Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Epilepsia ; 58(9): 1626-1636, 2017 09.
Article in English | MEDLINE | ID: mdl-28714074

ABSTRACT

OBJECTIVE: This study aimed to identify noninvasive biomarkers of human epilepsy that can reliably detect and localize epileptic brain regions. Having noninvasive biomarkers would greatly enhance patient diagnosis, patient monitoring, and novel therapy development. At the present time, only surgically invasive, direct brain recordings are capable of detecting these regions with precision, which severely limits the pace and scope of both clinical management and research progress in epilepsy. METHODS: We compared high versus low or nonspiking regions in nine medically intractable epilepsy surgery patients by performing integrated metabolomic-genomic-histological analyses of electrically mapped human cortical regions using high-resolution magic angle spinning proton magnetic resonance spectroscopy, cDNA microarrays, and histological analysis. RESULTS: We found a highly consistent and predictive metabolite logistic regression model with reduced lactate and increased creatine plus phosphocreatine and choline, suggestive of a chronically altered metabolic state in epileptic brain regions. Linking gene expression, cellular, and histological differences to these key metabolites using a hierarchical clustering approach predicted altered metabolic vascular coupling in the affected regions. Consistently, these predictions were validated histologically, showing both neovascularization and newly discovered, millimeter-sized microlesions. SIGNIFICANCE: Using a systems biology approach on electrically mapped human cortex provides new evidence for spatially segregated, metabolic derangements in both neurovascular and synaptic architecture in human epileptic brain regions that could be a noninvasively detectable biomarker of epilepsy. These findings both highlight the immense power of a systems biology approach and identify a potentially important role that magnetic resonance spectroscopy can play in the research and clinical management of epilepsy.


Subject(s)
Epilepsy/metabolism , Metabolomics , Adolescent , Biomarkers , Brain/metabolism , Child , Child, Preschool , Choline/metabolism , Creatine/metabolism , Epilepsy/genetics , Female , Genetic Markers , Humans , Infant , Lactic Acid/metabolism , Magnetic Resonance Spectroscopy , Male , Oligonucleotide Array Sequence Analysis , Phosphocreatine/metabolism
2.
Pediatr Transplant ; 21(2)2017 Mar.
Article in English | MEDLINE | ID: mdl-27885788

ABSTRACT

A decrease in live donor pediatric kidney transplants has occurred in the United States. This study investigates barriers that may influence access to live donor kidney transplants in children. Retrospective chart review was conducted for 91 children (69% male, mean age 11.9 years) who underwent pretransplant workup from 2005 to 2015 at an urban pediatric hospital. Fifty-four percent were African American, 32% Caucasian, 8% Arabic, 3% Hispanic, and 3% Others. Government-sponsored insurance (Medicaid/Medicare) was utilized by 73%, and 54% had dual caregivers. Only nine of 68 kidney transplants were live donor transplants. Live donor transplants (11%) were significantly (P=.008) lower than deceased donor transplants (59%) in African Americans. Private insurance was reported by 56% of live donor recipients and 25% of deceased donor recipients. Among live donor recipients, 78% were from dual caregiver families. Caregiver, health-related, financial, and religious/cultural barriers to live donor transplants were reported, several of which may be amenable to positive intervention.


Subject(s)
Kidney Failure, Chronic/surgery , Kidney Transplantation/methods , Living Donors/statistics & numerical data , Pediatrics/methods , Renal Insufficiency/surgery , Adolescent , Caregivers , Child , Female , Health Services Accessibility , Humans , Insurance, Health , Kidney Failure, Chronic/ethnology , Male , Prevalence , Retrospective Studies , Tissue Donors , Tissue and Organ Procurement , United States
3.
J Neurochem ; 136(6): 1196-1203, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26725566

ABSTRACT

Pavlovian conditioned approach (PCA) is a behavioral procedure that can be used to assess individual differences in the addiction vulnerability of drug-naïve rats and identify addiction vulnerability factors. Using proton magnetic resonance spectroscopy (1 H-MRS) ex vivo, we simultaneously analyzed concentrations of multiple neurochemicals throughout the mesocorticolimbic system 2 weeks after PCA training in order to identify potential vulnerability factors to addiction in drug-naïve rats for future investigations. Levels of myo-inositol (Ins), a 1 H-MRS-detectable marker of glial activity/proliferation, were increased in the nucleus accumbens (NAc) and ventral hippocampus, but not dorsal hippocampus or medial prefrontal cortex, of sign-trackers compared to goal-trackers or intermediate responders. In addition, Ins levels positively correlated with PCA behavior in the NAc and ventral hippocampus. Because the sign-tracker phenotype is associated with increased drug-seeking behavior, these results observed in drug-naïve rats suggest that alterations in glial activity/proliferation within these regions may represent an addiction vulnerability factor. Sign-tracking rats preferentially approach reward cues during Pavlovian conditioning, while goal-trackers instead approach the location of impending reward. Sign-trackers are also more prone to cue-induced drug-seeking behavior. We used magnetic resonance spectroscopy to show that myo-inositol levels are higher in the ventral hippocampus and nucleus accumbens of sign-trackers relative to goal-trackers. Thus, elevated myo-inositol may be a vulnerability factor for addiction.

4.
Mol Cell Neurosci ; 59: 119-26, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24534010

ABSTRACT

Working memory, which is dependent on higher-order executive function in the prefrontal cortex, is often disrupted in patients exposed to blast overpressure. In this study, we evaluated working memory and medial prefrontal neurochemical status in a rat model of blast neurotrauma. Adult male Sprague-Dawley rats were anesthetized with 3% isoflurane and exposed to calibrated blast overpressure (17 psi, 117 kPa) while sham animals received only anesthesia. Early neurochemical effects in the prefrontal cortex included a significant decrease in betaine (trimethylglycine) and an increase in GABA at 24 h, and significant increases in glycerophosphorylcholine, phosphorylethanolamine, as well as glutamate/creatine and lactate/creatine ratios at 48 h. Seven days after blast, only myo-inositol levels were altered showing a 15% increase. Compared to controls, short-term memory in the novel object recognition task was significantly impaired in animals exposed to blast overpressure. Working memory in control animals was negatively correlated with myo-inositol levels (r=-.759, p<0.05), an association that was absent in blast exposed animals. Increased myo-inositol may represent tardive glial scarring in the prefrontal cortex, a notion supported by GFAP changes in this region after blast overexposure as well as clinical reports of increased myo-inositol in disorders of memory.


Subject(s)
Blast Injuries/physiopathology , Brain Injuries/physiopathology , Inositol/metabolism , Memory, Short-Term , Prefrontal Cortex/metabolism , Animals , Betaine/metabolism , Blast Injuries/metabolism , Brain Injuries/metabolism , Creatine/metabolism , Ethanolamines/metabolism , Glutamic Acid/metabolism , Glycerylphosphorylcholine/metabolism , Lactic Acid/metabolism , Male , Pattern Recognition, Physiological , Prefrontal Cortex/injuries , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/metabolism
5.
Int J Psychiatry Clin Pract ; 18(1): 2-10, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24313739

ABSTRACT

OBJECTIVES: Currently, there are no guidelines for when to use an antiepileptic drug (AED) in nonepileptic panic disorder (PD) patients. We conducted this review to ascertain what guidance available literature can provide as to when to consider AEDs for PD patients. METHODS: The primary data sources were PubMed and Google-Scholars. Search was limited to "English" and "Humans". Only papers addressing use of nonbenzodiazepine AEDs in PD were included. Data regarding study subjects, the AED utilized, and clinical responses were collected. EEG data were used to classify reports of patients with abnormal versus those with normal and/or no EEG work-up. RESULTS: Ten reports were identified for use of AEDs in PD patients with abnormal EEGs with a total of 20 patients (17 responders). None of the 10 reports were controlled studies. Eighteen reports were identified for use of AEDs in panic patients with either normal EEGs or unselected groups (no EEG work-up). Out of the 18 reports, three were controlled studies. Included in the 18 studies were 253 patients (137 responders). CONCLUSIONS: We preliminary concluded that EEG work-up could be useful in guiding the treatment in PD as an abnormal EEG may be indicative of a higher likelihood of a positive response to an AED.


Subject(s)
Anticonvulsants/therapeutic use , Panic Disorder/drug therapy , Practice Guidelines as Topic/standards , Databases, Bibliographic , Electroencephalography , Epilepsy, Temporal Lobe/physiopathology , Humans , Panic Disorder/physiopathology , Patient Selection , Publication Bias , Treatment Outcome
6.
NMR Biomed ; 25(12): 1331-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22549883

ABSTRACT

Blast-induced neurotrauma is a major concern because of the complex expression of neuropsychiatric disorders after exposure. Disruptions in neuronal function, proximal in time to blast exposure, may eventually contribute to the late emergence of clinical deficits. Using magic angle spinning ¹H MRS and a rodent model of blast-induced neurotrauma, we found acute (24-48 h) decreases in succinate, glutathione, glutamate, phosphorylethanolamine and γ-aminobutyric acid, no change in N-acetylaspartate and increased glycerophosphorylcholine, alterations consistent with mitochondrial distress, altered neurochemical transmission and increased membrane turnover. Increased levels of the apoptotic markers Bax and caspase-3 suggested active cell death, consistent with increased FluoroJade B staining in the hippocampus. Elevated levels of glial fibrillary acidic protein suggested ongoing inflammation without diffuse axonal injury measured by no change in ß-amyloid precursor protein. In conclusion, blast-induced neurotrauma induces a metabolic cascade associated with neuronal loss in the hippocampus in the acute period following exposure.


Subject(s)
Blast Injuries/metabolism , Blast Injuries/pathology , Brain Injuries/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Animals , Blotting, Western , Brain Injuries/pathology , Caspase 3/metabolism , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Magnetic Resonance Spectroscopy , Male , Rats , Rats, Sprague-Dawley , bcl-2-Associated X Protein/metabolism
7.
NMR Biomed ; 22(4): 419-25, 2009 May.
Article in English | MEDLINE | ID: mdl-18985626

ABSTRACT

Despite the potential for deleterious (even fatal) effects on cardiac physiology, 3,4-methylenedioxymethamphetamine (MDMA; ecstasy) abuse abounds driven mainly by its euphoric effects. Acute exposure to MDMA has profound cardiovascular effects on blood pressure and heart rate in humans and animals. To determine the effects of MDMA on cardiac metabolites in rats, MDMA (0, 5, or 10 mg/kg) was injected every 2 h for a total of four injections; animals were sacrificed 2 h after the last injection (8 h drug exposure), and their hearts removed and tissue samples from left ventricular wall dissected. High resolution magic angle spinning proton magnetic resonance spectroscopy ((1)H-MRS) at 11.7 T, a specialized version of MRS aptly suited for analysis of semi-solid materials such as intact tissue samples, was used to measure the cardiac metabolomic profile, including alanine, lactate, succinate, creatine, and carnitine, in heart tissue from rats treated with MDMA. MDMA effects on MR-visible choline, glutamate, glutamine, and taurine were also determined. Body temperature was measured following each MDMA administration and serotonin and norepinephrine (NE) levels were measured by high pressure liquid chromatography (HPLC) in heart tissue from treated animals. MDMA significantly and dose-dependently increased body temperature, a hallmark of amphetamines. Serotonin, but not NE, levels were significantly and dose-dependently decreased by MDMA in the heart wall. MDMA significantly altered the MR-visible profile with an increase in carnitine and no change in other key compounds involved in cardiomyocyte energy metabolomics. Finally, choline levels were significantly decreased by MDMA in heart. The results are consistent with the notion that MDMA has significant effects on cardiovascular serotonergic tone and disrupts the metabolic homeostasis of energy regulation in cardiac tissue, potentially increasing utilization of fatty acid metabolism. The contributions of serotonergic signaling on MDMA-induced changes in cardiac metabolism remain to be determined.


Subject(s)
Heart/drug effects , Metabolome/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Animals , Body Temperature/drug effects , Choline/metabolism , Magnetic Resonance Spectroscopy , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Norepinephrine/metabolism , Organ Specificity/drug effects , Protons , Rats , Rats, Sprague-Dawley , Serotonin/metabolism
8.
Neurobiol Dis ; 29(3): 505-14, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18187333

ABSTRACT

Loss-of-function mutations in the DJ-1 gene account for an autosomal recessive form of Parkinson's disease (PD). To investigate the physiological functions of DJ-1 in vivo, we generated DJ-1 knockout (DJ-1(-/-)) mice. Younger (<1 year) DJ-1(-/-) mice were hypoactive and had mild gait abnormalities. Older DJ-1(-/-), however, showed decreased body weight and grip strength and more severe gait irregularities compared to wild-type littermates. The basal level of extracellular dopamine, evoked dopamine release and dopamine receptor D2 sensitivity appeared normal in the striatum of DJ-1(-/-) mice, which was consistent with similar results between DJ-1(-/-) and controls in behavioral paradigms specific for the dopaminergic system. An examination of spinal cord, nerve and muscle tissues failed to identify any pathological changes that were consistent with the noted motor deficits. Taken together, our findings suggest that loss of DJ-1 leads to progressive behavioral changes without significant alterations in nigrostriatal dopaminergic and spinal motor systems.


Subject(s)
Behavior, Animal/physiology , Corpus Striatum/physiology , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Substantia Nigra/physiology , Animals , Disease Progression , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology
9.
Neuropsychopharmacology ; 33(7): 1545-58, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17700647

ABSTRACT

Apoptosis has been proposed as a contributing cellular mechanism to the structural alterations that have been observed in stress-related mood disorders. Antidepressants, on the other hand, are hypothesized to exert trophic and/or neuroprotective actions. The present study examined the regulation of the major antiapoptotic (Bcl-2, Bcl-xl) and proapoptotic (Bax) genes by repeated unpredictable stress (an animal model of depression) and antidepressant treatments (ADT). In adult rats, exposure to unpredictable stress reduced Bcl-2 mRNA levels in the central nucleus of the amygdala (CeA), cingulate (Cg), and frontal (Fr) cortices. Bcl-xl mRNA was significantly decreased in hippocampal subfields. In contrast, chronic administration of clinically effective antidepressants from four different classes, ie fluoxetine, reboxetine, tranylcypromine, and electroconvulsive seizures (ECS) upregulated Bcl-2 mRNA expression in the Cg, Fr, and CeA. Reboxetine, tranylcypromine, and ECS selectively increased Bcl-xl, but not Bcl-2 mRNA expression in the hippocampus. Chemical ADT but not ECS, robustly enhanced Bcl-2 expression in the medial amygdaloid nucleus and ventromedial hypothalamus. Fluoxetine did not influence Bcl-xl expression in the hippocampus, but it was the only ADT that decreased Bax expression in this region. In the CeA, again in direct contrast to the stress effects, exposure to all classes of ADTs significantly increased Bcl-2 mRNA. The selective regulation of Bcl-xl and Bax in hippocampal subfields and of Bcl-2 in the Cg cortex, amygdala, and hypothalamus suggests that these cellular adaptations contribute to the long-term neural plastic adaptations to stress and ADTs in cortical, hypothalamic, and limbic brain structures.


Subject(s)
Antidepressive Agents/therapeutic use , Brain , Gene Expression Regulation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Stress, Psychological , Animals , Antidepressive Agents/pharmacology , Behavior, Animal , Brain/drug effects , Brain/metabolism , Brain/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Limbic System/drug effects , Limbic System/metabolism , Male , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Sprague-Dawley , Ribonucleases/pharmacology , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Stress, Psychological/pathology , bcl-X Protein/genetics , bcl-X Protein/metabolism
10.
Alcohol Clin Exp Res ; 32(10): 1697-707, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18631322

ABSTRACT

BACKGROUND: Maternal drinking during pregnancy can lead to a range of deleterious outcomes in the developing offspring that have been collectively termed fetal alcohol spectrum disorders (FASDs). There is interest and recognized value in using non-invasive neuroimaging techniques such as magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) to characterize, respectively, structural and biochemical alterations in individuals with FASDs. To date, however, results with MRS have been inconsistent regarding the degree and/or nature of abnormalities. METHODS: High-resolution magic angle spinning (HR-MAS) proton ((1)H) MRS is an ex vivo neuroimaging technique that can acquire spectra in small punches of intact tissue, providing clinically relevant neurochemical information about discrete brain regions. In this study, HR-MAS (1)H MRS was used to examine regional neurochemistry in frontal cortex, striatum, hippocampus, and cerebellum of young rats previously exposed to ethanol as neonates. Key neurochemicals of interest included N-acetyl-aspartate (NAA), glutamate, GABA, glutamine, creatine, choline and myo-inositol. RESULTS: Daily neonatal alcohol exposure from postnatal day 4 (PN4) through PN9 significantly reduced levels of NAA and taurine in the cerebellum and striatum, and induced sex-dependent reductions in cerebellar glutamate when measured on PN16. In addition, myo-inositol was significantly increased in cerebellum. The frontal cortex and hippocampus were virtually unaffected by this neonatal alcohol exposure. CONCLUSION: Results of this research may have implications for understanding the underlying neurobiology associated with FASDs and aid in testing treatments in the future. Ongoing studies are assessing the developmental persistence of and/or maturational recovery from these changes.


Subject(s)
Aspartic Acid/analogs & derivatives , Cerebellum/drug effects , Cerebrum/drug effects , Ethanol/adverse effects , Taurine/metabolism , Amino Acids/metabolism , Animals , Animals, Newborn , Aspartic Acid/metabolism , Body Weight/drug effects , Cerebellum/metabolism , Cerebrum/metabolism , Choline/metabolism , Female , Male , Pregnancy , Rats
11.
Sci Rep ; 8(1): 3798, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29491447

ABSTRACT

Neuronal origins of behavioral disorders have been examined for decades to construct frameworks for understanding psychiatric diseases and developing useful therapeutic strategies with clinical application. Despite abundant anecdotal evidence for white matter etiologies, including altered tractography in neuroimaging and diminished oligodendrocyte-specific gene expression in autopsy studies, mechanistic data demonstrating that dysfunctional myelin sheaths can cause behavioral deficits and perturb neurotransmitter biochemistry have not been forthcoming. At least in part, this impasse stems from difficulties in identifying model systems free of degenerative pathology to enable unambiguous assessment of neuron biology and behavior in a background of myelin dysfunction. Herein we examine myelin mutant mice lacking expression of the Claudin11 gene in oligodendrocytes and characterize two behavioral endophenotypes: perturbed auditory processing and reduced anxiety/avoidance. Importantly, these behaviors are associated with increased transmission time along myelinated fibers as well as glutamate and GABA neurotransmitter imbalances in auditory brainstem and amygdala, in the absence of neurodegeneration. Thus, our findings broaden the etiology of neuropsychiatric disease to include dysfunctional myelin, and identify a preclinical model for the development of novel disease-modifying therapies.


Subject(s)
Behavior, Animal , Claudins/deficiency , Claudins/genetics , Myelin Sheath/metabolism , Neurotransmitter Agents/metabolism , Oligodendroglia/metabolism , Amygdala/metabolism , Animals , Auditory Cortex/pathology , Axons/pathology , Hearing/genetics , Mice , Mutation , Myelin Sheath/physiology
12.
Neuropsychopharmacology ; 31(4): 751-6, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16132064

ABSTRACT

Proton magnetic resonance spectroscopy (1H-MRS) studies of schizophrenia suggest an effect of the disease or of antipsychotic medications on brain N-acetyl aspartate (NAA), a marker of neuronal viability. We studied in rat the effect of haloperidol on NAA, glutamate, and glutamine in several brain regions where metabolite reductions have been reported in chronically medicated patients with schizophrenia. Two groups of 16 rats each were treated with haloperidol depo (38 mg/kg/month) and vehicle for 6 months and were killed. Concentrations of metabolites were determined by high-resolution magic angle proton magnetic resonance spectroscopy (HR-MAS 1H-MRS) at 11.7 T in ex-vivo punch biopsies from the following brain regions: medial frontal and cingulate cortex, striatum, nucleus accumbens, dorsal and ventral hippocampus, amygdala, and temporal cortex. Factorial ANOVA of NAA concentrations revealed no significant effect of drug group (F(1,212) = 1.5; p = 0.22) or a group by brain region interaction (F(7,212) = 1.0; p = 0.43). There was a significant main effect of region (F(7,212) = 17.8; p < 0.001) with lower NAA in the striatum. A prolonged exposure to the dopamine D2 receptor blockade effects of haloperidol does not result in changes in NAA, glutamate, glutamine, and other metabolites in the proton spectrum. These results are consistent with the only other two studies of the effect of antipsychotic drugs on NAA in the rat brain. The documented lower NAA in chronically treated schizophrenia patients is most likely not a simple effect of antipsychotic medications.


Subject(s)
Aspartic Acid/analogs & derivatives , Brain/drug effects , Dopamine Antagonists/pharmacology , Haloperidol/pharmacology , Animals , Aspartic Acid/metabolism , Brain/metabolism , Brain Chemistry/drug effects , Drug Administration Schedule , Glutamic Acid/metabolism , Glutamine/metabolism , Magnetic Resonance Spectroscopy/methods , Male , Rats , Rats, Sprague-Dawley
14.
J Dev Behav Pediatr ; 37(6): 491-5, 2016.
Article in English | MEDLINE | ID: mdl-27011005

ABSTRACT

OBJECTIVE: Treatment of obsessive-compulsive disorder (OCD) is complicated by comorbid psychiatric disorders. Successful treatment of 2 pediatric patients with severe OCD and comorbid attention deficit/hyperactivity disorder (ADHD) is described. METHOD: A report on 2 pediatric clinical cases (Ages 9 and 10) with comorbid OCD and ADHD was used to describe response to medication management through the serotonin transporter inhibitor, sertraline, and the noradrenergic α2A receptor agonist, guanfacine, along with cognitive behavioral therapy. RESULTS: Cognitive behavioral therapy combined with titrated doses of the serotonin transporter inhibitor, sertraline, and the noradrenergic α2A receptor agonist, guanfacine resolved OCD symptoms and the underlying ADHD. CONCLUSION: The novel observations support a focused psychological and pharmacological approach to successful treatment of complex symptoms in patients with comorbid OCD and ADHD. Limitations to generalizability are discussed.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Attention Deficit Disorder with Hyperactivity/therapy , Guanfacine/pharmacology , Obsessive-Compulsive Disorder/therapy , Selective Serotonin Reuptake Inhibitors/pharmacology , Sertraline/pharmacology , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/epidemiology , Child , Cognitive Behavioral Therapy , Combined Modality Therapy , Comorbidity , Drug Therapy, Combination , Guanfacine/administration & dosage , Humans , Male , Obsessive-Compulsive Disorder/drug therapy , Obsessive-Compulsive Disorder/epidemiology , Selective Serotonin Reuptake Inhibitors/administration & dosage , Sertraline/administration & dosage
15.
J Neurotrauma ; 33(17): 1614-24, 2016 09 01.
Article in English | MEDLINE | ID: mdl-26529240

ABSTRACT

Mild traumatic brain injury (mTBI) contributes to development of affective disorders, including post-traumatic stress disorder (PTSD). Psychiatric symptoms typically emerge in a tardive fashion post-TBI, with negative effects on recovery. Patients with PTSD, as well as rodent models of PTSD, demonstrate structural and functional changes in brain regions mediating fear learning, including prefrontal cortex (PFC), amygdala (AMYG), and hippocampus (HC). These changes may reflect loss of top-down control by which PFC normally exhibits inhibitory influence over AMYG reactivity to fearful stimuli, with HC contribution. Considering the susceptibility of these regions to injury, we examined fear conditioning (FC) in the delayed post-injury period, using a mouse model of mTBI. Mice with mTBI displayed enhanced acquisition and delayed extinction of FC. Using proton magnetic resonance spectroscopy ex vivo, we examined PFC, AMYG, and HC levels of gamma-aminobutyric acid (GABA) and glutamate as surrogate measures of inhibitory and excitatory neurotransmission, respectively. Eight days post-injury, GABA was increased in PFC, with no significant changes in AMYG. In animals receiving FC and mTBI, glutamate trended toward an increase and the GABA/glutamate ratio decreased in ventral HC at 25 days post-injury, whereas GABA decreased and GABA/glutamate decreased in dorsal HC. These neurochemical changes are consistent with early TBI-induced PFC hypoactivation facilitating the fear learning circuit and exacerbating behavioral fear responses. The latent emergence of overall increased excitatory tone in the HC, despite distinct plasticity in dorsal and ventral HC fields, may be associated with disordered memory function, manifested as incomplete extinction and enhanced FC recall.


Subject(s)
Brain Concussion/physiopathology , Extinction, Psychological/physiology , Hippocampus/physiopathology , gamma-Aminobutyric Acid/metabolism , Animals , Behavior, Animal , Brain Concussion/psychology , Conditioning, Classical , Fear , Male , Mice , Mice, Inbred C57BL , Stress Disorders, Post-Traumatic/etiology , Stress Disorders, Post-Traumatic/physiopathology
16.
Behav Brain Res ; 303: 228-37, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26821287

ABSTRACT

Appropriate animal models of posttraumatic stress disorder (PTSD) are needed because human studies remain limited in their ability to probe the underlying neurobiology of PTSD. Although the single prolonged stress (SPS) model is an established rat model of PTSD, the development of a similarly-validated mouse model emphasizes the benefits and cross-species utility of rodent PTSD models and offers unique methodological advantages to that of the rat. Therefore, the aims of this study were to develop and describe a SPS model for mice and to provide data that support current mechanisms relevant to PTSD. The mouse single prolonged stress (mSPS) paradigm, involves exposing C57Bl/6 mice to a series of severe, multimodal stressors, including 2h restraint, 10 min group forced swim, exposure to soiled rat bedding scent, and exposure to ether until unconsciousness. Following a 7-day undisturbed period, mice were tested for cue-induced fear behavior, effects of paroxetine on cue-induced fear behavior, extinction retention of a previously extinguished fear memory, dexamethasone suppression of corticosterone (CORT) response, dorsal hippocampal glucocorticoid receptor protein and mRNA expression, and prefrontal cortex glutamate levels. Exposure to mSPS enhanced cue-induced fear, which was attenuated by oral paroxetine treatment. mSPS also disrupted extinction retention, enhanced suppression of stress-induced CORT response, increased mRNA expression of dorsal hippocampal glucocorticoid receptors and decreased prefrontal cortex glutamate levels. These data suggest that the mSPS model is a translationally-relevant model for future PTSD research with strong face, construct, and predictive validity. In summary, mSPS models characteristics relevant to PTSD and this severe, multimodal stress modifies fear learning in mice that coincides with changes in the hypothalamo-pituitary-adrenal (HPA) axis, brain glucocorticoid systems, and glutamatergic signaling in the prefrontal cortex.


Subject(s)
Disease Models, Animal , Fear , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/psychology , Stress, Psychological/complications , Animals , Behavior, Animal/drug effects , Conditioning, Classical , Corticosterone/blood , Cues , Extinction, Psychological , Fear/drug effects , Glutamic Acid/metabolism , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Paroxetine/administration & dosage , Prefrontal Cortex/metabolism , Receptors, Glucocorticoid/metabolism , Restraint, Physical , Selective Serotonin Reuptake Inhibitors/administration & dosage , Stress Disorders, Post-Traumatic/etiology , Swimming
17.
Sci Rep ; 5: 15075, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26537106

ABSTRACT

Few preclinical studies have assessed the long-term neuropathology and behavioral deficits after sustaining blast-induced neurotrauma (BINT). Previous studies have shown extensive astrogliosis and cell death at acute stages (<7 days) but the temporal response at a chronic stage has yet to be ascertained. Here, we used behavioral assays, immmunohistochemistry and neurochemistry in limbic areas such as the amygdala (Amy), Hippocampus (Hipp), nucleus accumbens (Nac), and prefrontal cortex (PFC), to determine the long-term effects of a single blast exposure. Behavioral results identified elevated avoidance behavior and decreased short-term memory at either one or three months after a single blast event. At three months after BINT, markers for neurodegeneration (FJB) and microglia activation (Iba-1) increased while index of mature neurons (NeuN) significantly decreased in all brain regions examined. Gliosis (GFAP) increased in all regions except the Nac but only PFC was positive for apoptosis (caspase-3). At three months, tau was selectively elevated in the PFC and Hipp whereas α-synuclein transiently increased in the Hipp at one month after blast exposure. The composite neurochemical measure, myo-inositol+glycine/creatine, was consistently increased in each brain region three months following blast. Overall, a single blast event resulted in enduring long-term effects on behavior and neuropathological sequelae.


Subject(s)
Amygdala/pathology , Brain Injuries/pathology , Hippocampus/pathology , Memory, Short-Term/physiology , Neurons/pathology , Nucleus Accumbens/pathology , Prefrontal Cortex/pathology , Amygdala/metabolism , Animals , Apoptosis/physiology , Brain Injuries/metabolism , Caspase 3/metabolism , Disease Models, Animal , Gliosis/metabolism , Gliosis/pathology , Hippocampus/metabolism , Male , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , alpha-Synuclein/metabolism
18.
Behav Brain Res ; 284: 218-24, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25712697

ABSTRACT

Posttraumatic stress disorder (PTSD) is often comorbid with substance use disorders (SUD). Single prolonged stress (SPS) is a well-validated rat model of PTSD that provides a framework to investigate drug-induced behaviors as a preclinical model of the comorbidity. We hypothesized that cocaine sensitization and self-administration would be increased following exposure to SPS. Male Sprague-Dawley rats were exposed to SPS or control treatment. After SPS, cocaine (0, 10 or 20 mg/kg, i.p.) was administered for 5 consecutive days and locomotor activity was measured. Another cohort was assessed for cocaine self-administration (0.1 or 0.32 mg/kg/i.v.) after SPS. Rats were tested for acquisition, extinction and cue-induced reinstatement behaviors. Control animals showed a dose-dependent increase in cocaine-induced locomotor activity after acute cocaine whereas SPS rats did not. Using a sub-threshold sensitization paradigm, control rats did not exhibit enhanced locomotor activity at Day 5 and therefore did not develop behavioral sensitization, as expected. However, compared to control rats on Day 5 the locomotor response to 20mg/kg repeated cocaine was greatly enhanced in SPS-treated rats, which exhibited enhanced cocaine locomotor sensitization. The effect of SPS on locomotor activity was unique in that SPS did not modify cocaine self-administration behaviors under a simple schedule of reinforcement. These data show that SPS differentially affects cocaine-mediated behaviors causing no effect to cocaine self-administration, under a simple schedule of reinforcement, but significantly augmenting cocaine locomotor sensitization. These results suggest that SPS shares common neurocircuitry with stimulant-induced plasticity, but dissociable from that underlying psychostimulant-induced reinforcement.


Subject(s)
Cocaine-Related Disorders/physiopathology , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Stress, Psychological/physiopathology , Akathisia, Drug-Induced/physiopathology , Animals , Catheters, Indwelling , Cohort Studies , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Male , Rats, Sprague-Dawley , Self Administration
19.
Neuroreport ; 14(7): 991-4, 2003 May 23.
Article in English | MEDLINE | ID: mdl-12802189

ABSTRACT

This study examined the effects of cued vs non-cued food delivery/consumption on extracellular glutamate and dopamine in the nucleus accumbens of food-deprived rats. Animals that always received a food pellet following a series of auditory tones showed a significant decrease in extracellular glutamate following food consumption, whereas animals that had not been previously exposed to tone-food pairing did not (p<0.05). In contrast, extracellular dopamine was significantly increased in the nucleus accumbens during the first time period after food consumption (p<0.05) regardless of whether animals had been exposed to prior tone-food pairing. Results suggest that food delivery/consumption is associated with a decrease in accumbal glutamate if food delivery has been previously paired with predictive environmental cues.


Subject(s)
Cues , Eating/physiology , Extracellular Space/metabolism , Food Deprivation/physiology , Glutamic Acid/metabolism , Nucleus Accumbens/metabolism , Acoustic Stimulation , Animals , Male , Rats , Rats, Sprague-Dawley
20.
Article in English | MEDLINE | ID: mdl-24246571

ABSTRACT

Gamma-aminobutyric acid (GABA) is the major inhibitory amino acid neurotransmitter in the brain and is primarily responsible for modulating excitatory tone. Clinical neuroimaging studies show decreased GABA levels in the anterior cingulate of patients with mood disorders, including major depressive disorder. Chronic unpredictable stress (CUS) is an animal model thought to mimic the stressful events that may precipitate clinical depression in humans. In this study male Sprague-Dawley rats were subjected to a modified CUS paradigm that used a random pattern of unpredictable stressors twice daily for 10 days to explore the early developmental stages of depression-like endophenotypes. Control rats were handled daily for 10 days. Some rats from each treatment group received an injection of ketamine (40 mg/kg) after the final stressor. One day following the final stressor rats were tested for behavioral effects in the forced swim test and then euthanized to collect trunk blood and anterior cingulate brain samples. GABA levels were measured in anterior cingulate samples ex vivo using proton magnetic resonance spectroscopy ((1)H-MRS) at 11.7 T. Animals subjected to CUS had lower body weights, higher levels of blood corticosterone, and increased immobility in the forced swim test; all of which suggest that the stress paradigm induced a depression-like phenotype. GABA levels in the anterior cingulate were significantly increased in the stressed animals compared to controls. Administration of ketamine on the last day of treatment blunted the depression-like behavior and increased GABA levels in the anterior cingulate following CUS. These data indicate that stress disrupts GABAergic signaling, which may over time lead to symptoms of depression and ultimately lower basal levels of cortical (1)H-MRS GABA that are seen in humans with depression. Furthermore, the data suggests that ketamine modulates cortical GABA levels as a mechanism of its antidepressant activity.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/drug therapy , Depression/pathology , Gyrus Cinguli/metabolism , Ketamine/therapeutic use , gamma-Aminobutyric Acid/metabolism , Analysis of Variance , Animals , Chromatography, High Pressure Liquid , Depression/etiology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gyrus Cinguli/drug effects , Immobility Response, Tonic/drug effects , Magnetic Resonance Spectroscopy , Male , Protons , Rats , Rats, Sprague-Dawley , Stress, Psychological/complications , Swimming/psychology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL