Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Brain Res ; 1522: 67-75, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23743262

ABSTRACT

Although the mechanisms underlying mild traumatic brain injury (mTBI) are becoming well understood, treatment options are still limited. In the present study, mTBI was induced by a weight drop model to produce a closed head injury to mice and the effect of inhaled nitric oxide (INO) was evaluated by a short term memory task (object recognition task) and immunohistochemical staining of glial fibrillary acidic protein (GFAP) and CD45 for the detection of reactive astrocytes and microglia. Results showed that mTBI model did not produce brain edema, skull fracture or sensorimotor coordination dysfunctions. Mice did however exhibit a significant deficit in short term memory (STM) and strong inflammatory reaction in the ipsilateral cortex and hippocampus compared to sham-injured controls 24h after mTBI. Additional groups of untreated mice tested 3 and 7 days later, demonstrated that recognition memory had recovered to normal levels by Day 3. Mice treated with 10ppm INO for 4 or 8h, beginning immediately after TBI demonstrated significantly improved STM at 24h when compared with room air controls (p<0.05). Whereas mice treated with 10ppm INO for 24h showed no improvement in STM. Mice treated with INO 10ppm for 8h exhibited significantly reduced microglia and astrocyte activation compared with room air controls. These data demonstrate that mTBI produces a disruption of STM which is evident 24h after injury and persists for 2-3 days. Treatment with low concentration or short durations of INO prevents this memory loss and also attenuates the inflammatory response. These findings may have relevance for the treatment of patients diagnosed with concussion.


Subject(s)
Brain Concussion/pathology , Free Radical Scavengers/administration & dosage , Memory, Short-Term/drug effects , Nitric Oxide/administration & dosage , Administration, Inhalation , Animals , Behavior, Animal/drug effects , Brain Concussion/metabolism , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Inflammation/pathology , Leukocyte Common Antigens/metabolism , Male , Mice
2.
Brain Res ; 1507: 134-45, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23473843

ABSTRACT

Inhaled nitric oxide (iNO) has been shown to reduce ischemia/reperfusion (I/R) injury in several different organ systems including the brain. We investigated whether iNO was neuroprotective in a mouse model of transient focal ischemia. Male Swiss Webster mice underwent middle cerebral artery occlusion for 1 h followed by reperfusion for 47 h. Mice were divided into 5 concentration groups and administered nitric oxide (NO) at either 10, 20, 40, 60 or 80 ppm. Each of the 5 concentration groups was subdivided into 4 duration groups which were treated with iNO for 5, 8, 16 or 24 h beginning immediately after artery occlusion. Results showed both concentration and duration determined efficacy. At 10 ppm only the 24hr duration group exhibited reduced infarct volume while at 20, 40 and 60 ppm only 8 and 16 h of exposure led to smaller infarctions. At these concentrations the dose response curves were strongly U shaped indicating a loss of benefit at long durations. At 80 ppm, reduction in infarct volume was not observed at any duration. Additional experiments showed that 60 ppm iNO could be transported from lung to brain and that iNO administered for 8h improved recovery from subarachnoid hemorrhage and reduced the inflammatory response accompanying ischemic stroke. Enhanced blood flow during reperfusion may be an important mediator of these effects.


Subject(s)
Brain/drug effects , Neuroprotective Agents/administration & dosage , Nitric Oxide/administration & dosage , Stroke/drug therapy , Administration, Inhalation , Animals , Brain/blood supply , Disease Models, Animal , Infarction, Middle Cerebral Artery/drug therapy , Male , Mice , Neuroprotective Agents/therapeutic use , Nitric Oxide/therapeutic use , Reperfusion Injury/drug therapy
3.
Methods Mol Biol ; 849: 529-40, 2012.
Article in English | MEDLINE | ID: mdl-22528113

ABSTRACT

In the last couple of decades, substantial progress has been made in the development of transgenic mouse models developing amyloid-ß deposits and/or neurofibrillary tangles. These mouse models of Alzheimer's disease and related disorders provide an excellent tool for investigating etiology, pathogenic mechanisms, and potential treatments. An essential component of their characterization is a detailed behavioral assessment, which clarifies the functional consequences of these pathologies. We have selected and refined a series of cognitive and sensorimotor tasks that are ideal for studying these models and the efficacy of various treatments.


Subject(s)
Alzheimer Disease/physiopathology , Cognition , Disease Models, Animal , Motor Activity/physiology , Animals , Maze Learning/physiology , Mice , Rotarod Performance Test
4.
Anesth Analg ; 114(5): 1034-41, 2012 May.
Article in English | MEDLINE | ID: mdl-22366846

ABSTRACT

BACKGROUND: Acute hypotension may be implicated in cognitive dysfunction. L-type calcium channel blockers in the setting of hypoxia are protective of learning and memory. We tested the hypothesis that hypotension induced by nimodipine (NIMO) and nicardipine (NICA) would be protective of long- and short-term memory compared to hypotension induced by nitroglycerin (NTG). METHODS: Forty Swiss-Webster mice (30 to 35 g, 6 to 8 weeks) were randomized into 4 groups for i.p. injection immediately after passive avoidance (PA) learning on day 0: (1) NTG (30 mg/kg); (2) NICA (40 mg/kg); (3) NIMO (40 mg/kg); and (4) saline. PA training latencies (seconds) were recorded for entry from a suspended platform into a Plexiglas tube where a shock (0.3 mA; 2-second duration) was automatically delivered. On day 2 latencies were recorded during a testing trial during which no shock was delivered. Latencies >900 seconds were assigned this value. Lower testing latency is indicative of an impairment of long-term associative memory. Forty-nine additional mice were randomized into similar groups for object recognition testing (ORT) and given i.p. injections on day 0. ORT measures short-term memory by exploiting the tendency of mice to prefer novel objects where a familiar object is present. On day 5 during training, 2 identical objects were placed in a circular arena and mice explored both for 15 minutes. A testing trial was conducted 1 hour later for 3 minutes after a novel object replaced a familiar one. Mice with intact memory spend about 65% of the time exploring the novel object. Mice with impaired memory devote equal time to each object. Recognition index (RI) is defined as the ratio of time spent exploring the novel object to time spent exploring both objects was the measure of memory. Mean arterial blood pressure (MAP), cerebral bloodflow, and body and brain oxygenation (PO(2)) studies were done in separate groups of mice to determine the dosages for matched degrees of hypotension and the physiological profile of each treatment. RESULTS: The median PA latencies for the different conditions were as follows: NTG (219.5 ± 93.5 second semi-interquartile range [SIQR]), NICA (372.5 ± 75.5 second SIQR), NIMO (540 ± 200 second SIQR) and saline (804 ± 257.5 second SIQR). Rank methods were used to analyze the PA latencies for significant differences. NTG latency was significantly shorter than NIMO latency (P = 0.012) and saline latency (P = 0.006), but not NICA latency (P = 0.126). ORT RI values showed a similar pattern. We found that NTG RI (47.2 ± 5.9% SEM) was different from NIMO RI (60.2 ± 4.6% SEM, P = 0.031) and different from saline RI (66.9 + 3.7% SEM, P = 0.006). Physiological experiments showed that MAP decreased to 45 to 50 mm Hg in all animals who became minimally responsive to external stimuli within 10 to 15 minutes of injection. Intergroup differences for MAP, body and brain oxygenation, and cerebral bloodflow were not statistically significant. CONCLUSION: Acute hypotension induced by NIMO was protective of 2 categories of memory formation relevant to the clinical posttreatment period. Both immediate long-term associative memory consolidation as measured by the PA learning paradigm and delayed short-term working memory function as measured by the ORT paradigm were significantly improved compared to matched levels of hypotension induced by NTG. These results indicate the utility of further investigation of l-type calcium channel blockers as a potential means of preserving cognition in the setting of hypotensive and low flow states.


Subject(s)
Hypotension/chemically induced , Memory, Long-Term/drug effects , Memory, Short-Term/drug effects , Nimodipine/pharmacology , Nitroglycerin/pharmacology , Vasodilator Agents/pharmacology , Animals , Avoidance Learning/drug effects , Blood Pressure/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Cerebrovascular Circulation , Data Interpretation, Statistical , Hypotension/psychology , Mice , Nicardipine/pharmacology , Oxygen Consumption/drug effects , Recognition, Psychology/drug effects
5.
Brain Res Rev ; 67(1-2): 193-208, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21315760

ABSTRACT

Although the central noradrenergic system has been shown to be involved in a number of behavioral and neurophysiological processes, the relation of these to its role in depressive illness has been difficult to define. The present review discusses the hypothesis that one of its chief functions that may be related to affective illness is the inhibition of behavioral activation, a prominent symptom of the disorder. This hypothesis is found to be consistent with most previous neuropsychopharmacological and immunohistochemical experiments on active behavior in rodents in a variety of experimental conditions using manipulation of neurotransmission at both locus coeruleus and forebrain adrenergic receptors. The findings support a mechanism in which high rates of noradrenergic neural activity suppress the neural activity of principal neurons in forebrain regions mediating active behavior. The suppression may be mediated through postsynaptic galaninergic and adrenergic receptors, and via the release of corticotrophin-releasing hormone. The hypothesis is consistent with clinical evidence for central noradrenergic system hyperactivity in depressives and with the view that this hyperactivity is a contributing etiological factor in the disorder. A similar mechanism may underlie the ability of the noradrenergic system to suppress seizure activity suggesting that inhibition of the spread of neural activation may be a unifying function.


Subject(s)
Adrenergic Neurons/physiology , Depressive Disorder/pathology , Inhibition, Psychological , Locus Coeruleus/pathology , Adrenergic Neurons/pathology , Animals , Depressive Disorder/psychology , Humans , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Neural Inhibition/physiology , Neural Pathways/cytology , Neural Pathways/pathology
6.
Int J Neuropsychopharmacol ; 14(3): 319-31, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20459885

ABSTRACT

The present study examined the ability of 6-fluoronorepinephrine (6FNE), a full selective α-adrenoceptor agonist, to produce antidepressant-like effects in mice. The drug, administered in the 4th ventricle, produced marked anti-immobility effects at mid-dose range in the acute forced swim, tail suspension and repeated open-space forced swim tests with minimal effect on open-field motor activity and also reversed anhedonia following lipopolysaccharide administration. Its antidepressant effects were equal to or greater than that of an established systemic antidepressant, desmethylimipramine, given subacutely. Experiments with α-adrenoceptor antagonists indicated that the drug acts primarily via the α2-receptor in contrast to endogenous catecholamines which appear to control depressive behaviour primarily via the α1-receptor. Antidepressant activity declined at higher doses signifying a possible pro-depressant effect of one of the α-adrenoceptor subtypes. Compared to the selective α2-agonist, dexmedetomidine, 6FNE showed equivalent antidepressant action in the tail suspension test but appeared to have a greater efficacy or speed of action in the repeated open-space forced swim test which produces a more sustained depression. Studies of regional brain Fos expression induced during the antidepressant tests showed that 6FNE tended to inhibit neural activity in two stress-responsive regions (locus coeruleus and paraventricular hypothalamus) but to enhance activity in two areas involved in motivated behaviour (nucleus accumbens shell and lateral septal nucleus) producing a neural pattern consistent with antidepressant action. It is concluded that 6FNE elicits a rapid and effective antidepressant and anti-stress response that may compare favourably with available antidepressants.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Antidepressive Agents/pharmacology , Depression/drug therapy , Desipramine/pharmacology , Norepinephrine/analogs & derivatives , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Animals , Antidepressive Agents/administration & dosage , Behavior, Animal/drug effects , Brain/drug effects , Hindlimb Suspension , Injections, Intraventricular , Male , Mice , Motor Activity/drug effects , Norepinephrine/administration & dosage , Norepinephrine/pharmacology , Pleasure/drug effects , Random Allocation , Swimming
7.
J Neurosci ; 30(49): 16559-66, 2010 Dec 08.
Article in English | MEDLINE | ID: mdl-21147995

ABSTRACT

Harnessing the immune system to clear protein aggregates is emerging as a promising approach to treat various neurodegenerative diseases. In Alzheimer's disease (AD), several clinical trials are ongoing using active and passive immunotherapy targeting the amyloid-ß (Aß) peptide. Limited emphasis has been put into clearing tau/tangle pathology, another major hallmark of the disease. Recent findings from the first Aß vaccination trial suggest that this approach has limited effect on tau pathology and that Aß plaque clearance may not halt or slow the progression of dementia in individuals with mild-to-moderate AD. To assess within a reasonable timeframe whether targeting tau pathology with immunotherapy could prevent cognitive decline, we developed a new model with accelerated tangle development. It was generated by crossing available strains that express all six human tau isoforms and the M146L presenilin mutation. Here, we show that this unique approach completely prevents severe cognitive impairment in three different tests. This remarkable effect correlated well with extensive clearance of abnormal tau within the brain. Overall, our findings indicate that immunotherapy targeting pathological tau is very feasible for tauopathies, and should be assessed in clinical trials in the near future.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/immunology , Cognition Disorders , Immunotherapy/methods , Mutation/immunology , tau Proteins/immunology , Alzheimer Disease/blood , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Antibodies/blood , Antibodies/therapeutic use , Behavior, Animal/physiology , Brain/metabolism , Brain/pathology , Cognition Disorders/etiology , Cognition Disorders/immunology , Cognition Disorders/therapy , DNA-Binding Proteins , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Female , Gliosis/etiology , Humans , Male , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/immunology , Mice , Mice, Transgenic , Motor Activity/genetics , Motor Activity/immunology , Mutation/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Polycomb-Group Proteins , Presenilin-1/genetics , Psychomotor Performance/drug effects , Recognition, Psychology/physiology , Rotarod Performance Test , Statistics, Nonparametric , Transcription Factors/metabolism , tau Proteins/genetics
8.
Anesth Analg ; 109(6): 1943-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19923525

ABSTRACT

BACKGROUND: Hypotension and a resultant decrease in cerebral blood flow have been implicated in the development of cognitive dysfunction. We tested the hypothesis that nimodipine (NIMO) administered at the onset of nitroglycerin (NTG)-induced hypotension would preserve long-term associative memory. METHODS: The passive avoidance (PA) paradigm was used to assess memory retention. For PA training, latencies (seconds) were recorded for entry from a suspended platform into a Plexiglas tube where a shock was automatically delivered. Latencies were recorded 48 h later for a testing trial. Ninety-six Swiss-Webster mice (30-35 g, 6-8 wk), were randomized into 6 groups 1) saline (control), 2) NTG immediately after learning, 3) NTG 3 h after learning, 4) NTG and NIMO, 5) vehicle, and 6) NIMO alone. The extent of hypotension and changes in brain tissue oxygenation (PbtO(2)) and in cerebral blood flow were studied in a separate group of animals. RESULTS: All groups exhibited similar training latencies (17.0 +/- 4.6 s). Mice subjected to hypotensive episodes showed a significant decrease in latency time (178 +/- 156 s) compared with those injected with saline, NTG + NIMO, or delayed NTG (580 +/- 81 s, 557 +/- 67 s, and 493 +/- 146 s, respectively). A Kruskal-Wallis 1-way analysis of variance indicated a significant difference among the 4 treatment groups (H = 15.34; P < 0.001). In a separate group of mice not subjected to behavioral studies, the same dose of NTG (n = 3) and NTG + NIMO (n = 3) caused mean arterial blood pressure to decrease from 85.9 +/- 3.8 mm Hg sem to 31.6 +/- 0.8 mm Hg sem and from 86.2 +/- 3.7 mm Hg sem to 32.6 +/- 0.2 mm Hg sem, respectively. Mean arterial blood pressure in mice treated with NIMO alone decreased from 88.1 +/- 3.8 mm Hg to 80.0 +/- 2.9 mm Hg. The intergroup difference was statistically significant (P < 0.05). PbtO(2) decreased from 51.7 +/- 4.5 mm Hg sem to 33.8 +/- 5.2 mm Hg sem in the NTG group and from 38.6 +/- 6.1 mm Hg sem to 25.4 +/- 2.0 mm Hg sem in the NTG + NIMO groups, respectively. There were no significant differences among groups. CONCLUSION: In a PA retention paradigm, the injection of NTG immediately after learning produced a significant impairment of long-term associative memory in mice, whereas delayed induced hypotension had no effect. NIMO attenuated the disruption in consolidation of long-term memory caused by NTG but did not improve latency in the absence of hypotension. The observed effect of NIMO may have been attributable to the preservation of calcium homeostasis during hypotension, because there were no differences in the PbtO(2) indices among groups.


Subject(s)
Behavior, Animal/drug effects , Calcium Channel Blockers/pharmacology , Cerebrovascular Circulation/drug effects , Cognition/drug effects , Hypotension/drug therapy , Memory Disorders/prevention & control , Memory/drug effects , Nimodipine/pharmacology , Animals , Avoidance Learning/drug effects , Blood Pressure/drug effects , Disease Models, Animal , Heart Rate/drug effects , Hypotension/chemically induced , Hypotension/physiopathology , Hypotension/psychology , Male , Memory Disorders/etiology , Memory Disorders/physiopathology , Memory Disorders/psychology , Mice , Nitroglycerin , Oxygen/blood , Reaction Time/drug effects , Time Factors
9.
J Alzheimers Dis ; 18(4): 961-72, 2009.
Article in English | MEDLINE | ID: mdl-19749432

ABSTRACT

Immunotherapy holds great promise for Alzheimer's disease (AD) and other conformational disorders but certain adverse reactions need to be overcome. Prior to the side effects in the first Elan/Wyeth AD vaccine trial, we proposed using amyloid-beta (Abeta) derivatives as a safer approach. The route of administration may also affect vaccine safety. To assess the feasibility of oral immunization that promotes mucosal immunity, Tg2576 AD model mice were treated prophylactically three times over 6 weeks starting at 3-5 months of age with a Salmonella vaccine expressing K6Abeta(1-30). At 22-24 months of age, cortical Abeta plaque burden and total Abeta(40/42) levels were reduced by 48-75% in the immunized mice compared to controls, which received unmodified Salmonella. Plaque clearance was not associated with increased microglial activation, which may be explained by the long treatment period. Furthermore, cerebral microhemorrhages were not increased in the treated mice in contrast to several passive Abeta antibody studies. These results further support our findings with this immunogen delivered subcutaneously and demonstrate its efficacy when given orally, which may provide added benefits for human use.


Subject(s)
Alzheimer Disease/prevention & control , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/immunology , Administration, Oral , Alzheimer Vaccines/administration & dosage , Animals , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/prevention & control , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulins/blood , Immunohistochemistry , Male , Mice , Mice, Transgenic , Peptide Fragments , Salmonella/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
10.
Brain Res ; 1291: 21-31, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19632210

ABSTRACT

alpha(1)-Adrenoceptors are concentrated in the locus coeruleus (LC) where they appear to regulate various active behaviors but have been difficult to stimulate effectively. The present study examined the behavioral, pharmacological and neural effects of possible stimulation of these receptors with 6-fluoronorepinephrine (6FNE), the only known selective alpha-agonist that has full efficacy at all brain alpha-receptors. Infusion of this compound in the mouse LC was found to produce extreme activation of diverse motivated behaviors of exploration, wheel-running and operant approach responding in different environments consistent with a global behavioral function of the dorsal noradrenergic system. Infusion of selective antagonists of alpha(1)- (terazosin) or alpha(2)- (atipamezole) receptors or of either the partial alpha(1)-agonist, phenylephrine, or full alpha(2)-agonist, dexmedetomidine, indicated that the behavioral effects of 6FNE were due largely due to activation of LC alpha(1)-receptors consistent with the known greater density of alpha(1)- than alpha(2)-adrenoreceptors in the mouse nucleus. Immunohistochemistry of fos in tyrosine hydroxylase-positive LC neurons following IV ventricular infusions indicated that 6FNE markedly depressed whereas terazosin strongly enhanced the apparent functional activity of the nucleus. The changes in fos expression following 6FNE and terazosin were significantly greater than those following dexmedetomidine and atipamezole. It is hypothesized that the alpha(1)-receptors of the mouse LC are strongly activated by 6FNE and serve to potently inhibit its tonic or stress-induced activity which in turn disinhibits prepotent motivated behaviors.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Conditioning, Operant/drug effects , Exploratory Behavior/drug effects , Locus Coeruleus/physiology , Motor Activity/drug effects , Receptors, Adrenergic, alpha-1/physiology , Analysis of Variance , Animals , Catheters, Indwelling , Fluorescent Antibody Technique , Locus Coeruleus/drug effects , Male , Mice , Neurons/metabolism , Oxidopamine/toxicity , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Adrenergic, alpha-2/physiology , Tyrosine 3-Monooxygenase/metabolism
11.
J Neurosurg Anesthesiol ; 21(2): 140-4, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19295393

ABSTRACT

BACKGROUND: Cognitive changes associated with moderate hypoxia may be related to the elevation of cytosolic calcium (Ca) levels which may, in turn, affect neurotransmitter synthesis and metabolism. We tested whether treatment with nimodipine (NIMO), an L-type Ca channel blocker, would preserve working memory after hypoxic hypoxia. METHODS: We randomized 157 Swiss-Webster, 30 to 35 g mice (6 to 8 wk) to 6 groups, which were exposed to the following gas mixtures for 1 hour: (1) O2 21%; (2) O2 21% followed by 0.1 mg/kg of subcutaneous NIMO; (3) O2 21% followed by vehicle (60% polyethylene glycol/40% methanol); (4) O2 10%; (5) O2 10% then NIMO; (6) O2 10% then vehicle. The Object Recognition Test (ORT) was given once either on Day 1 or Day 7 to assess changes in short-term memory. ORT exploits the tendency of mice to prefer novel over familiar objects. Two identical objects were placed in an arena for 15 minutes of training. During the testing 1 hour later, one of the objects was replaced by a new object. Recognition Index (RI) was used to compare performance. It is defined as the time spent exploring the novel object divided by the time spent exploring both objects, the novel plus the familiar, and this ratio is converted to a percentage. RI was analyzed with analysis of variance. Tukey Honestly Significant Difference tests were used for post hoc comparisons when appropriate. P values <0.05 were considered significant. RESULTS: RI for the control group was 68.3% (SE+/-3.6%). RI was 53.7% (SE+/-3.8%) for the 10% O2 group on the first posttreatment day. O2 saturation (SpO2) for the hypoxic group was 71.7% (SE+/-0.5%). By Day 7, RI for the 10% O2 group increased to 64.2% (SE+/-4.7%), which was not significantly different from control. On Day 1, RI was 68.6% (SE+/-5.2%) for hypoxic rodents treated with NIMO. These results were statistically significant. Low RI indicates impaired working memory and high RI indicates intact working memory. These results suggest that NIMO prevented impairment of working memory after moderate hypoxia. CONCLUSIONS: NIMO reverses the disturbance of short-term working memory caused by moderate hypoxia in mice. The results may have implications for cognitive changes linked to Ca homeostasis in the postoperative period.


Subject(s)
Calcium Channel Blockers/therapeutic use , Cognition Disorders/etiology , Cognition Disorders/prevention & control , Hypoxia, Brain/complications , Nimodipine/therapeutic use , Animals , Attention/drug effects , Blood Gas Analysis , Calcium Channels, L-Type/drug effects , Cognition Disorders/psychology , Hypoxia, Brain/psychology , Male , Memory/drug effects , Mice , Motor Activity/drug effects , Oxygen Consumption/drug effects , Recognition, Psychology/drug effects
12.
Blood ; 113(24): 6051-60, 2009 Jun 11.
Article in English | MEDLINE | ID: mdl-19218546

ABSTRACT

Anti-platelet integrin GPIIIa49-66 antibody (Ab) induces complement-independent platelet oxidative fragmentation and death by generation of platelet peroxide following NADPH oxidase activation. A C-terminal 385-amino acid fragment of ADAMTS-18 (a disintegrin metalloproteinase with thrombospondin motifs produced in endothelial cells) induces oxidative platelet fragmentation in an identical kinetic fashion as anti-GPIIIa49-66 Ab. Endothelial cell ADAMTS-18 secretion is enhanced by thrombin and activated by thrombin cleavage to fragment platelets. Platelet aggregates produced ex vivo with ADP or collagen and fibrinogen are destroyed by the C-terminal ADAMTS-18 fragment. Anti-ADAMTS-18 Ab shortens the tail vein bleeding time. The C-terminal fragment protects against FeCI3-induced carotid artery thrombosis as well as cerebral infarction in a postischemic stroke model. Thus, a new mechanism is proposed for platelet thrombus clearance, via platelet oxidative fragmentation induced by thrombin cleavage of ADAMTS-18.


Subject(s)
ADAM Proteins/pharmacology , Blood Platelets/metabolism , Carotid Stenosis/prevention & control , Peptide Fragments/pharmacology , Platelet Aggregation/drug effects , Stroke/prevention & control , ADAM Proteins/metabolism , ADAMTS Proteins , Animals , Bleeding Time , Blood Platelets/pathology , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Cyclic AMP/pharmacology , Endothelium, Vascular/metabolism , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Peptide Library , Platelet Glycoprotein GPIIb-IIIa Complex/pharmacology , Polymerase Chain Reaction , Stroke/metabolism , Stroke/pathology , Thrombin/pharmacology , Umbilical Veins/cytology
13.
Pharmacol Biochem Behav ; 91(1): 190-5, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18692087

ABSTRACT

The present study evaluated the extension of a new rat model of depression, repeated open-space swimming, which overcomes drawbacks of existing models, to mice. Mice were swum for 15 min daily in a large tank of tepid water for 4 days and thereafter at 4 day intervals for a period of 3 weeks. Some of the animals were provided with an active coping (escape) response. Variables measured included time floating, distance swum, immobility on a subsequent tail-suspension test, sucrose preference and brain cell proliferation (Ki67 immunohistochemistry) as well as responses to 2 antidepressant drugs, desmethylimipramine and fluoxetine, and 2 non-antidepressant drugs, haloperidol and diazepam. The repeated swims were found to increase time floating and tail-suspension immobility and to decrease distance swum, sucrose preference and brain cell proliferation. Both chronic antidepressant drugs as well as the active coping response attenuated the increased time floating while neither of the non-antidepressant drugs had this effect. The distance swum measure was found to be more variable. Chronic fluoxetine also reversed the increased tail-suspension immobility, reduced sucrose preference and reduced brain cell proliferation caused by the model. It is concluded that repeated open-space swim represents a useful new model of depression in the mouse.


Subject(s)
Depression/psychology , Swimming/psychology , Adaptation, Psychological , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents, Second-Generation/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Antipsychotic Agents/pharmacology , Cell Proliferation/drug effects , Chronic Disease , Desipramine/pharmacology , Diazepam/pharmacology , Disease Models, Animal , Fluoxetine/pharmacology , Food Preferences/psychology , Haloperidol/pharmacology , Hindlimb Suspension/psychology , Hypnotics and Sedatives/pharmacology , Male , Mice , Motor Activity/physiology , Neurons/drug effects
14.
J Neurosci Res ; 86(12): 2784-91, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18615702

ABSTRACT

Memantine, an N-methyl-D-aspartate (NMDA) receptor antagonist, has been shown to improve learning and memory in several preclinical models of Alzheimer's disease (AD). Memantine has also been shown to reduce the levels of amyloid beta (A beta) peptides in human neuroblastoma cells as well as to inhibit A beta oligomer-induced synaptic loss. In this study, we assessed whether NMDA receptor inhibition by memantine in transgenic mice expressing human amyloid-beta precursor protein (APP) and presenilin 1 (PS1) is associated with cognitive benefit and amyloid burden reduction by using object recognition, micromagnetic resonance imaging (micro MRI), and histology. APP/PS1 Tg mice were treated either with memantine or with vehicle for a period of 4 months starting at 3 months of age. After treatment, the mice were subjected to an object recognition test and analyzed by ex vivo micro MRI, and histological examination of amyloid burden. micro MRI was performed following injection with gadolinium-DTPA-A beta(1-40). We found that memantine-treated Tg mice performed the same as wild-type control mice, whereas the performance of vehicle-treated Tg mice was significantly impaired (P = 0.0081, one-way ANOVA). Compared with vehicle-treated animals, memantine-treated Tg mice had a reduced plaque burden, as determined both histologically and by micro MRI. This reduction in amyloid burden correlates with an improvement in cognitive performance. Thus, our findings provide further evidence of the potential role of NMDA receptor antagonists in ameliorating AD-related pathology. In addition, our study shows, for the first time, the utility of micro MRI in conjunction with gadolinium-labeled A beta labeling agents to monitor the therapeutic response to amyloid-reducing agents.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Disease Models, Animal , Memantine/therapeutic use , Alzheimer Disease/pathology , Animals , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Magnetic Resonance Imaging/methods , Memantine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recognition, Psychology/drug effects , Recognition, Psychology/physiology
15.
Synapse ; 62(7): 516-23, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18435418

ABSTRACT

alpha(1)-Adrenoceptors of the locus coeruleus (LC) have been implicated in behavioral activation in novel surroundings, but the endogenous agonist that activates these receptors has not been established. In addition to the canonical activation of alpha(1)-receptors by norepinephrine (NE), there is evidence that dopamine (DA) may also activate certain brain alpha(1)-receptors. This study examined the contribution of DA to exploratory activity in a novel cage by determining the effect of infusion of various dopaminergic and adrenergic drugs into the mouse LC. It was found that the D2/D3 agonist, quinpirole, which selectively blocks the release of CNS DA, produced a dose-dependent and virtually complete abolition of exploration and all movement in the novel cage test. The quinpirole-induced inactivity was significantly attenuated by coinfusion of DA but not by the D1 agonist, SKF38390. Furthermore, the DA attenuation of quinpirole inactivity was blocked by coinfusion of the alpha(1)-adrenergic receptor antagonist, terazosin, but not by the D1 receptor antagonist, SCH23390. LC infusions of either quinpirole or terazosin also produced profound inactivity in DA-beta-hydroxylase knockout (Dbh -/-) mice that lack NE, indicating that their behavioral effects were not due to an alteration of the release or action of LC NE. Measurement of endogenous DA, NE, and 5HT and their metabolites in the LC during exposure to the novel cage indicated an increase in the turnover of DA and NE but not 5HT. These results indicate that DA is a candidate as an endogenous agonist for behaviorally activating LC alpha(1)-receptors and may play a role in the activation of this nucleus by novel surroundings.


Subject(s)
Arousal/physiology , Dopamine/metabolism , Exploratory Behavior/physiology , Locus Coeruleus/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Synaptic Transmission/physiology , Adrenergic alpha-1 Receptor Agonists , Animals , Arousal/drug effects , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Dopamine beta-Hydroxylase/genetics , Dose-Response Relationship, Drug , Drug Interactions/physiology , Exploratory Behavior/drug effects , Female , Locus Coeruleus/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Norepinephrine/biosynthesis , Serotonin/metabolism , Synaptic Transmission/drug effects
16.
Neurosci Biobehav Rev ; 32(3): 508-24, 2008.
Article in English | MEDLINE | ID: mdl-18023876

ABSTRACT

Functional neuroimaging studies of depressed patients have converged with functional brain mapping studies of depressed animals in showing that depression is accompanied by a hypoactivity of brain regions involved in positively motivated behavior together with a hyperactivity in regions involved in stress responses. Both sets of changes are reversed by diverse antidepressant treatments. It has been proposed that this neural pattern underlies the symptoms common to most forms of the depression, which are the loss of positively motivated behavior and increased stress. The paper discusses how this framework can organize diverse findings ranging from effects of monoamine neurotransmitters, cytokines, corticosteroids and neurotrophins on depression. The hypothesis leads to new insights concerning the relationship between the prolonged inactivity of the positive motivational network during a depressive episode and the loss of neurotrophic support, the potential antidepressant action of corticosteroid treatment, and to the key question of whether antidepressants act by inhibiting the activity of the stress network or by enhancing the activity of the positive motivational system.


Subject(s)
Brain Mapping , Depressive Disorder/physiopathology , Motivation , Neural Pathways , Stress, Psychological/physiopathology , Adrenal Cortex Hormones/therapeutic use , Animals , Antidepressive Agents/therapeutic use , Cytokines/therapeutic use , Depressive Disorder/complications , Depressive Disorder/drug therapy , Disease Models, Animal , Humans , Mice , Nerve Growth Factors/therapeutic use , Reward , Stress, Psychological/complications
17.
J Neurosci ; 27(34): 9115-29, 2007 Aug 22.
Article in English | MEDLINE | ID: mdl-17715348

ABSTRACT

Immunotherapies for various neurodegenerative diseases have recently emerged as a promising approach for clearing pathological protein conformers in these disorders. This type of treatment has not been assessed in models that develop neuronal tau aggregates as observed in frontotemporal dementia and Alzheimer's disease. Here, we present that active immunization with a phosphorylated tau epitope, in P301L tangle model mice, reduces aggregated tau in the brain and slows progression of the tangle-related behavioral phenotype. Females had more tau pathology than males but were also more receptive to the immunotherapy. The tau antibodies generated in these animals recognized pathological tau on brain sections. Performance on behavioral assays that require extensive motor coordination correlated with tau pathology in corresponding brain areas, and antibody levels against the immunogen correlated inversely with tau pathology. Interestingly, age-dependent autoantibodies that recognized recombinant tau protein but not the immunogen were detected in the P301L mice. To confirm that anti-tau antibodies could enter the brain and bind to pathological tau, FITC-tagged antibodies purified from a P301L mouse, with a high antibody titer against the immunogen, were injected into the carotid artery of P301L mice. These antibodies were subsequently detected within the brain and colocalized with PHF1 and MC1 antibodies that recognize pathological tau. Currently, no treatment is available for clearing tau aggregates. Our present findings may lead to a novel therapy targeting one of the major hallmarks of Alzheimer's disease and frontotemporal dementia.


Subject(s)
Antibodies/therapeutic use , Brain/pathology , Dementia/therapy , Immunotherapy/methods , tau Proteins/immunology , tau Proteins/metabolism , Age Factors , Analysis of Variance , Animals , Behavior, Animal , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Dementia/genetics , Dementia/pathology , Disease Models, Animal , Humans , Leucine/genetics , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Neuroblastoma , Proline/genetics , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Sex Factors , tau Proteins/genetics
18.
Anesth Analg ; 105(3): 739-43, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17717233

ABSTRACT

BACKGROUND: Cognitive changes associated with moderate hypoxia in rodents may result from the diminished functioning of central cholinergic neurotransmission. We designed this study to examine whether treatment with physostigmine (PHY), an acetylcholinesterase inhibitor, could improve the impairment of working memory after hypoxic hypoxia. METHODS: We randomized 90 Swiss Webster, 30-35 g mice (6-8 wks) to three hypoxia groups at fraction of inspired oxygen, FiO2 = 0.10 (1. no treatment; 2. PHY 0.1 mg/kg intraperitoneally administered immediately before; or 3. after hypoxia), or to two room air groups (given either no treatment or PHY after an insult). An object recognition test was used to assess short-term memory function. The object recognition test exploits the tendency of mice to prefer exploring novel objects in an environment when a familiar object is also present. During the 15 min training trial, two identical objects were placed in two defined sites of the box. During the test trial performed 1 h later, one of the objects was replaced by a new object with a different shape. The time spent exploring the two objects was automatically recorded by a video camera and associated software. The performance was analyzed with ANOVA, followed by post hoc comparisons using the Newman-Keuls test when appropriate. P values <0.05 were considered significant. RESULTS: Untreated mice subjected to hypoxia at Fio2 = 0.1 spent significantly less time exploring a novel object on testing day 1 than did untreated mice breathing room air. Performance of the mice subjected to hypoxia, who received physostigmine after, but not before, the insult did not differ from the control group. CONCLUSION: Moderate hypoxia impairs rodents' performance in a working memory task. It appears that changes are transient, because the cognitive functioning of the mice returned to the baseline level 7 days after treatment. Postinsult administration of PHY prevented deterioration of cognitive function. An increased level of acetylcholine in the central nervous system may be responsible for the improved performance of the hypoxia-treated mice.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Cholinesterase Inhibitors/pharmacology , Cognition Disorders/prevention & control , Cognition/drug effects , Hypoxia, Brain/drug therapy , Nootropic Agents/pharmacology , Physostigmine/pharmacology , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Animals , Brain/enzymology , Cholinesterase Inhibitors/therapeutic use , Cognition Disorders/enzymology , Cognition Disorders/etiology , Disease Models, Animal , Exploratory Behavior/drug effects , Hypoxia, Brain/enzymology , Hypoxia, Brain/psychology , Male , Memory/drug effects , Mice , Nootropic Agents/therapeutic use , Physostigmine/therapeutic use , Severity of Illness Index , Time Factors
19.
Neurol Res ; 29(5): 469-75, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17535591

ABSTRACT

OBJECTIVES: The purpose of this experiment was to determine if the low molecular weight heparin (LMWH) enoxaparin could extend the treatment window for thrombolysis in a mouse model of embolic stroke. METHODS: To establish the treatment window, mice were treated with tPA 2, 3 or 4 hours after clot insertion. Results showed that only the 2 hour treatment group exhibited infarct volumes significantly smaller than untreated controls. We attempted to widen this window by pre-treating mice with enoxaparin (10 mg/kg, s.c.; n=36) 1 hour before embolization. A control group (n=24) was given a saline injection. The enoxaparin-treated animals were subdivided and treated with tPA either 4 (n=12) or 6 hours (n=12) after clot insertion, while the third group (n=12) was given saline. The saline-pre-treated mice were dived into two groups: one group (n=12) received tPA and the other group (n=12) received saline 4 hours post-stroke. Embolization was confirmed by laser Doppler flowmetry and the effects of the resulting infarcts were evaluated by triphenyltetrazolium chloride staining and by behavioral testing. RESULTS: Results showed large infarcts and impaired sensorimotor coordination in the saline pre-treated animals confirming the narrow treatment window. Enoxaparin pre-treatment produced significantly smaller infarcts and improved motor behavior in groups treated with tPA both 4 and 6 hours after embolization. Neither the 4 nor the 6 hour tPA-treated groups showed evidence of intracerebral hemorrhage or external bleeding. CONCLUSION: These data indicate that the LMWH enoxaparin can significantly increase the therapeutic time window in a mouse model of embolic stroke.


Subject(s)
Enoxaparin/therapeutic use , Fibrinolytic Agents/therapeutic use , Intracranial Embolism/drug therapy , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , Animals , Behavior, Animal/drug effects , Brain Infarction/drug therapy , Brain Infarction/etiology , Disease Models, Animal , Drug Interactions , Intracranial Embolism/complications , Laser-Doppler Flowmetry/methods , Male , Mice , Motor Activity/drug effects , Motor Activity/physiology , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Reflex/drug effects , Reflex/physiology , Stroke/complications , Tetrazolium Salts , Time Factors
20.
Prog Neuropsychopharmacol Biol Psychiatry ; 31(6): 1196-207, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17513031

ABSTRACT

A previous study showed that two mouse models of behavioral depression, immune system activation and depletion of brain monoamines, are accompanied by marked reductions in stimulated neural activity in brain regions involved in motivated behavior. The present study tested whether this effect is common to other depression models by examining the effects of repeated forced swimming, chronic subordination stress or acute intraventricular galanin injection - three additional models - on baseline or stimulated c-fos expression in several brain regions known to be involved in motor or motivational processes (secondary motor, M2, anterior piriform cortex, APIR, posterior cingulate gyrus, CG, nucleus accumbens, NAC). Each of the depression models was found to reduce the fos response stimulated by exposure to a novel cage or a swim stress in all four of these brain areas but not to affect the response of a stress-sensitive region (paraventricular hypothalamus, PVH) that was included for control purposes. Baseline fos expression in these structures was either unaffected or affected in an opposite direction to the stimulated response. Pretreatment with either desmethylimipramine (DMI) or tranylcypromine (tranyl) attenuated these changes. It is concluded that the pattern of a reduced neural function of CNS motor/motivational regions with an increased function of stress areas is common to 5 models of behavioral depression in the mouse and is a potential experimental analog of the neural activity changes occurring in the clinical condition.


Subject(s)
Depression/metabolism , Depression/pathology , Gene Expression Regulation/physiology , Proto-Oncogene Proteins c-fos/metabolism , Analysis of Variance , Animals , Antidepressive Agents/administration & dosage , Behavior, Animal , Depression/etiology , Depression/prevention & control , Desipramine/administration & dosage , Disease Models, Animal , Dominance-Subordination , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Galanin , Gene Expression Regulation/drug effects , Male , Mice , Stress, Psychological/complications , Swimming/psychology , Time Factors , Tranylcypromine/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...