Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Pharmacol Ther ; 256: 108609, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38369062

ABSTRACT

Traumatic brain injury (TBI) is a highly prevalent medical condition for which no medications specific for the prophylaxis or treatment of the condition as a whole exist. The spectrum of symptoms includes coma, headache, seizures, cognitive impairment, depression, and anxiety. Although it has been known for years that the inhibitory neurotransmitter γ-amino-butyric acid (GABA) is involved in TBI, no novel therapeutics based upon this mechanism have been introduced into clinical practice. We review the neuroanatomical, neurophysiological, neurochemical, and neuropharmacological relationships of GABA neurotransmission to TBI with a view toward new potential GABA-based medicines. The long-standing idea that excitatory and inhibitory (GABA and others) balances are disrupted by TBI is supported by the experimental data but has failed to invent novel methods of restoring this balance. The slow progress in advancing new treatments is due to the complexity of the disorder that encompasses multiple dynamically interacting biological processes including hemodynamic and metabolic systems, neurodegeneration and neurogenesis, major disruptions in neural networks and axons, frank brain lesions, and a multitude of symptoms that have differential neuronal and neurohormonal regulatory mechanisms. Although the current and ongoing clinical studies include GABAergic drugs, no novel GABA compounds are being explored. It is suggested that filling the gap in understanding the roles played by specific GABAA receptor configurations within specific neuronal circuits could help define new therapeutic approaches. Further research into the temporal and spatial delivery of GABA modulators should also be useful. Along with GABA modulation, research into the sequencing of GABA and non-GABA treatments will be needed.


Subject(s)
Brain Injuries, Traumatic , Humans , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , gamma-Aminobutyric Acid/metabolism , Seizures/metabolism , Synaptic Transmission/physiology , Neurons/metabolism
2.
Exp Biol Med (Maywood) ; 248(7): 596-604, 2023 04.
Article in English | MEDLINE | ID: mdl-37208920

ABSTRACT

Hyperexcitability is a major mechanism implicated in several neuropsychiatric disorders, such as organophosphate-induced status epilepticus (SE), primary epilepsy, stroke, spinal cord injury, traumatic brain injury, schizophrenia, and autism spectrum disorders. Underlying mechanisms are diverse, but a functional impairment and loss of GABAergic inhibitory neurons are common features in many of these disorders. While novel therapies abound to correct for the loss of GABAergic inhibitory neurons, it has been difficult at best to improve the activities of daily living for the majority of patients. Alpha-linolenic acid (ALA) is an essential omega-3 polyunsaturated fatty acid found in plants. ALA exerts pleiotropic effects in the brain that attenuate injury in chronic and acute brain disease models. However, the effect of ALA on GABAergic neurotransmission in hyperexcitable brain regions involved in neuropsychiatric disorders, such as the basolateral amygdala (BLA) and CA1 subfield of the hippocampus, is unknown. Administration of a single dose of ALA (1500 nmol/kg) subcutaneously increased the charge transfer of inhibitory postsynaptic potential currents mediated by GABAA receptors in pyramidal neurons by 52% in the BLA and by 92% in the CA1 compared to vehicle animals a day later. Similar results were obtained in pyramidal neurons from the BLA and CA1 when ALA was bath-applied in slices from naïve animals. Importantly, pretreatment with the high-affinity, selective TrkB inhibitor, k252, completely abolished the ALA-induced increase in GABAergic neurotransmission in the BLA and CA1, suggesting a brain-derived neurotrophic factor (BDNF)-mediated mechanism. Addition of mature BDNF (20 ng/mL) significantly increased GABAA receptor inhibitory activity in the BLA and CA1 pyramidal neurons similar to the results obtained with ALA. ALA may be an effective treatment for neuropsychiatric disorders where hyperexcitability is a major feature.


Subject(s)
Basolateral Nuclear Complex , Rats , Humans , Animals , Basolateral Nuclear Complex/metabolism , alpha-Linolenic Acid/pharmacology , Brain-Derived Neurotrophic Factor , Rats, Sprague-Dawley , Activities of Daily Living , Synaptic Transmission/physiology , Receptors, GABA-A/metabolism
3.
Am J Vet Res ; 84(3)2023 Jan 26.
Article in English | MEDLINE | ID: mdl-36662603

ABSTRACT

Nearly all of the American horses exported to Mexico and Canada are slaughtered for human consumption, and their meat is either exported around the world or consumed locally. Previous work showed that 18 Thoroughbred racehorses purchased by rescues that would have otherwise been sold for export for the sole purpose of slaughter to produce meat for human consumption were administered phenylbutazone. We report the number of American horses exported to Canada and Mexico from 2016 to 2021, the presence of contaminated horsemeat from Canadian slaughterhouses, and the human use and idiosyncratic effects of veterinary phenylbutazone and side effects of clenbuterol, 2 of the drugs that were found in contaminated Canadian horsemeat. The number of live American horses exported to Canada declined precipitously from 2016 to 2017, and a second decline occurred in 2020. All food-producing animals are under strict regulatory control to prevent animals administered banned drugs to enter the food chain. A major principle of this program is zero tolerance for banned drugs and testing for compliance. No regulatory process is in place to remove horses administered banned drugs such as phenylbutazone. The efficacy lasts for more than 24 hours as a result of the irreversible binding to cyclooxygenase, slow elimination, and long elimination half-life of its metabolite oxyphenbutazone. High or frequent doses of phenylbutazone result in disproportionately increased plasma concentrations, which result in the residual presence in tissues. It is this fact that underlies the ban of this drug in food-producing animals. No human clinical surveillance program is in place to monitor individuals on the possible short- and long-term consequences of banned drugs in contaminated horsemeat. If the United States is unable to put in place a regulatory program to remove horses administered banned drugs as exists for all food-producing animals, the exportation of American horses across both borders for the sole purpose of slaughter for human consumption must end.


Subject(s)
Phenylbutazone , Public Health , Horses , Humans , Animals , United States , Canada , Oxyphenbutazone
4.
Amino Acids ; 54(9): 1229-1249, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35798984

ABSTRACT

Traumatic brain injury (TBI) has reached epidemic proportions around the world and is a major public health concern in the United States. Approximately 2.8 million individuals sustain a traumatic brain injury and are treated in an Emergency Department yearly in the U.S., and about 50,000 of them die. Persistent symptoms develop in 10-15% of the cases including neuropsychiatric disorders. Anxiety is the second most common neuropsychiatric disorder that develops in those with persistent neuropsychiatric symptoms after TBI. Abnormalities or atrophy in the temporal lobe has been shown in the overwhelming number of TBI cases. The basolateral amygdala (BLA), a temporal lobe structure that consolidates, stores and generates fear and anxiety-based behavioral outputs, is a critical brain region in the anxiety circuitry. In this review, we sought to capture studies that characterized the relationship between human post-traumatic anxiety and structural/functional alterations in the amygdala. We compared the human findings with results obtained with a reproducible mild TBI animal model that demonstrated a direct relationship between the alterations in the BLA and an anxiety-like phenotype. From this analysis, both preliminary insights, and gaps in knowledge, have emerged which may open new directions for the development of rational and more efficacious treatments.


Subject(s)
Basolateral Nuclear Complex , Brain Injuries, Traumatic , Animals , Anxiety , Brain , Humans
5.
Mol Neurobiol ; 57(1): 23-31, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31520315

ABSTRACT

Traumatic brain injury (TBI) is a major public health concern in the USA. There are approximately 2.5 million brain injuries annually, 90% of which may be classified as mild since these individuals do not display clear morphological abnormalities following injury on imaging. The majority of individuals develop neurocognitive deficits such as learning and memory impairment and recovery occurs over 3 to 6 months after mild TBI (mTBI). The hippocampus is highly susceptible to injury from mTBI due to the anatomic localization and has been implicated in the neurocognitive impairments after mTBI. Here, we investigated whether the mTBI-induced morphological and pathophysiological alterations of GABAergic interneurons in the CA1 subfield of the hippocampus recovers after 30 days in the controlled cortical impact (CCI) model of TBI. Design-based stereology shows a significant reduction in the number of GABAergic interneurons 7 days after CCI. However, the number of GABAergic interneurons is not significantly reduced at 30 days after CCI. The total number of neurons is not altered over the course of 30 days. GABAergic inhibitory currents in the CA1 subfield also show that, although there is a significant reduction in the CCI group at 7 days, the currents are not significantly different from sham controls at 30 days. We suggest that the recovery of GABAergic function in the CA1 subfield of the hippocampus observed 30 days after CCI is one of the mechanisms associated with the recovery of memory after mTBI.


Subject(s)
Brain Injuries, Traumatic/physiopathology , CA1 Region, Hippocampal/physiopathology , Hippocampus/physiopathology , Inhibitory Postsynaptic Potentials/physiology , Animals , Brain Injuries/complications , Brain Injuries/physiopathology , Brain Injuries, Traumatic/complications , Disease Models, Animal , GABAergic Neurons , Interneurons/metabolism , Male , Memory/physiology , Memory Disorders/complications , Rats, Sprague-Dawley
6.
Epilepsia ; 59 Suppl 2: 92-99, 2018 10.
Article in English | MEDLINE | ID: mdl-30159887

ABSTRACT

Nerve agents are organophosphate (OP) compounds and among the most powerful poisons known to man. A terrorist attack on civilian or military populations causing mass casualties is a real threat. The OP nerve agents include soman, sarin, cyclosarin, tabun, and VX. The major mechanism of acute toxicity is the irreversible inhibition of acetylcholinesterase. Acetylcholinesterase inhibition results in the accumulation of excessive acetylcholine levels in synapses, leading to progression of toxic signs including hypersecretions, tremors, status epilepticus, respiratory distress, and death. Miosis and rhinorrhea are the most common clinical findings in those individuals acutely exposed to OP nerve agents. Prolonged seizures are responsible for the neuropathology. The brain region that shows the most severe damage is the amygdala, followed by the piriform cortex, hippocampus, cortex, thalamus, and caudate/putamen. Current medical countermeasures are only modestly effective in attenuating the seizures and neuropathology. Anticonvulsants such as benzodiazepines decrease seizure activity and improve outcome, but their efficacy depends upon the administration time after exposure to the nerve agent. Administration of benzodiazepines may increase the risk for seizure recurrence. Recent studies document long-term neurologic and behavior deficits, and technological advances demonstrate structural brain changes on magnetic resonance imaging.


Subject(s)
Brain/drug effects , Nerve Agents/toxicity , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Brain/metabolism , Humans , Miosis/drug therapy , Miosis/etiology , Respiration Disorders/chemically induced , Respiration Disorders/drug therapy , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Time Factors
7.
Mol Neurobiol ; 55(1): 187-200, 2018 01.
Article in English | MEDLINE | ID: mdl-28844093

ABSTRACT

Approximately, 1.7 million Americans suffer a TBI annually and TBI is a major cause of death and disability. The majority of the TBI cases are of the mild type and while most patients recover completely from mild TBI (mTBI) about 10% result in persistent symptoms and some result in lifelong disability. Anxiety disorders are the second most common diagnosis post-TBI. Of note, TBI-induced anxiety disorders are difficult to treat and remain a chronic condition suggesting that new therapies are needed. Previous work from our laboratory demonstrated that a mild TBI induced an anxiety-like phenotype, a key feature of the human condition, associated with loss of GABAergic interneurons and hyperexcitability in the basolateral amygdala (BLA) in rodents 7 and 30 days after a controlled cortical impact (CCI) injury. We now confirm that animals display significantly increased anxiety-like behavior 30 days after CCI. The anxiety-like behavior was associated with a significant loss of GABAergic interneurons and significant reductions in the frequency and amplitude of spontaneous and miniature GABAA-receptor-mediated inhibitory postsynaptic currents (IPSCs) in the BLA. Significantly, subchronic treatment with alpha-linolenic acid (ALA) after CCI prevents the development of anxiety-like behavior, the loss of GABAergic interneurons, hyperexcitability in the BLA and reduces the impact injury. Taken together, administration of ALA after CCI is a potent therapy against the neuropathology and pathophysiological effects of mTBI in the BLA.


Subject(s)
Anxiety/prevention & control , Brain Injuries, Traumatic/drug therapy , Contusions/drug therapy , alpha-Linolenic Acid/therapeutic use , Animals , Anxiety/etiology , Anxiety/physiopathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/physiopathology , Contusions/etiology , Contusions/physiopathology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Rats , Rats, Sprague-Dawley , Treatment Outcome , alpha-Linolenic Acid/pharmacology
8.
Toxicol Sci ; 161(1): 103-114, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29029261

ABSTRACT

Domoic acid (DOM) is an excitatory amino acid analog of kainic acid (KA) that acts through glutamic acid (GLU) receptors, inducing a fast and potent neurotoxic response. Here, we present evidence for an enhancement of excitotoxicity following exposure of cultured cerebellar granule cells to DOM in the presence of lower than physiological Na+ concentrations. The concentration of DOM that reduced by 50% neuronal survival was approximately 3 µM in Na+-free conditions and 16 µM in presence of a physiological concentration of extracellular Na+. The enhanced neurotoxic effect of DOM was fully prevented by AMPA/KA receptor antagonist, while N-methyl-D-aspartate-receptor-mediated neurotoxicity did not seem to be involved, as the absence of extracellular Na+ failed to potentiate GLU excitotoxicity under the same experimental conditions. Lowering of extracellular Na+ concentration to 60 mM eliminated extracellular recording of spontaneous electrophysiological activity from cultured neurons grown on a multi electrode array and prevented DOM stimulation of the electrical activity. Although changes in the extracellular Na+ concentration did not alter the magnitude of the rapid increase in intracellular Ca2+ levels associated to DOM exposure, they did change significantly the contribution of voltage-sensitive calcium channels (VScaCs) and the recovery time to baseline. The prevention of Ca2+ influx via VSCaCs by nifedipine failed to prevent DOM toxicity at any extracellular Na+ concentration, while the reduction of extracellular Ca2+ concentration ameliorated DOM toxicity only in the absence of extracellular Na+, enhancing it in physiological conditions. Our data suggest a crucial role for extracellular Na+ concentration in determining excitotoxicity by DOM.


Subject(s)
Cerebellum/drug effects , GABAergic Neurons/drug effects , Kainic Acid/analogs & derivatives , Neurotoxins/toxicity , Sodium/metabolism , Animals , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Extracellular Space , GABAergic Neurons/metabolism , Kainic Acid/toxicity , Mice , Primary Cell Culture , Rats , Receptors, Glutamate
9.
Aging Cell ; 16(5): 1125-1135, 2017 10.
Article in English | MEDLINE | ID: mdl-28772063

ABSTRACT

The dietary intake of ω-3 polyunsaturated fatty acids has been linked to a reduction in the incidence of aging-associated disease including cardiovascular disease and stroke. Additionally, long-lived Caenorhabditis elegans glp-1 germ line-less mutant animals show a number of changes in lipid metabolism including the increased production of the ω-3 fatty acid, α-linolenic acid (ALA). Here, we show that the treatment of C. elegans with ALA produces a dose-dependent increase in lifespan. The increased longevity of the glp-1 mutant animals is known to be dependent on both the NHR-49/PPARα and SKN-1/Nrf2 transcription factors, although the mechanisms involved are incompletely understood. We find that ALA treatment increased the lifespan of wild-type worms and that these effects required both of these transcription factors. Specifically, NHR-49 was activated by ALA to promote the expression of genes involved in the ß-oxidation of lipids, whereas SKN-1 is not directly activated by ALA, but instead, the exposure of ALA to air results in the oxidation of ALA to a group of compounds termed oxylipins. At least one of the oxylipins activates SKN-1 and enhances the increased longevity resulting from ALA treatment. The results show that ω-3 fatty acids inhibit aging and that these effects could reflect the combined effects of the ω-3 fatty acid and the oxylipin metabolites. The benefits of ω-3 fatty acid consumption on human health may similarly involve the production of oxylipins, and differences in oxylipin conversion could account for at least part of the variability found between observational vs. interventional clinical trials.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/drug effects , Longevity/drug effects , Oxylipins/metabolism , PPAR alpha/genetics , Receptors, Cytoplasmic and Nuclear/genetics , alpha-Linolenic Acid/pharmacology , Animals , Biotransformation , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Developmental , Lipid Metabolism , Longevity/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , PPAR alpha/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , alpha-Linolenic Acid/metabolism
10.
Molecules ; 20(11): 20355-80, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26569216

ABSTRACT

α-Linolenic acid (ALA) is a nutraceutical found in vegetable products such as flax and walnuts. The pleiotropic properties of ALA target endogenous neuroprotective and neurorestorative pathways in brain and involve the transcription factor nuclear factor kappa B (NF-κB), brain-derived neurotrophic factor (BDNF), a major neuroprotective protein in brain, and downstream signaling pathways likely mediated via activation of TrkB, the cognate receptor of BDNF. In this review, we discuss possible mechanisms of ALA efficacy against the highly toxic OP nerve agent soman. Organophosphate (OP) nerve agents are highly toxic chemical warfare agents and a threat to military and civilian populations. Once considered only for battlefield use, these agents are now used by terrorists to inflict mass casualties. OP nerve agents inhibit the critical enzyme acetylcholinesterase (AChE) that rapidly leads to a cholinergic crisis involving multiple organs. Status epilepticus results from the excessive accumulation of synaptic acetylcholine which in turn leads to the overactivation of muscarinic receptors; prolonged seizures cause the neuropathology and long-term consequences in survivors. Current countermeasures mitigate symptoms and signs as well as reduce brain damage, but must be given within minutes after exposure to OP nerve agents supporting interest in newer and more effective therapies. The pleiotropic properties of ALA result in a coordinated molecular and cellular program to restore neuronal networks and improve cognitive function in soman-exposed animals. Collectively, ALA should be brought to the clinic to treat the long-term consequences of nerve agents in survivors. ALA may be an effective therapy for other acute and chronic neurodegenerative disorders.


Subject(s)
Dietary Supplements , Nerve Agents/adverse effects , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Organophosphates/adverse effects , Signal Transduction/drug effects , alpha-Linolenic Acid/pharmacology , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Cognition/drug effects , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/metabolism , Humans , Models, Animal , Nervous System Diseases/chemically induced , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neuropathology , Neuroprotective Agents/therapeutic use , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Linolenic Acid/therapeutic use
11.
Neurotoxicology ; 51: 38-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26386148

ABSTRACT

Exposure to nerve agents results in severe seizures or status epilepticus caused by the inhibition of acetylcholinesterase, a critical enzyme that breaks down acetylcholine to terminate neurotransmission. Prolonged seizures cause brain damage and can lead to long-term consequences. Current countermeasures are only modestly effective against the brain damage supporting interest in the evaluation of new and efficacious therapies. The nutraceutical alpha-linolenic acid (LIN) is an essential omega-3 polyunsaturated fatty acid that has a wide safety margin. Previous work showed that a single intravenous injection of alpha-linolenic acid (500 nmol/kg) administered before or after soman significantly protected against soman-induced brain damage when analyzed 24h after exposure. Here, we show that administration of three intravenous injections of alpha-linolenic acid over a 7 day period after soman significantly improved motor performance on the rotarod, enhanced memory retention, exerted an anti-depressant-like activity and increased animal survival. This dosing schedule significantly reduced soman-induced neuronal degeneration in four major vulnerable brain regions up to 21 days. Taken together, alpha-linolenic acid reduces the profound behavioral deficits induced by soman possibly by decreasing neuronal cell death, and increases animal survival.


Subject(s)
Antidepressive Agents/administration & dosage , Cognition/drug effects , Neuroprotective Agents/administration & dosage , Soman/toxicity , alpha-Linolenic Acid/administration & dosage , Animals , Avoidance Learning/drug effects , Body Weight/drug effects , Brain/drug effects , Brain/pathology , Dietary Supplements , Male , Motor Activity/drug effects , Neurons/drug effects , Rats, Sprague-Dawley , Rotarod Performance Test
12.
J Mol Neurosci ; 57(2): 282-303, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26319264

ABSTRACT

We have previously demonstrated that mild controlled cortical impact (mCCI) injury to rat cortex causes indirect, concussive injury to underlying hippocampus and other brain regions, providing a reproducible model for mild traumatic brain injury (mTBI) and its neurochemical, synaptic, and behavioral sequelae. Here, we extend a preliminary gene expression study of the hippocampus-specific events occurring after mCCI and identify 193 transcripts significantly upregulated, and 21 transcripts significantly downregulated, 24 h after mCCI. Fifty-three percent of genes altered by mCCI within 24 h of injury are predicted to be expressed only in the non-neuronal/glial cellular compartment, with only 13% predicted to be expressed only in neurons. The set of upregulated genes following mCCI was interrogated using Ingenuity Pathway Analysis (IPA) augmented with manual curation of the literature (190 transcripts accepted for analysis), revealing a core group of 15 first messengers, mostly inflammatory cytokines, predicted to account for >99% of the transcript upregulation occurring 24 h after mCCI. Convergent analysis of predicted transcription factors (TFs) regulating the mCCI target genes, carried out in IPA relative to the entire Affymetrix-curated transcriptome, revealed a high concordance with TFs regulated by the cohort of 15 cytokines/cytokine-like messengers independently accounting for upregulation of the mCCI transcript cohort. TFs predicted to regulate transcription of the 193-gene mCCI cohort also displayed a high degree of overlap with TFs predicted to regulate glia-, rather than neuron-specific genes in cortical tissue. We conclude that mCCI predominantly affects transcription of non-neuronal genes within the first 24 h after insult. This finding suggests that early non-neuronal events trigger later permanent neuronal changes after mTBI, and that early intervention after mTBI could potentially affect the neurochemical cascade leading to later reported synaptic and behavioral dysfunction.


Subject(s)
Brain Injuries/metabolism , Hippocampus/metabolism , Transcriptome , Animals , Brain Injuries/pathology , Cerebral Cortex/injuries , Cerebral Cortex/metabolism , Cytokines/genetics , Cytokines/metabolism , Male , Neuroglia/metabolism , Neurons/metabolism , Organ Specificity , Rats , Rats, Sprague-Dawley
13.
Exp Neurol ; 273: 11-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26238734

ABSTRACT

Patients that suffer mild traumatic brain injuries (mTBI) often develop cognitive impairments, including memory and learning deficits. The hippocampus shows a high susceptibility to mTBI-induced damage due to its anatomical localization and has been implicated in cognitive and neurological impairments after mTBI. However, it remains unknown whether mTBI cognitive impairments are a result of morphological and pathophysiological alterations occurring in the CA1 hippocampal region. We investigated whether mTBI induces morphological and pathophysiological alterations in the CA1 using the controlled cortical impact (CCI) model. Seven days after CCI, animals subjected to mTBI showed cognitive impairment in the passive avoidance test and deficits to long-term potentiation (LTP) of synaptic transmission. Deficiencies in inducing or maintaining LTP were likely due to an observed reduction in the activation of NMDA but not AMPA receptors. Significant reductions in the frequency and amplitude of spontaneous and miniature GABAA-receptor mediated inhibitory postsynaptic currents (IPSCs) were also observed 7 days after CCI. Design-based stereology revealed that although the total number of neurons was unaltered, the number of GABAergic interneurons is significantly reduced in the CA1 region 7 days after CCI. Additionally, the surface expression of α1, ß2/3, and γ2 subunits of the GABAA receptor were reduced, contributing to a reduced mIPSC frequency and amplitude, respectively. Together, these results suggest that mTBI causes a significant reduction in GABAergic inhibitory transmission and deficits to NMDA receptor mediated currents in the CA1, which may contribute to changes in hippocampal excitability and subsequent cognitive impairments after mTBI.


Subject(s)
Brain Injuries/pathology , CA1 Region, Hippocampal/pathology , GABAergic Neurons/pathology , Inhibitory Postsynaptic Potentials/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Avoidance Learning/physiology , Brain Injuries/complications , Disease Models, Animal , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Glucose Transporter Type 1/metabolism , Glutamate Decarboxylase/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/pathology , Male , Memory Disorders/etiology , Rats , Rats, Sprague-Dawley , Reaction Time/physiology , Receptors, GABA-A/metabolism , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Time Factors
14.
Neuromolecular Med ; 17(3): 251-69, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25920465

ABSTRACT

Exposure to organophosphorous (OP) nerve agents such as soman inhibits the critical enzyme acetylcholinesterase (AChE) leading to excessive acetylcholine accumulation in synapses, resulting in cholinergic crisis, status epilepticus and brain damage in survivors. The hippocampus is profoundly damaged after soman exposure leading to long-term memory deficits. We have previously shown that treatment with three sequential doses of alpha-linolenic acid, an essential omega-3 polyunsaturated fatty acid, increases brain plasticity in naïve animals. However, the effects of this dosing schedule administered after a brain insult and the underlying molecular mechanisms in the hippocampus are unknown. We now show that injection of three sequential doses of alpha-linolenic acid after soman exposure increases the endogenous expression of mature BDNF, activates Akt and the mammalian target of rapamycin complex 1 (mTORC1), increases neurogenesis in the subgranular zone of the dentate gyrus, increases retention latency in the passive avoidance task and increases animal survival. In sharp contrast, while soman exposure also increases mature BDNF, this increase did not activate downstream signaling pathways or neurogenesis. Administration of the inhibitor of mTORC1, rapamycin, blocked the alpha-linolenic acid-induced neurogenesis and the enhanced retention latency but did not affect animal survival. Our results suggest that alpha-linolenic acid induces a long-lasting neurorestorative effect that involves activation of mTORC1 possibly via a BDNF-TrkB-mediated mechanism.


Subject(s)
Avoidance Learning/drug effects , Hippocampus/drug effects , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Soman/toxicity , alpha-Linolenic Acid/pharmacology , Animals , Antigens, Nuclear/biosynthesis , Antigens, Nuclear/genetics , Atropine Derivatives/therapeutic use , Avoidance Learning/physiology , Brain Damage, Chronic/etiology , Brain Damage, Chronic/physiopathology , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , DNA Replication/drug effects , Diazepam/therapeutic use , Doublecortin Domain Proteins , Electroshock , Exploratory Behavior/drug effects , Hippocampus/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1 , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuropeptides/biosynthesis , Neuropeptides/genetics , Neuroprotective Agents/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Neurotoxins/metabolism , Oximes/therapeutic use , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Pyridinium Compounds/therapeutic use , Rats , Rats, Sprague-Dawley , Receptor, trkB/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology , Status Epilepticus/chemically induced , Status Epilepticus/complications , Status Epilepticus/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/biosynthesis , TOR Serine-Threonine Kinases/genetics , alpha-Linolenic Acid/antagonists & inhibitors , alpha-Linolenic Acid/therapeutic use
15.
Biomed Res Int ; 2015: 519830, 2015.
Article in English | MEDLINE | ID: mdl-25789320

ABSTRACT

Alpha-linolenic acid (ALA) is plant-based essential omega-3 polyunsaturated fatty acids that must be obtained through the diet. This could explain in part why the severe deficiency in omega-3 intake pointed by numerous epidemiologic studies may increase the brain's vulnerability representing an important risk factor in the development and/or deterioration of certain cardio- and neuropathologies. The roles of ALA in neurological disorders remain unclear, especially in stroke that is a leading cause of death. We and others have identified ALA as a potential nutraceutical to protect the brain from stroke, characterized by its pleiotropic effects in neuroprotection, vasodilation of brain arteries, and neuroplasticity. This review highlights how chronic administration of ALA protects against rodent models of hypoxic-ischemic injury and exerts an anti-depressant-like activity, effects that likely involve multiple mechanisms in brain, and may be applied in stroke prevention. One major effect may be through an increase in mature brain-derived neurotrophic factor (BDNF), a widely expressed protein in brain that plays critical roles in neuronal maintenance, and learning and memory. Understanding the precise roles of ALA in neurological disorders will provide the underpinnings for the development of new therapies for patients and families who could be devastated by these disorders.


Subject(s)
Fatty Acids, Omega-3/metabolism , Neuroprotective Agents/metabolism , Stroke/prevention & control , alpha-Linolenic Acid/metabolism , Animals , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Humans , Stroke/metabolism
16.
PLoS One ; 9(7): e102627, 2014.
Article in English | MEDLINE | ID: mdl-25047645

ABSTRACT

Traumatic brain injury (TBI) is a major public health concern affecting a large number of athletes and military personnel. Individuals suffering from a TBI risk developing anxiety disorders, yet the pathophysiological alterations that result in the development of anxiety disorders have not yet been identified. One region often damaged by a TBI is the basolateral amygdala (BLA); hyperactivity within the BLA is associated with increased expression of anxiety and fear, yet the functional alterations that lead to BLA hyperexcitability after TBI have not been identified. We assessed the functional alterations in inhibitory synaptic transmission in the BLA and one mechanism that modulates excitatory synaptic transmission, the α7 containing nicotinic acetylcholine receptor (α7-nAChR), after mTBI, to shed light on the mechanisms that contribute to increased anxiety-like behaviors. Seven and 30 days after a mild controlled cortical impact (CCI) injury, animals displayed significantly greater anxiety-like behavior. This was associated with a significant loss of GABAergic interneurons and significant reductions in the frequency and amplitude of spontaneous and miniature GABAA-receptor mediated inhibitory postsynaptic currents (IPSCs). Decreases in the mIPSC amplitude were associated with reduced surface expression of α1, ß2, and γ2 GABAA receptor subunits. However, significant increases in the surface expression and current mediated by α7-nAChR, were observed, signifying increases in the excitability of principal neurons within the BLA. These results suggest that mTBI causes not only a significant reduction in inhibition in the BLA, but also an increase in neuronal excitability, which may contribute to hyperexcitability and the development of anxiety disorders.


Subject(s)
Anxiety Disorders/etiology , Basolateral Nuclear Complex/physiopathology , Brain Injuries/complications , Receptors, GABA-A/metabolism , Animals , Basolateral Nuclear Complex/metabolism , Brain Injuries/metabolism , Brain Injuries/physiopathology , Inhibitory Postsynaptic Potentials , Interneurons/metabolism , Interneurons/pathology , Male , Rats , Rats, Sprague-Dawley , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism
17.
PLoS One ; 9(6): e99818, 2014.
Article in English | MEDLINE | ID: mdl-24955574

ABSTRACT

Organophosphorus (OP) nerve agents are deadly chemical weapons that pose an alarming threat to military and civilian populations. The irreversible inhibition of the critical cholinergic degradative enzyme acetylcholinesterase (AChE) by OP nerve agents leads to cholinergic crisis. Resulting excessive synaptic acetylcholine levels leads to status epilepticus that, in turn, results in brain damage. Current countermeasures are only modestly effective in protecting against OP-induced brain damage, supporting interest for evaluation of new ones. (-)-Phenserine is a reversible AChE inhibitor possessing neuroprotective and amyloid precursor protein lowering actions that reached Phase III clinical trials for Alzheimer's Disease where it exhibited a wide safety margin. This compound preferentially enters the CNS and has potential to impede soman binding to the active site of AChE to, thereby, serve in a protective capacity. Herein, we demonstrate that (-)-phenserine protects neurons against soman-induced neuronal cell death in rats when administered either as a pretreatment or post-treatment paradigm, improves motoric movement in soman-exposed animals and reduces mortality when given as a pretreatment. Gene expression analysis, undertaken to elucidate mechanism, showed that (-)-phenserine pretreatment increased select neuroprotective genes and reversed a Homer1 expression elevation induced by soman exposure. These studies suggest that (-)-phenserine warrants further evaluation as an OP nerve agent protective strategy.


Subject(s)
Chemical Warfare Agents/toxicity , Cholinesterase Inhibitors/pharmacology , Physostigmine/analogs & derivatives , Soman/toxicity , Status Epilepticus , Animals , Carrier Proteins/biosynthesis , Gene Expression Regulation/drug effects , Homer Scaffolding Proteins , Male , Physostigmine/pharmacology , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Status Epilepticus/metabolism
18.
J Neurotrauma ; 31(7): 683-90, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24344922

ABSTRACT

Mild traumatic brain injury (mTBI) often has long-term effects on cognitive function and social behavior. Altered gene expression may be predictive of long-term psychological effects of mTBI, even when acute clinical effects are minimal or transient. Controlled cortical impact (CCI), which causes concussive, but nonpenetrant, trauma to underlying (non-cortical) brain, resulting in persistent changes in hippocampal synaptic function, was used as a model of mTBI. The hippocampal transcriptomes of sham-operated or injured male rats at 1, 7, and 30 days postinjury were examined using microarrays comprising a comprehensive set of expressed genes, subsequently confirmed by quantitative reverse-transcriptase polymerase chain reaction. Transcripts encoding the chemokines, chemokine (C-C motif) ligand (Ccl)2 and Ccl7, inflammatory mediators lipocalin-2 (Lcn2) and tissue inhibitor of metalloproteinase 1 (Timp1), immunocyte activators C-C chemokine receptor type 5 (Ccr5) and Fc fragment of IgG, low affinity IIb, receptor (CD32) (Fcgr2b), the major histocompatibility complex II immune response-related genes, Cd74 and RT1 class II, locus Da (RT1-Da), the complement component, C3, and the transcription factor, Kruppel-like factor 4 (Klf4), were identified as early (Ccl2, Ccl7, Lcn2, and Timp1), intermediate (Ccr5, Fcgr2b, Cd74, RT1-Da, and C3), and late (Klf4) markers for bilateral hippocampal response to CCI. Ccl2 and Ccl7 transcripts were up-regulated within 24 h after CCI, and their elevation subsided within 1 week of injury. Other transcriptional changes occurred later and were more stable, some persisting for at least 1 month, suggesting that short-term inflammatory responses trigger longer-term alteration in the expression of genes previously associated with injury, aging, and neuronal function in the brain. These transcriptional responses to mTBI may underlie long-term changes in excitatory and inhibitory neuronal imbalance in hippocampus, leading to long-term behavioral consequences of mTBI.


Subject(s)
Brain Concussion/genetics , Brain Concussion/physiopathology , Hippocampus/metabolism , Transcriptome , Animals , Brain Concussion/metabolism , Disease Models, Animal , Hippocampus/physiopathology , Kruppel-Like Factor 4 , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
19.
PLoS One ; 8(1): e54887, 2013.
Article in English | MEDLINE | ID: mdl-23382994

ABSTRACT

Neuronal dysfunction and demise together with a reduction in neurogenesis are cardinal features of Alzheimer's disease (AD) induced by a combination of oxidative stress, toxic amyloid-ß peptide (Aß) and a loss of trophic factor support. Amelioration of these was assessed with the Aß lowering AD experimental drugs (+)-phenserine and (-)-phenserine in neuronal cultures, and actions in mice were evaluated with (+)-phenserine. Both experimental drugs together with the metabolite N1-norphenserine induced neurotrophic actions in human SH-SY5Y cells that were mediated by the protein kinase C (PKC) and extracellular signal-regulated kinases (ERK) pathways, were evident in cells expressing amyloid precursor protein Swedish mutation (APP(SWE)), and retained in the presence of Aß and oxidative stress challenge. (+)-Phenserine, together with its (-) enantiomer as well as its N1- and N8-norphenserine and N1,N8-bisnorphenserine metabolites, likewise provided neuroprotective activity against oxidative stress and glutamate toxicity via the PKC and ERK pathways. These neurotrophic and neuroprotective actions were evident in primary cultures of subventricular zone (SVZ) neural progenitor cells, whose neurosphere size and survival were augmented by (+)-phenserine. Translation of these effects in vivo was assessed in wild type and AD APPswe transgenic (Tg2576) mice by doublecortin (DCX) immunohistochemical analysis of neurogenesis in the SVZ, which was significantly elevated by 16 day systemic (+)-phenserine treatment, in the presence of a (+)-phenserine-induced elevation in brain- derived neurotrophic factor (BDNF).


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Physostigmine/analogs & derivatives , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Doublecortin Protein , Drug Discovery , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Transgenic , Mutation , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Physostigmine/chemistry , Physostigmine/pharmacology , Physostigmine/therapeutic use , Stereoisomerism
20.
Neurotoxicology ; 33(5): 1219-29, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22884490

ABSTRACT

Nerve agents are deadly threats to military and civilian populations around the world. Nerve agents cause toxicity to peripheral and central sites through the irreversible inhibition of acetylcholinesterase, the enzyme that metabolizes acetylcholine. Excessive acetylcholine accumulation in synapses results in status epilepticus in the central nervous system. Prolonged status epilepticus leads to brain damage, neurological dysfunction and poor outcome. Anticonvulsants are effective but must be given rapidly following exposure. Because these agents cause mass casualties, effective neuroprotective agents are needed to reduce brain damage and improve cognitive outcome. α-Linolenic acid is an omega-3 fatty acid that is found in vegetable products and has no known side effects. α-Linolenic acid is neuroprotective against kainic acid-induced brain damage in vivo, but its neuroprotective efficacy against nerve agents is unknown. α-Linolenic acid also exerts anti-depressant and anti-inflammatory activities and enhances synaptic plasticity in vivo. These properties make this polyunsaturated fatty acid (PUFA) a potential candidate against nerve agent-induced neuropathology. Here we show that α-linolenic acid is neuroprotective against soman-induced neuropathology in either a pretreatment or post-treatment paradigm. We also show that subcutaneous injection of α-linolenic acid shows greater neuroprotective efficacy compared with intravenous injection in a brain region-specific manner.


Subject(s)
Cholinesterase Inhibitors/toxicity , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , alpha-Linolenic Acid/therapeutic use , Analysis of Variance , Animals , Brain Injuries/etiology , Brain Injuries/prevention & control , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Electrophoretic Mobility Shift Assay , Fluoresceins , Male , NF-kappa B/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Neurotoxicity Syndromes/complications , Organic Chemicals , Rats , Rats, Sprague-Dawley , Soman/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...