Furthermore, brain P450s catalyze the formation of important brain signaling molecules, such as neurosteroids and eicosanoids, and metabolize substrates as diverse as vitamins A and D, cholesterol, bile acids, as well as centrally acting drugs, anesthetics and environmental neurotoxins. These unique characteristics allow this family of proteins and their metabolites to perform such vital functions in brain as neurotrophic support, neuroprotection, control of cerebral blood flow, temperature control, neuropeptide release, maintenance of brain cholesterol homoeostasis, elimination of retinoids from CNS, regulation of neurotransmitter levels and other functions important in brain physiology, development and disease.


Subject(s)
Brain Diseases/enzymology , Cytochrome P-450 Enzyme System/metabolism , Anesthetics/metabolism , Animals , Arachidonic Acid/metabolism , Bile Acids and Salts/metabolism , Brain Diseases/metabolism , Cholesterol/metabolism , Cytochrome P-450 Enzyme System/genetics , Humans , Pharmaceutical Preparations/metabolism , Steroids/biosynthesis
7.
Neuroscience ; 114(1): 217-27, 2002.
Article in English | MEDLINE | ID: mdl-12207967

ABSTRACT

Caspases are proteases involved in various physiological and pathological processes in the nervous system, including development and pathogenesis. GRASP-1 is a recently identified neuronal substrate of caspase-3-subfamily caspases. It is a Ras-guanine nucleotide exchange factor (RasGEF) that interacts with the glutamate receptor interacting protein (GRIP). This alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptor/GRIP protein complex has been proposed to be involved in AMPA receptor synaptic targeting. The caspase-3 cleavage of GRASP-1 separates the N-terminal RasGEF catalytic domain from the C-terminal GRIP-interacting region, potentially disrupting regulation of the RasGEF activity by GRIP. To examine the regulation and regional distribution of the caspase-3 cleavage of GRASP-1 in vivo, we generated a cleavage site-specific antibody, termed CGP, against the cleaved N-terminal fragment of GRASP-1. Using this antibody, we have examined the caspase cleavage of GRASP-1 during postnatal development and following ischemia in mice. We found that caspase cleavage of GRASP-1 occurs in specific brain regions in a time-dependent manner during development and ischemia. This data provides an important account of the brain areas that might require caspase-3 activity in postnatal development and ischemic damage, which has not been documented. It also demonstrates that the CGP antibody is a powerful tool for studying neuronal activity of the caspase-3-subfamily caspases in vivo.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , Caspases/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , ras Guanine Nucleotide Exchange Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Antibody Specificity/immunology , Brain/cytology , Brain/growth & development , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Caspase 3 , Catalytic Domain/physiology , Cell Death/physiology , Cells, Cultured , Functional Laterality/physiology , Immunohistochemistry , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Membrane Proteins , Mice , Neostriatum/metabolism , Neostriatum/physiopathology , Neurons/cytology , Protein Structure, Tertiary/physiology
8.
CNS Drug Rev ; 8(3): 309-30, 2002.
Article in English | MEDLINE | ID: mdl-12353060

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.


Subject(s)
Central Nervous System/drug effects , Estrogen Receptor Modulators/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Breast Neoplasms/drug therapy , Cardiovascular System/drug effects , Central Nervous System/physiology , Cognition/drug effects , Estrogen Antagonists/adverse effects , Estrogen Antagonists/therapeutic use , Estrogen Receptor Modulators/chemistry , Estrogen Receptor Modulators/pharmacology , Female , Hormone Replacement Therapy , Humans , Hypothalamo-Hypophyseal System/drug effects , Male , Neuroprotective Agents/pharmacology , Osteoporosis, Postmenopausal/drug therapy , Piperidines/pharmacology , Piperidines/therapeutic use , Raloxifene Hydrochloride/pharmacology , Raloxifene Hydrochloride/therapeutic use , Receptors, Neurotransmitter/drug effects , Structure-Activity Relationship , Tamoxifen/adverse effects , Tamoxifen/therapeutic use , Thiophenes/pharmacology , Thiophenes/therapeutic use , Uterus/drug effects
9.
Am J Respir Crit Care Med ; 164(8 Pt 1): 1470-5, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11704598

ABSTRACT

Obese females are less predisposed to sleep-disordered breathing and have higher serum leptin levels than males of comparable body weight. Because leptin is a powerful respiratory stimulant, especially during sleep, we hypothesized that the elevated leptin level is necessary to maintain normal ventilatory control in obese females. We examined ventilatory control during sleep and wakefulness in male and female leptin-deficient obese C57BL/6J-Lep(ob) mice, wild-type C57BL/6J mice with dietary-induced obesity and high serum leptin levels, and normal weight wild-type C57BL/6J mice. Both male and female C57BL/6J-Lep(ob) mice had depressed hypercapnic ventilatory response (HCVR) in comparison with wild-type animals. In comparison with male C57BL/6J-Lep(ob) mice, female C57BL/6J-Lep(ob) mice had reduced HCVR and respiratory drive (a ratio of tidal volume to inspiratory time) both during non-rapid eye movement (NREM) sleep and wakefulness. In contrast, the HCVR did not differ between sexes in wild-type mice during NREM sleep and wakefulness, but was lower in females during REM sleep. Thus, leptin deficiency in female obesity is even more detrimental to hypercapnic ventilatory control during wakefulness and NREM sleep than in obese, leptin-deficient males.


Subject(s)
Leptin/deficiency , Obesity/complications , Respiratory Insufficiency/etiology , Animals , Carbon Dioxide , Female , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Obesity/metabolism , Obesity/physiopathology , Respiratory Insufficiency/physiopathology , Severity of Illness Index , Sex Factors
10.
Proc Natl Acad Sci U S A ; 98(20): 11824-8, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11553785

ABSTRACT

The relationship between stressful life events and the onset of disease is well documented. However, the role of psychological stress as a risk factor for life-threatening cerebrovascular insults such as stroke remains unspecified, but could explain individual variation in stroke outcome. To discover the mechanisms through which psychological stress may alter stroke outcome, we modeled the effects of chronic social intimidation and stress on ischemia-induced bcl-2 expression and early neuronal cell loss resulting from cerebral artery occlusion in mice (C57BL/6). The bcl-2 protooncogene promotes cell survival and protects against apoptosis and cellular necrosis in numerous neurodegenerative disorders, including stroke. In our study, male mice were chronically exposed to aggressive social stimuli before induction of a controlled, mild ischemic insult. Stressed mice expressed approximately 70% less bcl-2 mRNA than unstressed mice after ischemia. In addition, social stress greatly exacerbated infarct in wild-type mice but not in transgenic mice that constitutively express increased neuronal bcl-2. Despite similar postischemic concentrations of corticosterone, the major stress hormone in mice, high corticosterone concentrations were significantly correlated with larger infarcts in wild-type mice but not bcl-2 transgenic mice. Thus, enhanced bcl-2 expression offsets the potentially deleterious consequences of high postischemic plasma corticosterone concentrations. Taken together, these data demonstrate that stressful prestroke social milieu strongly compromises an endogenous molecular mechanism of neuroprotection in injured brain and offer a new behavioral target for stroke therapy.


Subject(s)
Genes, bcl-2 , Ischemic Attack, Transient/psychology , Music/psychology , Proto-Oncogene Proteins c-bcl-2/genetics , Suppression, Genetic , Animals , Corticosterone/blood , Ischemic Attack, Transient/genetics , Ischemic Attack, Transient/physiopathology , Male , Mice , Mice, Inbred C57BL , Regression Analysis , Reperfusion , Ribonucleases , Time Factors , Treatment Outcome , WT1 Proteins/genetics
11.
J Neurosci ; 21(19): 7543-50, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567044

ABSTRACT

Female rodents producing endogenous estrogens are protected from stroke damage in comparison with male counterparts. This natural protection is lost after ovariectomy or reproductive senescence. The aim of this study is to determine whether estrogen reduces early neuronal injury and cell loss after ischemia by increasing the expression of Bcl-2. Male, intact female, ovariectomized, and estrogen-repleted ovariectomized rats were subjected to middle cerebral artery occlusion, and 22 hr later the level and localization of Bcl-2 mRNA and protein were determined. The levels of post-ischemic bcl-2 mRNA and protein were increased exclusively in neurons within the peri-infarct region. Intact females and estrogen-treated castrates demonstrated increased bcl-2 mRNA and protein expression compared with males and estrogen-deficient females, accompanied by a decrease in infarct size. To test the hypothesis that the neuroprotective mechanism of estrogen functions via Bcl-2, we compared ischemic outcome in male, female, and ovariectomized wild-type mice and mice overexpressing Bcl-2 exclusively in neurons. Wild-type female mice sustained smaller infarcts compared with males. Bcl-2 overexpression reduced infarct size in males, but provided no added protection in the female. Moreover, ovariectomy exacerbated infarction in wild-type females, but had no effect in Bcl-2 overexpressors. These data indicate that overexpression of Bcl-2 simulates the protection against ischemic injury conferred by endogenous female sex steroids. We concluded that estrogen rescues neurons after focal cerebral ischemia by increasing the level of Bcl-2 in peri-infarct regions and that estrogen-induced bcl-2 gene expression is an important downstream component of neuronal protection in female stroke.


Subject(s)
Cerebral Infarction/prevention & control , Estrogens/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Stroke/metabolism , Transgenes , Animals , Cell Death/drug effects , Cerebral Infarction/metabolism , Cerebral Infarction/pathology , Disease Susceptibility , Estrogen Replacement Therapy , Estrogens/pharmacology , Female , Gene Expression , Gene Expression Regulation/drug effects , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Ovariectomy , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sex Factors , Stroke/genetics , Stroke/pathology , Transcriptional Activation
12.
Stroke ; 32(8): 1920-5, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11486126

ABSTRACT

BACKGROUND AND PURPOSE: It is not known whether preischemic exposure to anesthetic agents affects the amount of damage from transient focal ischemia that occurs after cessation of the anesthetic. We compared the effect of prior exposure to halothane or propofol on infarction size after transient middle cerebral artery occlusion (MCAO) induced in the awakening animal to test the hypothesis that anesthetic type and exposure duration would independently affect the amount of brain injury. METHODS: Male Wistar rats (weight, 200 to 300 g) were anesthetized briefly with halothane for placement of hemodynamic instrumentation. Twenty-four hours later, rats were treated with either a short (approximately 1 hour) or long (8 hours) duration of inhaled halothane (1% to 2%) or intravenous propofol (10 mg/kg bolus, 30 mg/kg per hour infusion). Each cohort (n=8 per group) was then subjected to 2-hour MCAO by the intraluminal suture technique. All anesthesia was discontinued once MCAO was achieved. Infarct volume was measured at 22 hours of reperfusion. In a second cohort, regional cerebral blood flow (CBF) was measured ([(14)C]iodoantipyrine autoradiography) at end-occlusion in short-duration halothane (n=5) or short-duration propofol (n=5) anesthesia groups and in corresponding surgical shams (n=3 each). RESULTS: Pericranial temperature, PaO(2), PaCO(2), and blood pressure were controlled and not different among groups before or during occlusion. MCAO resulted in a similar immediate reduction in laser-Doppler flow signal after discontinuation of anesthesia in the awakening animals. Infarct volume was smaller in rats exposed to short-duration halothane in cortex (87.5+/-16.6 mm(3)) (mean+/-SEM) and caudoputamen (38.3+/-13.7 mm(3)) compared with rats exposed to short-duration propofol (cortex, 177.5+/-16.9 mm(3); caudoputamen, 47.8+/-2.9 mm(3)). Infarct volume was not different in long-duration halothane versus long-duration propofol treatment. Absolute cortical or caudoputamen intraischemic CBF was not different between short-duration halothane or short-duration propofol treatment. CONCLUSIONS: These data demonstrate that short-duration halothane exposure before MCAO in the awakening animal attenuates infarction volume compared with propofol. This protection by halothane is not mediated through preservation of intraischemic CBF. Longer durations of halothane exposure may activate secondary injury pathways, which negate the protective effects of short-term halothane preischemic treatment.


Subject(s)
Anesthetics, Inhalation/administration & dosage , Anesthetics, Intravenous/administration & dosage , Halothane/administration & dosage , Propofol/administration & dosage , Stroke/drug therapy , Administration, Inhalation , Anesthesia Recovery Period , Animals , Blood Flow Velocity/drug effects , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Drug Administration Schedule , Infarction, Middle Cerebral Artery/complications , Injections, Intravenous , Intraoperative Period , Laser-Doppler Flowmetry , Male , Rats , Rats, Wistar , Stroke/etiology , Stroke/physiopathology
13.
Stroke ; 32(7): 1613-20, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11441209

ABSTRACT

BACKGROUND AND PURPOSE: The potent final sigma(1)-receptor ligand 4-phenyl-1-(4-phenylbutyl) piperidine (PPBP) provides neuroprotection in experimental stroke. We tested the hypothesis that PPBP attenuates striatal tissue damage after middle cerebral artery occlusion (MCAO) by a mechanism involving reduction of ischemia-evoked nitric oxide (NO) production. Furthermore, we determined whether the agent fails to protect ischemic brain when neuronal nitric oxide synthase (nNOS) is genetically deleted or pharmacologically inhibited (selective nNOS inhibitor, 7-nitroindazole [7-NI]). METHODS: Halothane-anesthetized adult male Wistar rats were subjected to 2 hours of MCAO by the intraluminal filament occlusion technique. All physiological variables were controlled during the ischemic insult. In vivo striatal NO production was estimated via microdialysis by quantification of local, labeled citrulline recovery after labeled arginine infusion. In a second series of experiments, nNOS null mutants (nNOSKOs) and the genetically matched wild-type (WT) strain were treated with 90 minutes of MCAO. Brains were harvested at 22 hours of reperfusion for measurement of infarction volume by triphenyltetrazolium chloride histology. RESULTS: PPBP attenuated infarction volume at 22 hours of reperfusion in cerebral cortex and striatum and markedly attenuated NO production in ischemic and nonischemic striatum during occlusion and early reperfusion. Treatment with 7-NI mimicked the effects of PPBP. In WT mice, infarction volume was robustly decreased by both PPBP and 7-NI, but the efficacy of PPBP was not altered by pharmacological nNOS inhibition in combined therapy. In contrast, PPBP did not decrease infarction volume in nNOSKO mice. CONCLUSIONS: These data suggest that the mechanism of neuroprotection of PPBP in vivo is through attenuation of nNOS activity and ischemia-evoked NO production. Neuroprotective effects of PPBP are lost when nNOS is not present or is inhibited; therefore, PPBP likely acts upstream from NO generation and its subsequent neurotoxicity.


Subject(s)
Haloperidol/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Neurons/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide/biosynthesis , Receptors, sigma/agonists , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Ischemia/complications , Brain Ischemia/metabolism , Citrulline/analysis , Enzyme Inhibitors/pharmacology , Haloperidol/analogs & derivatives , Indazoles/pharmacology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Mice , Mice, Knockout , Neurons/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Rats , Rats, Wistar , Sigma-1 Receptor
14.
Neurosci Biobehav Rev ; 25(4): 325-42, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11445138

ABSTRACT

Stroke in humans is associated with deficits in sensorimotor and cognitive function. Consequently, many stroke researchers recently have expanded their techniques to assess cognitive and behavioral correlates of histologically-determined stroke damage in animal models. Although the incorporation of functional outcome assessment represents an important step forward in stroke research, reports of middle cerebral artery occlusion (MCAO) induced behavioral deficits often conflict, and a significant correlation between post-stroke histology and behavior has been reported in few stroke studies. Discrepancies in behavioral outcomes among studies may be due to several factors, such as method of MCAO, duration of occlusion, strain, the timing and method of the behavioral testing and the laboratory environment. Furthermore, proper experimental and control groups, necessary to rule out potential confounding factors during cognitive testing, often are not incorporated. The goal of this review is: (1) to provide a description of the techniques most commonly employed to assess functional outcome after (MCAO) in rodents and (2) to identify potential confounding factors that may interfere with a clear interpretation of the behavioral data.


Subject(s)
Behavior, Animal/physiology , Cognition/physiology , Stroke/psychology , Animals , Anxiety/psychology , Disease Models, Animal , Humans , Psychomotor Performance/physiology
15.
Magn Reson Med ; 46(1): 183-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11443725

ABSTRACT

MRI studies using mouse brain models of ischemia are becoming a valuable tool for understanding the mechanism of stroke, since transgenic models are now available. However, the small size of the mouse brain and the surgical complexity of creating ischemia in mice make it technically challenging to obtain high-quality MRI data. Therefore, there are few reports of MRI studies in murine cerebral ischemia. In this project a newly developed rapid 3D diffusion-weighted imaging (DWI) technique was applied to study experimental stroke in a mouse model of reversible middle cerebral artery occlusion (MCAO). Ischemic volumes were successfully delineated using this 3D whole-brain imaging technique with high spatial (0.34 x 0.5 x 1.0 mm(3) before zero-filling) and temporal (7 min) resolution. The 3D observation revealed the characteristic evolution of stroke after transient MCAO. There was a temporarily high diffusion constant in the cortex during early reperfusion, followed by a secondary energy failure in the cortex and caudate-putamen at 6 and 21 h of reperfusion. Magn Reson Med 46:183-188, 2001.


Subject(s)
Brain/pathology , Magnetic Resonance Imaging/methods , Stroke/pathology , Animals , Brain Ischemia/pathology , Imaging, Three-Dimensional , Mice
16.
Am J Physiol Heart Circ Physiol ; 281(1): H155-60, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11406480

ABSTRACT

Estrogen protects the brain from experimental cerebral ischemia, likely through both vascular and neuronal cellular mechanisms. The purpose of this study was to determine whether chronic estrogen treatment in males and repletion in ovariectomized (Ovx) females reverses abnormalities in pial arteriolar reactivity during reperfusion from global forebrain ischemia (4-vessel occlusion, 15 min) and whether the site of protection is vascular endothelium. Male and Ovx female rats were implanted with either placebo or a 25-microg 17 beta-estradiol pellet 10 days before ischemia. With the use of intravital microscopy, pial vessel dilation to ACh (10 microM) and S-nitroso-N-acetyl-penicillamine (SNAP; 1 microM) and vasoconstriction to serotonin (10 microM) was examined in situ at 30--60 min of reperfusion. Postischemic changes in vessel diameter were compared with preischemic values for each agent. Postischemic response to both ACh and SNAP was lost in males and Ovx females, but not in estrogen pellet-implanted males and estrogen-implanted Ovx females, suggesting that estrogen protects both endothelial and smooth muscle-mediated vasodilation. Ischemia blunted vessel constriction to serotonin regardless of treatment. These data demonstrate that estrogen acts as a vasoprotective agent within the cerebral circulation and can improve microvascular function under conditions of an acutely evolving ischemic pathology.


Subject(s)
Brain Ischemia/physiopathology , Estradiol/pharmacology , Pia Mater/blood supply , Reperfusion Injury/physiopathology , Vasodilation , Acetylcholine/pharmacology , Animals , Female , Male , Microcirculation/drug effects , Ovariectomy , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Rats , Rats, Wistar , S-Nitroso-N-Acetylpenicillamine , Serotonin/pharmacology , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
17.
Contemp Top Lab Anim Sci ; 40(2): 9-12, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300680

ABSTRACT

We examined retrospectively the effects of brief halothane anesthesia during daily gavage administration of vehicle on gavage-related complications and body weight in ovariectomized female Wistar rats. The number of gavage-related deaths or animals requiring euthanasia due to gavage problems was dramatically reduced, but the occurrence of incomplete vehicle retention during gavage was increased appreciably in halothane-anesthetized animals. Halothane-anesthetized rats maintained daily body weight for a longer period than did awake animals. Our observations suggest that the use of brief inhalational anesthesia reduces gavage-associated death and euthanasia due to esophageal trauma and minimizes stress-related weight loss.


Subject(s)
Anesthesia/veterinary , Anesthetics, Inhalation/administration & dosage , Halothane/administration & dosage , Intubation, Gastrointestinal/adverse effects , Intubation, Gastrointestinal/veterinary , Animals , Body Weight/drug effects , Esophagus/injuries , Female , Longevity/drug effects , Ovariectomy , Rats , Rats, Wistar , Retrospective Studies , Stress, Physiological , Weight Loss
18.
Brain Res ; 900(1): 137-42, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11325356

ABSTRACT

Estrogen has demonstrated great potential as a therapeutic agent in focal ischemic brain injury, as exogenous beta-estradiol has proven beneficial in a variety of focal stroke models. In contrast, the relatively few studies of estrogen's efficacy in transient forebrain ischemia have produced inconsistent results. The present study was therefore designed to clarify estrogen's neuroprotective potential in selective hippocampal neuronal injury resulting from four-vessel occlusion in the rat. Female Wistar rats (normal, ovariectomized, or ovariectomized and estradiol-treated) received 5 or 10 min of ischemia. No differences in hippocampal cell loss were found amongst the groups with 10 min of ischemia. Amongst the groups with 5 min of ischemia, the mildest injury was found in the ovariectomized animals, which lost only 32% of their CA1 pyramidal cells. In comparison, mean cell losses were 54% and 49%, respectively, in intact females and in ovariectomized animals with estradiol replacement. Linear regression analysis demonstrated a highly significant relationship between cell loss and plasma estradiol levels. The mechanism by which exogenous and endogenous estrogen exacerbated the injury is unclear, as estrogen has many neuroprotective effects. On the other hand, many other reported effects of estrogen in hippocampal area CA1 might confer increased sensitivity to ischemia, either by modulating the excitatory effects of glutamate or by modifying the inhibitory effects of GABA. Determining how to modulate the various competing effects of estrogen is of both theoretical and practical importance, as it is now clear that one cannot assume that estrogen administration will always improve outcome in cerebral ischemia.


Subject(s)
Estradiol/therapeutic use , Hippocampus/blood supply , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Estradiol/blood , Estradiol/toxicity , Estrogen Replacement Therapy/adverse effects , Female , Hippocampus/pathology , Ischemic Attack, Transient/pathology , Neuroprotective Agents/toxicity , Organ Specificity , Ovariectomy , Progesterone/blood , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Rats , Rats, Wistar
19.
Stroke ; 32(3): 783-7, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11239202

ABSTRACT

BACKGROUND AND PURPOSE: Recent results suggest that selective inhibitors of presynaptic neuronal ion channels can diminish glutamate release during cerebral ischemia and modulate excitotoxic cell death. The aim of the present study was to evaluate lamotrigine (LTG), an antiepileptic that inhibits presynaptic sodium and voltage-sensitive calcium channels, as a potential stroke resuscitation agent in the rat. Three dosages of LTG were examined for effect on infarction volume and sensorimotor behavioral recovery after middle cerebral artery (MCA) occlusion. METHODS: Halothane-anesthetized male Wistar rats were subjected to 2 hours of MCA occlusion by the intraluminal occlusion technique. Physiological variables were controlled, and ipsilateral cortical perfusion was monitored by laser Doppler flowmetry throughout ischemia. At onset of reperfusion, rats received intravenous LTG 5, 10, or 20 mg/kg or PBS (n=9 to 11 per group) during 15 minutes. Behavioral assessment was completed at 3 and 7 days after stroke, and the brain was harvested for histology (triphenyltetrazolium chloride staining). RESULTS: Values are mean+/-SE. Cortical infarction volumes were unchanged in LTG-treated animals: 14+/-6% of contralateral cortex at 5 mg/kg LTG, 17+/-7% at 10 mg/kg, and 30+/-6% at 20 mg/kg, versus saline-treated cohorts (12+/-3%; P:=0.19; n=9). Caudate-putamen infarction injury was also unchanged (37+/-11% of contralateral caudate-putamen at 5 mg/kg LTG, 44+/-8% at 10 mg/kg, and 65+/-9% at 20 mg/kg versus saline (38+/-11%; P:=0.18). Total infarction was not different among groups (P:=0.15). Consistent with histology, behavioral outcomes were unimproved by treatment. CONCLUSIONS: Histological damage and behavioral recovery at 7 days after MCA occlusion was not altered by LTG treatment over the dosage range used in the present study.


Subject(s)
Anticonvulsants/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Stroke/drug therapy , Triazines/administration & dosage , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Infarction, Middle Cerebral Artery/complications , Lamotrigine , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Recovery of Function/drug effects , Reperfusion , Stroke/etiology , Treatment Failure
20.
Stroke ; 32(3): 796-802, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11239204

ABSTRACT

BACKGROUND AND PURPOSE: Estrogen is a known neuroprotective and vasoprotective agent in experimental cerebral ischemia. Preischemic steroid treatment protects animals of both sexes from focal cerebral ischemia. This study determined whether intravenous estrogen acts as a vasodilator when administered on reperfusion and whether the resulting increase in cerebral blood flow (CBF) provides tissue protection from middle cerebral artery occlusion. METHODS: Adult male Wistar rats were treated with reversible middle cerebral artery occlusion (2 hours), then infused with intravenous estrogen (Premarin; 1 mg/kg) or vehicle during the first minutes of reperfusion (n=15 per group). Cortical laser-Doppler flowmetry was used to assess adequacy of occlusion. Ischemic lesion volume was determined at 22 hours after occlusion by 2,3,5-triphenyltetrazolium chloride staining and image analysis. Cortical and striatal CBF was measured by (14)[C]iodoantipyrine autoradiography at 10 (n=10) or 90 (n=11) minutes of reperfusion. RESULTS: As expected, supraphysiological plasma estrogen levels were achieved during reperfusion (estrogen, 198+/-45 pg/mL; vehicle, 6+/-5; P:=0.001). Physiological variables were controlled and not different between groups. Total hemispheric infarction was reduced in estrogen-treated rats (estrogen, 49+/-4% of ipsilateral structure; vehicle, 33+/-5%; P:=0.02), which was most pronounced in striatum (estrogen, 40+/-6% of ipsilateral striatum; vehicle, 60+/-3%; P:=0.01). CBF recovery was strikingly increased by estrogen infusion at 10 minutes in frontal (estrogen, 102+/-12 mL/100 g per minute; vehicle, 45+/-15; P:=0.01) and parietal cortex (estrogen, 74+/-15 mL/100 g per minute; vehicle, 22+/-13; P:=0.028) and throughout striatum (estrogen, 87+/-13 mL/100 g per minute; vehicle, 25+/-20; P:=0.02). Hemispheric volume with low CBF recovery (eg, <20 mL/100 g per minute) was smaller in estrogen-treated animals (estrogen, 73+/-18 mm(3); vehicle, 257+/-46; P:=0.002). However, differences in CBF recovery could not be appreciated between groups by 90 minutes of reperfusion. CONCLUSIONS: Acute estrogen therapy during reperfusion improves tissue outcome from experimental stroke. The steroid rapidly promotes CBF recovery and reduces hemispheric no-reflow zones. This beneficial effect appears only during early reperfusion and likely complements other known mechanisms by which estrogen salvages brain from focal necrosis.


Subject(s)
Brain Ischemia/prevention & control , Cerebrovascular Circulation/drug effects , Estrogens/administration & dosage , Stroke/drug therapy , Animals , Brain/blood supply , Brain/drug effects , Brain/pathology , Brain Ischemia/etiology , Brain Ischemia/physiopathology , Disease Models, Animal , Estrogens/metabolism , Hemodynamics/drug effects , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/physiopathology , Infusions, Intravenous , Male , Rats , Rats, Wistar , Stroke/etiology , Stroke/pathology , Stroke/physiopathology , Tissue Distribution , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL