Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Vitam Horm ; 113: 291-312, 2020.
Article in English | MEDLINE | ID: mdl-32138953

ABSTRACT

Vasopressin is a peptide hormone produced in the hypothalamus and released from the posterior pituitary. Secretion of vasopressin is followed by activation of its receptors V1a, V1b, and V2 throughout the body. Each receptor type is responsible for a specific function of vasopressin. For example, V1a receptor activation triggers vasoconstriction, V1b receptor is responsible for modulation of mood and behavior, and V2 receptor induces water reabsorption in the kidney. Vasopressin is known to regulate blood pressure, blood osmolality, and blood volume. The effects of V1a and V2 receptors can be amplified when vasopressin is secreted in excessive amounts, and this condition may be experienced by patients undergoing a disease or stress. In pathological conditions such as stroke, traumatic brain injury, subarachnoid hemorrhage, liver disease, and other diseases, vasopressin can exacerbate brain edema. Oversecretion of vasopressin unleashes deleterious pathways leading to hyponatremia and brain edema. This book chapter describes important mechanisms and pathways linking vasopressin and brain edema triggered by various conditions.


Subject(s)
Brain Edema/metabolism , Receptors, Vasopressin/metabolism , Vasopressins/metabolism , Humans , Signal Transduction
2.
PLoS One ; 12(8): e0183985, 2017.
Article in English | MEDLINE | ID: mdl-28854286

ABSTRACT

BACKGROUND: Ischemic stroke is often complicated by brain edema, disruption of blood-brain barrier (BBB), and uncontrolled release of arginine-vasopressin (AVP). Conivaptan, a V1a and V2 receptor antagonist, reduces brain edema and minimizes damage to the blood-brain barrier after stroke. Most stroke patients do not receive treatment immediately after the onset of brain ischemia. Delays in therapy initiation may worsen stroke outcomes. Therefore, we designed a translational study to explore the therapeutic time window for conivaptan administration. METHODS: Mice were treated with conivaptan beginning 3, 5, or 20 hours after 60-minute focal middle cerebral artery occlusion. Treatments were administered by continuous IV infusion for a total of 48 hours. Brain edema and blood-brain barrier (BBB) disruption were evaluated at endpoint. RESULTS: Conivaptan therapy initiated at 3 hours following ischemia reduced edema in the ipsilateral hemisphere, which corresponded with improvements in neurological deficits. Stroke-triggered BBB disruption was also reduced in mice when conivaptan treatments were initiated at 3 hours of reperfusion. However, 5 and 20-hour delays of conivaptan administration failed to reduce edema or protect BBB. CONCLUSION: Timing of conivaptan administration is important for successful reduction of brain edema and BBB disruption. Our experimental data open new possibilities to repurpose conivaptan, and make an important "bench-to-bedside translation" of the results into clinical practice.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/therapeutic use , Benzazepines/therapeutic use , Blood-Brain Barrier/drug effects , Brain Edema/drug therapy , Brain Edema/etiology , Infarction, Middle Cerebral Artery/complications , Animals , Blood-Brain Barrier/pathology , Brain Edema/pathology , Infarction, Middle Cerebral Artery/pathology , Male , Mice , Mice, Inbred C57BL
3.
J Vis Exp ; (115)2016 09 01.
Article in English | MEDLINE | ID: mdl-27684044

ABSTRACT

Stroke is one of the major causes of morbidity and mortality in the world. Stroke is complicated by brain edema and other pathophysiological events. Among the most important players in the development and evolution of stroke-evoked brain edema is the hormone arginine-vasopressin and its receptors, V1a and V2. Recently, the V1a and V2 receptor blocker conivaptan has been attracting attention as a potential drug to reduce brain edema after stroke. However, animal models which involve conivaptan applications in stroke research need to be modified based on feasible routes of administration. Here the outcomes of 48 hr continuous intravenous (IV) are compared with intraperitoneal (IP) conivaptan treatments after experimental stroke in mice. We developed a protocol in which middle cerebral artery occlusion was combined with catheter installation into the jugular vein for IV treatment of conivaptan (0.2 mg) or vehicle. Different cohorts of animals were treated with 0.2 mg bolus of conivaptan or vehicle IP daily. Experimental stroke-evoked brain edema was evaluated in mice after continuous IV and IP treatments. Comparison of the results revealed that the continuous IV administration of conivaptan alleviates post-ischemic brain edema in mice, unlike the IP administration of conivaptan. We conclude that our model can be used for future studies of conivaptan applications in the context of stroke and brain edema.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/administration & dosage , Benzazepines/administration & dosage , Brain Edema/drug therapy , Stroke/drug therapy , Animals , Brain Edema/metabolism , Brain Edema/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery , Infusions, Intravenous , Male , Mice , Mice, Inbred C57BL , Random Allocation , Receptors, Vasopressin/metabolism , Stroke/metabolism , Stroke/pathology
5.
PLoS One ; 10(8): e0136121, 2015.
Article in English | MEDLINE | ID: mdl-26275173

ABSTRACT

BACKGROUND: Stroke is a major cause of morbidity and mortality. Stroke is complicated by brain edema and blood-brain barrier (BBB) disruption, and is often accompanied by increased release of arginine-vasopressin (AVP). AVP acts through V1a and V2 receptors to trigger hyponatremia, vasospasm, and platelet aggregation which can exacerbate brain edema. The AVP receptor blockers conivaptan (V1a and V2) and tolvaptan (V2) are used to correct hyponatremia, but their effect on post-ischemic brain edema and BBB disruption remains to be elucidated. Therefore, we conducted this study to investigate if these drugs can prevent brain edema and BBB disruption in mice after stroke. METHODS: Experimental mice underwent the filament model of middle cerebral artery occlusion (MCAO) with reperfusion. Mice were treated with conivaptan, tolvaptan, or vehicle. Treatments were initiated immediately at reperfusion and administered IV (conivaptan) or orally (tolvaptan) for 48 hours. Physiological variables, neurological deficit scores (NDS), plasma and urine sodium and osmolality were recorded. Brain water content (BWC) and Evans Blue (EB) extravasation index were evaluated at the end point. RESULTS: Both conivaptan and tolvaptan produced aquaresis as indicated by changes in plasma and urine sodium levels. However plasma and urine osmolality was changed only by conivaptan. Unlike tolvaptan, conivaptan improved NDS and reduced BWC in the ipsilateral hemisphere: from 81.66 ± 0.43% (vehicle) to 78.28 ± 0.48% (conivaptan, 0.2 mg, p < 0.05 vs vehicle). Conivaptan also attenuated the EB extravasation from 1.22 ± 0.08 (vehicle) to 1.01 ± 0.02 (conivaptan, 0.2 mg, p < 0.05). CONCLUSION: Continuous IV infusion with conivaptan for 48 hours after experimental stroke reduces brain edema, and BBB disruption. Conivaptan but not tolvaptan may potentially be used in patients to prevent brain edema after stroke.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/pharmacology , Benzazepines/pharmacology , Blood-Brain Barrier/metabolism , Brain Edema/prevention & control , Receptors, Vasopressin/metabolism , Stroke/prevention & control , Animals , Blood-Brain Barrier/pathology , Brain Edema/metabolism , Brain Edema/pathology , Disease Models, Animal , Mice , Stroke/metabolism , Stroke/pathology , Water-Electrolyte Balance/drug effects
6.
PLoS One ; 9(7): e102194, 2014.
Article in English | MEDLINE | ID: mdl-25010766

ABSTRACT

BACKGROUND: Several studies demonstrate that estrogen treatment improves cerebral blood flow in ischemic brain regions of young ovariectomized (OVX) rats. Estrogen receptor-α (ER-α) may mediate estrogen's beneficial actions via its effects on the cerebral microvasculature. However, estrogen-derived benefit may be attenuated in aged, reproductively senescent (RS) rats. Our goal was to determine the effects of aging, estrogen deprivation and estrogen repletion with oral conjugated estrogens (CE) on postischemic cerebral microvascular protein expression of ER-α and ER-ß. METHODS: Fisher-344 (n = 37) female rats were randomly divided into the following groups: OVX, OVX CE-treated, RS untreated, and RS CE-treated. After 30 days pretreatment with CE (0.01 mg/kg) rats were subjected to 15 min. transient global cerebral ischemia. Non-ischemic naïve, OVX and RS rats were used as controls. Expression of ER-α and ER-ß in isolated cortical cerebral microvessels (20 to 100 µm in diameter) was assessed using Western blot and immunohistochemistry techniques. RESULTS: Age and reproductive status blunted nonischemic ER-α expression in microvessels of OVX rats (0.31 ± 0.05) and RS rats (0.33 ± 0.06) compared to naïve rats (0.45 ± 0.02). Postischemic microvascular expression of ER-α in OVX rats (0.01 ± 0.0) was increased by CE treatment (0.04 ± 0.01). Expression of ER-α in microvessels of RS rats (0.03 ± 0.02) was unaffected by CE treatment (0.01 ± 0.02). Western blot data are presented as a ratio of ER-α or ER-ß proteins to ß-actin and. Oral CE treatment had no effect on ER-ß expression in postischemic microvessels of OVX and RS rats. Statistical analysis was performed by One-Way ANOVA and a Newman-Keuls or Student's post-hoc test. CONCLUSION: Chronic treatment with CE increases ER-α but not ER-ß expression in cerebral microvessels of OVX rats. Aging appears to reduce the normal ability of estrogen to increase ER-α expression in postischemic cerebral microvessels.


Subject(s)
Aging/genetics , Cellular Senescence/genetics , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Aging/pathology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/genetics , Brain Ischemia/pathology , Cellular Senescence/drug effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Estrogens, Conjugated (USP)/administration & dosage , Female , Humans , Microvessels/drug effects , Microvessels/pathology , Rats
7.
J Vis Exp ; (57): e3203, 2011 Nov 29.
Article in English | MEDLINE | ID: mdl-22143194

ABSTRACT

Many of estrogen's effects on vascular reactivity are mediated through interaction with estrogen receptors (1, 2, 3). Although two sub-types exist (estrogen receptor -α and ß),estrogen receptor-α has been identified in both the smooth muscle and in endothelial cells of pial arterial segments using fluorescent staining combined with confocal laser scanning microscopy (4). Furthermore, ER-α is located in the nuclei and in the cytoplasm of rat basilar arteries (5). The receptors are abundant and fluoresce brightly, but clear visualization of discrete groups of receptors is difficult likely due to the numbers located in many cell layers of pial vessel segments. Additionally, many reports using immunohistochemical techniques paired with confocal microscopy poorly detail the requirements critical for reproduction of experiments (6). Our purpose for this article is to describe a simple technique to optimize the staining and visualization of ER-α using cross-sectional slices of pial arterioles obtain from female rat brains. We first perfuse rats with Evans blue dye to easily identify surface pial arteries which we isolate under a dissecting microscope. Use of a cryostat to slice 8 µm cross sections of the arteries allows us to obtain thin vessel sections so that different vessel planes are more clearly visualized. Cutting across the vessel rather than use of a small vessel segment has the advantage of easier viewing of the endothelial and smooth muscle layers. In addition, use of a digital immunofluorescent microscope with extended depth software produces clear images of ten to twelve different vessel planes and is less costly than use of a confocal laser scanning microscope.


Subject(s)
Estrogen Receptor alpha/analysis , Microscopy, Fluorescence/methods , Pia Mater/blood supply , Animals , Arterioles/chemistry , Estrogen Receptor alpha/chemistry , Female , Rats
8.
J Neurosci ; 30(4): 1413-6, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20107067

ABSTRACT

D-Serine, formed from L-serine by serine racemase (SR), is a physiologic coagonist at NMDA receptors. Using mice with targeted deletion of SR, we demonstrate a role for D-serine in NMDA receptor-mediated neurotoxicity and stroke. Brain cultures of SR-deleted mice display markedly diminished nitric oxide (NO) formation and neurotoxicity. In intact SR knock-out mice, NO formation and nitrosylation of NO targets are substantially reduced. Infarct volume following middle cerebral artery occlusion is dramatically diminished in several regions of the brains of SR mutant mice despite evidence of increased NMDA receptor number and sensitivity.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/genetics , Cytoprotection/genetics , Neurotoxins/metabolism , Racemases and Epimerases/genetics , Serine/metabolism , Animals , Brain/blood supply , Brain/enzymology , Brain/physiopathology , Brain Infarction/enzymology , Brain Infarction/genetics , Brain Infarction/therapy , Brain Ischemia/therapy , Cells, Cultured , Disease Models, Animal , Down-Regulation/genetics , Gene Deletion , Gene Expression Regulation, Enzymologic/genetics , Genetic Therapy/methods , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/therapy , Isomerism , Male , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/genetics , Nitro Compounds/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism
9.
Brain Res ; 1294: 183-92, 2009 Oct 19.
Article in English | MEDLINE | ID: mdl-19646426

ABSTRACT

Matrix metalloproteinase (MMP)-9 has been shown to contribute to blood-brain barrier (BBB) disruption, infarct formation, and hemorrhagic transformation after ischemic stroke. The cellular source of MMP-9 detectable in the ischemic brain remains controversial since extracellular molecules in the brain may be derived from blood. We here demonstrate that bone marrow-derived cells are the major source of MMP-9 in the ischemic brain. We made bone marrow chimeric mice with MMP-9 null and wild-type as donor and recipient. After 90 min of transient focal cerebral ischemia, MMP-9 null mice receiving wild-type bone marrow showed comparable outcomes to wild-type in brain MMP-9 levels and BBB disruption (endogenous albumin extravasation) at 1 h post-reperfusion and infarct size at 24 h post-reperfusion. In contrast, wild-type animals replaced with MMP-9 null bone marrow showed barely detectable levels of MMP-9 in the ischemic brain, with attenuations in BBB disruption and infarct size. MMP-9 null mice receiving wild-type bone marrow showed enhanced Evans blue extravasation as early as 1 h post-reperfusion compared to wild-type mice replaced with MMP-9 null bone marrow. These findings suggest that MMP-9 released from bone marrow-derived cells influences the progression of BBB disruption in the ischemic brain.


Subject(s)
Blood-Brain Barrier/physiopathology , Bone Marrow Cells/physiology , Brain Infarction/physiopathology , Brain Ischemia/physiopathology , Matrix Metalloproteinase 9/metabolism , Stroke/physiopathology , Animals , Animals, Genetically Modified , Blood-Brain Barrier/pathology , Bone Marrow Transplantation , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Infarction/pathology , Brain Ischemia/pathology , Capillary Permeability , Gelatinases/metabolism , Male , Matrix Metalloproteinase 9/genetics , Mice , Stroke/pathology , Time Factors , Transplantation Chimera
10.
Neurobiol Dis ; 35(2): 264-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19465127

ABSTRACT

Ischemic preconditioning (IPC) protects brain against ischemic injury by activating specific mechanisms. Our goal was to determine if the inducible heme oxygenase 1 (HO1) is required for such protection. IPC before transient or permanent ischemia reduced cortical infarct volumes by 57.4% and 33.9%, respectively at 48 h in wildtype adult mice. Interestingly, IPC failed to protect the HO1 gene deleted mice against permanent ischemic brain injury. IPC also resulted in a significant increase in HO1 protein levels in the brain and correlated with reduced neurological deficits after permanent and transient brain ischemia. Our study demonstrates that neuroprotective effects of IPC are at least partially mediated via HO1. Elucidating the physiological/cellular role by which HO1 is protective against brain ischemia may aid the development of selective drugs to treat stroke and its associated neurological disorders.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Heme Oxygenase-1/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Preconditioning , Animals , Blotting, Western , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain Ischemia/pathology , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/genetics , Infarction, Middle Cerebral Artery/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Severity of Illness Index
11.
Microcirculation ; 16(5): 403-13, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19347762

ABSTRACT

OBJECTIVE: The aims of this work were to determine if 1) ischemia alters pial artery responsiveness to the partially nitric oxide (NO)-dependent dilator, ADP, 2) the alteration depends on 17beta-estradial (E2), and 3) NO contributes to E2 protective effects. MATERIALS AND METHODS: Response to ADP and the non-NO-dependent dilator, PGE(2), were examined through closed cranial windows. Ovariectomized (OVX) and E2-replaced (E25, 0.025 mg; or E50, 0.05 mg) rats were subjected to 15-minute forebrain ischemia and one-hour reperfusion. Endothelial NO synthase (eNOS) expression was determined in pre- and postischemic isolated cortical microvessels. RESULTS: In OVX rats, ischemia depressed pial responses to ADP, but not to PGE(2). Both doses of E2 maintained responses to ADP and had no effect on the response to PGE(2). eNOS inhibition decreased the ADP response by 60% in the E25 rats and 50% in the E50 rats, but had no effect in the OVX rats. Compared to the OVX group, microvessel expression of eNOS was increased by E2, but postischemic eNOS was unchanged in both groups. CONCLUSIONS: The nearly complete loss of postischemic dilation to ADP suggests that normal non-NO-mediated dilatory mechanisms may be acutely impaired after ischemic injury. Estrogen's protective action on ADP dilation may involve both NO- and non-NO-mediated mechanisms.


Subject(s)
Adenosine Diphosphate/pharmacology , Brain Ischemia/metabolism , Cerebral Arteries/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Nitric Oxide/metabolism , Adenosine Diphosphate/metabolism , Animals , Dinoprostone/metabolism , Dinoprostone/pharmacology , Estradiol/metabolism , Estrogens/metabolism , Female , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Wistar , Vasodilation/drug effects
12.
Neurotox Res ; 15(2): 133-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19384576

ABSTRACT

Carbon monoxide (CO) is associated with central nervous system toxicity. However, evidence also indicates that CO can be protective, depending on its concentration. To determine if CO can be neuroprotective after ischemic brain injury, we subjected mice to transient middle cerebral artery occlusion and exposed them to different concentrations of CO. We found that in mice, low CO levels protected the brain from injury following 90-min transient focal ischemia and 48 h of reperfusion. When inhalation of 125 or 250 ppm CO began immediately at the onset of reperfusion, total hemispheric infarct volume was reduced by 32.1 +/- 8.9% and 62.2 +/- 14.4%, respectively; with an extended therapeutic window of 1-3 h after ischemia, CO inhalation also attenuated infarct volume significantly. Furthermore, early CO exposure limited brain edema formation by 3.2 +/- 0.8% (125 ppm) and 2.6 +/- 0.3% (250 ppm). Finally, CO inhalation significantly improved neurological deficit scores at 48 h of survival time after ischemia. Transient elevation of carboxyhemoglobin levels returned rapidly to baseline when CO exposure was stopped. These findings suggest a potential application of CO to treat brain ischemic stroke.


Subject(s)
Carbon Monoxide/therapeutic use , Infarction, Middle Cerebral Artery/prevention & control , Neuroprotective Agents/therapeutic use , Analysis of Variance , Animals , Brain Edema/etiology , Brain Edema/prevention & control , Carboxyhemoglobin/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/mortality , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/prevention & control , Time Factors
13.
Crit Care Med ; 36(9): 2634-40, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18679106

ABSTRACT

OBJECTIVE: Osmotherapy with hypertonic saline ameliorates cerebral edema associated with experimental ischemic stroke. We tested the hypothesis that hypertonic saline exerts its antiedema effect by promoting an efflux of water from brain via the perivascular aquaporin-4 pool. We used mice with targeted disruption of the gene encoding alpha-syntrophin (alpha-Syn(-/-)) that lack the perivascular aquaporin-4 pool but retain the endothelial pool of this protein. DESIGN: Prospective laboratory animal study. SETTING: Research laboratory in a university teaching hospital. MEASUREMENTS AND MAIN RESULTS: Halothane-anesthetized adult male wildtype C57B/6 and alpha-Syn(-/-) mice were subjected to 90 min of transient middle cerebral artery occlusion and treated with either a continuous intravenous infusion of 0.9% saline or 3% hypertonic saline (1.5 mL/kg/hr) for 48 hr. In the first series of experiments (n = 59), increased brain water content analyzed by wet-to-dry ratios in the ischemic hemisphere of wildtype mice was attenuated after hypertonic saline (79.9% +/- 0.5%; mean +/- SEM) but not after 0.9% saline (82.3% +/- 1.0%) treatment. In contrast in alpha-Syn(-/-) mice, hypertonic saline had no effect on the postischemic edema (hypertonic saline: 80.3% +/- 0.7%; 0.9% saline: 80.3% +/- 0.4%). In the second series of experiments (n = 32), treatment with hypertonic saline attenuated postischemic blood-brain barrier disruption at 48 hr in wildtype mice but not in alpha-Syn(-/-) mice; alpha-Syn(-/-) deletion alone had no effect on blood-brain barrier integrity. In the third series of experiments (n = 34), alpha-Syn(-/-) mice treated with either hypertonic saline or 0.9% saline had smaller infarct volume as compared with their wildtype counterparts. CONCLUSIONS: These data demonstrate that 1) osmotherapy with hypertonic saline exerts antiedema effects via the perivascular pool of aquaporin-4, 2) hypertonic saline attenuates blood-brain barrier disruption depending on the presence of perivascular aquaporin-4, and 3) deletion of the perivascular pool of aquaporin-4 alleviates tissue damage after stroke, in mice subjected to osmotherapy and in nontreated mice.


Subject(s)
Aquaporin 4/metabolism , Brain Edema/drug therapy , Brain Edema/physiopathology , Brain Ischemia/complications , Calcium-Binding Proteins/genetics , Membrane Proteins/genetics , Muscle Proteins/genetics , Saline Solution, Hypertonic/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Body Water/metabolism , Brain Edema/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmosis
14.
Proc Natl Acad Sci U S A ; 103(36): 13532-6, 2006 Sep 05.
Article in English | MEDLINE | ID: mdl-16938871

ABSTRACT

The aquaporin-4 (AQP4) pool in the perivascular astrocyte membranes has been shown to be critically involved in the formation and dissolution of brain edema. Cerebral edema is a major cause of morbidity and mortality in stroke. It is therefore essential to know whether the perivascular pool of AQP4 is up- or down-regulated after an ischemic insult, because such changes would determine the time course of edema formation. Here we demonstrate by quantitative immunogold cytochemistry that the ischemic striatum and neocortex show distinct patterns of AQP4 expression in the reperfusion phase after 90 min of middle cerebral artery occlusion. The striatal core displays a loss of perivascular AQP4 at 24 hr of reperfusion with no sign of subsequent recovery. The most affected part of the cortex also exhibits loss of perivascular AQP4. This loss is of magnitude similar to that of the striatal core, but it shows a partial recovery toward 72 hr of reperfusion. By freeze fracture we show that the loss of perivascular AQP4 is associated with the disappearance of the square lattices of particles that normally are distinct features of the perivascular astrocyte membrane. The cortical border zone differs from the central part of the ischemic lesion by showing no loss of perivascular AQP4 at 24 hr of reperfusion but rather a slight increase. These data indicate that the size of the AQP4 pool that controls the exchange of fluid between brain and blood during edema formation and dissolution is subject to large and region-specific changes in the reperfusion phase.


Subject(s)
Aquaporin 4/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neocortex/metabolism , Animals , Aquaporin 4/deficiency , Aquaporin 4/ultrastructure , Astrocytes/metabolism , Blood-Brain Barrier/physiology , Brain Edema/physiopathology , Brain Ischemia/etiology , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Freeze Fracturing , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neocortex/physiopathology , Neocortex/ultrastructure , Reperfusion , Time Factors
15.
J Cereb Blood Flow Metab ; 26(3): 414-20, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16049424

ABSTRACT

We have previously shown that treatment with selective kappa-opioid receptor agonist BRL 52537 hydrochloride [(+/-)-1-(3,4-dichlorophenyl) acetyl-2-(1-pyrrolidinyl) methylpiperidine] (1) has a long therapeutic window for providing ischemic neuroprotection and (2) attenuates ischemia-evoked nitric oxide (NO) production in vivo in rats. Neuronally derived NO has been shown to be deleterious in the male, but not in the female, rodent model of focal ischemic stroke. We sought to determine if the agent fails to protect ischemic brain when neuronal NO synthase (nNOS) is genetically deleted in male, but not female, mice. Halothane-anesthetized adult male and female nNOS null mutants (nNOS(-/-)) and the genetically matched wildtype (WT) strain were subjected to transient (2 h) middle cerebral artery occlusion by the intraluminal filament technique. Vehicle or BRL 52537 treatment with continuous intravenous infusion was instituted at the onset of reperfusion and continued for 22 h. In WT male mice, infarct volumes measured at 72 h of reperfusion were robustly decreased with BRL 52537 treatment. In contrast, BRL 52537 did not decrease infarct volume in male nNOS(-/-) mice. BRL 52537 had no effect in the WT or nNOS(-/-) female mice. These data support that BRL 52537's mechanism of neuroprotection in vivo is through attenuation of nNOS activity and ischemia-evoked NO production. Neuroprotective effects of BRL 52537 are lost in the male when nNOS is not present; therefore, BRL 52537 likely acts upstream from NO generation and its subsequent neurotoxicity.


Subject(s)
Neurons/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide/metabolism , Piperidines/pharmacology , Pyrrolidines/pharmacology , Receptors, Opioid, kappa/agonists , Animals , Brain Ischemia/metabolism , Disease Models, Animal , Female , Infusions, Intravenous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/chemistry , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase Type I/deficiency , Nitric Oxide Synthase Type I/drug effects , Nitric Oxide Synthase Type I/metabolism , Piperidines/administration & dosage , Pyrrolidines/administration & dosage , Reperfusion Injury , Sex Factors , Stroke/metabolism
16.
J Neurosurg ; 102(2): 348-54, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15739565

ABSTRACT

OBJECT: Aneurysmal subarachnoid hemorrhage affects approximately 10/100,000 people per year. Endovascular coil embolization is used increasingly to treat cerebral aneurysms and its safety and durability is rapidly developing. The long-term durability of coil embolization of cerebral aneurysms remains in question; patients treated using this modality require multiple follow-up angiography studies and occasional repeated treatments. METHODS: Optical coherence tomography (OCT) is an emerging imaging modality that uses backscattered light to produce high-resolution tomography of optically accessible biological tissues such as the eye, luminal surface of blood vessels, and gastrointestinal tract. Vascular OCT probes in the form of imaging microwires are presently available--although not Food and Drug Administration-approved--and may be adapted for use in the cerebral circulation. In this study the authors describe the initial use of OCT to make visible the neck of aneurysms created in a canine model and treated with coil embolization. Optical coherence tomography images demonstrate changes that correlate with the histological findings of healing at the aneurysm neck and thus may be capable of demonstrating human cerebral aneurysm healing. CONCLUSIONS: Optical coherence tomography may obviate the need for subsequent follow-up angiography studies as well as aid in the understanding of endovascular tissue healing. Data in this study demonstrate that further investigation of in vivo imaging with such probes is warranted.


Subject(s)
Embolization, Therapeutic , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Intracranial Aneurysm/pathology , Tomography, Optical Coherence , Wound Healing/physiology , Animals , Carotid Arteries/pathology , Carotid Artery Diseases/pathology , Carotid Artery Diseases/therapy , Cerebral Angiography , Cerebral Arteries/pathology , Disease Models, Animal , Dogs , Intracranial Aneurysm/therapy , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...