Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 292: 179-192, 2017 06.
Article in English | MEDLINE | ID: mdl-28341460

ABSTRACT

A neurochemical target at which cannabinoids interact to have global effects on behavior is brain noradrenergic circuitry. Acute and repeated administration of a cannabinoid receptor synthetic agonist is capable of increasing multiple indices of noradrenergic activity. This includes cannabinoid-induced 1) increases in norepinephrine (NE) release in the medial prefrontal cortex (mPFC); 2) desensitization of cortical α2-adrenoceptor-mediated effects; 3) activation of c-Fos in brainstem locus coeruleus (LC) noradrenergic neurons; and 4) increases in anxiety-like behaviors. In the present study, we sought to examine adaptations in adrenoceptor expression and function under conditions of cannabinoid receptor type 1 (CB1r) deletion using knockout (KO) mice and compare these to wild type (WT) controls. Electrophysiological analysis of α2-adrenoceptor-mediated responses in mPFC slices in WT mice showed a clonidine-induced α2-adrenoceptor-mediated increase in mPFC cell excitability coupled with an increase in input resistance. In contrast, CB1r KO mice showed an α2-adrenoceptor-mediated decrease in mPFC cell excitability. We then examined protein expression levels of α2- and ß1-adrenoceptor subtypes in the mPFC as well as TH expression in the locus coeruleus (LC) of mice deficient in CB1r. Both α2- and ß1-adrenoceptors exhibited a significant decrease in expression levels in CB1r KO mice when compared to WT in the mPFC, while a significant increase in TH was observed in the LC. To better define whether the same cortical neurons express α2A-adrenoceptor and CB1r in mPFC, we utilized high-resolution immunoelectron microscopy. We localized α2A-adrenoceptors in a knock-in mouse that expressed a hemoagglutinin (HA) tag downstream of the α2A-adrenoceptor promoter. Although the α2A-adrenoceptor was often identified pre-synaptically, we observed co-localization of CB1r with α2-adrenoceptors post-synaptically in the same mPFC neurons. Finally, using receptor binding, we confirmed prior results showing that α2A-adrenoceptor is unchanged in mPFC following acute or chronic exposure to the synthetic cannabinoid receptor agonist, WIN 55,212-2, but is increased, following chronic treatment followed by a period of abstinence. Taken together, these data provide convergent lines of evidence indicating cannabinoid regulation of the cortical adrenergic system.


Subject(s)
Locus Coeruleus/drug effects , Neurons/drug effects , Prefrontal Cortex/drug effects , Receptors, Cannabinoid/metabolism , Animals , Benzoxazines/pharmacology , Brain/drug effects , Brain/metabolism , Cannabinoids/pharmacology , Locus Coeruleus/metabolism , Male , Mice, Knockout , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/metabolism , Norepinephrine/metabolism , Prefrontal Cortex/metabolism , Receptors, Cannabinoid/deficiency , Synapses/drug effects , Synapses/metabolism
2.
Exp Neurol ; 236(2): 327-35, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22677142

ABSTRACT

The cannabinoid receptor agonist, WIN 55,212-2, increases extracellular norepinephrine levels in the rat frontal cortex under basal conditions, likely via desensitization of inhibitory α2-adrenergic receptors located on norepinephrine terminals. Here, the effect of WIN 55,212-2 on stress-induced norepinephrine release was assessed in the medial prefrontal cortex (mPFC), in adult male Sprague-Dawley rats using in vivo microdialysis. Systemic administration of WIN 55,212-2 30 min prior to stressor exposure prevented stress-induced cortical norepinephrine release induced by a single exposure to swim when compared to vehicle. To further probe cortical cannabinoid-adrenergic interactions, postsynaptic α2-adrenergic receptor (AR)-mediated responses were assessed in mPFC pyramidal neurons using electrophysiological analysis in an in vitro cortical slice preparation. We confirm prior studies showing that clonidine increases cortical pyramidal cell excitability and that this was unaffected by exposure to acute stress. WIN 55,212-2, via bath application, blocked postsynaptic α2-AR mediated responses in cortical neurons irrespective of exposure to stress. Interestingly, stress exposure prevented the desensitization of α2-AR mediated responses produced by a history of cannabinoid exposure. Together, these data indicate the stress-dependent nature of cannabinoid interactions via both pre- and postsynaptic ARs. In summary, microdialysis data indicate that cannabinoids restrain stress-induced cortical NE efflux. Electrophysiology data indicate that cannabinoids also restrain cortical cell excitability under basal conditions; however, stress interferes with these CB1-α2 AR interactions, potentially contributing to over-activation of pyramidal neurons in mPFC. Overall, cannabinoids are protective of the NE system and cortical excitability but stress can derail this protective effect, potentially contributing to stress-related psychopathology. These data add to the growing evidence of complex, stress-dependent modulation of monoaminergic systems by cannabinoids and support the potential use of cannabinoids in the treatment of stress-induced noradrenergic dysfunction.


Subject(s)
Benzoxazines/administration & dosage , Cannabinoids/administration & dosage , Morpholines/administration & dosage , Naphthalenes/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Receptors, Adrenergic, alpha-2/physiology , Stress, Psychological/physiopathology , Animals , Cannabinoids/toxicity , Male , Organ Culture Techniques , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/metabolism , Stress, Psychological/psychology , Swimming/psychology
3.
Neuroscience ; 216: 143-57, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22542679

ABSTRACT

The locus coeruleus (LC) is the major loci of noradrenergic innervation to the forebrain. Due to the extensive central nervous system innervation of the LC noradrenergic system, a reduction in the number of LC neurons could result in significant changes in noradrenergic function in many forebrain regions. LC noradrenergic neurons were lesioned in adult male C57Bl/6 mice with the unilateral administration of 6-hydroxydopamine (6OHDA) (vehicle on the alternate side). Noradrenergic markers were measured 3 weeks later to determine the consequence of LC loss in the forebrain. Direct administration of 6OHDA into the LC results in the specific reduction of noradrenergic neurons in the LC (as measured by electrophysiology, immunoreactivity and in situ hybridization), the lateral tegmental neurons and dopaminergic neurons in the substantia nigra (SN) and ventral tegmental region were unaffected. The loss of LC noradrenergic neurons did not result in compensatory changes in the expression of mRNA for norepinephrine (NE)-synthesizing enzymes. The loss of LC noradrenergic neurons is associated with reduced NE tissue concentration and NE transporter (NET) binding sites in the frontal cortex and hippocampus, as well as other forebrain regions such as the amygdala and SN. Adrenoreceptor (AR) binding sites (α(1)- and α(2)-AR) were not significantly affected on the 6OHDA-treated side compared to the vehicle-treated side, although there is a reduction of AR binding sites on both the vehicle- and 6OHDA-treated side in specific forebrain regions. These studies indicate that unilateral stereotaxic injection of 6OHDA into mice reduces noradrenergic LC neurons and reduces noradrenergic innervation to many forebrain regions, including the contralateral side.


Subject(s)
Locus Coeruleus/metabolism , Oxidopamine/toxicity , Signal Transduction/drug effects , Adrenergic Agents/toxicity , Adrenergic Neurons/drug effects , Adrenergic Neurons/metabolism , Animals , Behavior, Animal , Brain Mapping , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Electrophysiological Phenomena , Hippocampus/metabolism , Locus Coeruleus/drug effects , Male , Mice , Mice, Inbred C57BL , Norepinephrine/metabolism , RNA, Messenger/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
4.
Neuroscience ; 166(1): 279-91, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20045445

ABSTRACT

Degeneration of the noradrenergic neurons in the locus coeruleus (LC) is a major component of Alzheimer's (AD) and Parkinson's disease (PD), but the consequence of noradrenergic neuronal loss has different effects on the surviving neurons in the two disorders. Therefore, understanding the consequence of noradrenergic neuronal loss is important in determining the role of this neurotransmitter in these neurodegenerative disorders. The goal of the study was to determine if the neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4) could be used as a model for either (or both) AD or PD. Rats were administered DSP4 and sacrificed 3 days 2 weeks and 3 months later. DSP4-treatment resulted in a rapid, though transient reduction in norepinephrine (NE) and NE transporter (NET) in many brain regions receiving variable innervation from the LC. Alpha(1)-adrenoreceptors binding site concentrations were unchanged in all brain regions at all three time points. However, an increase in alpha(2)-AR was observed in many different brain regions 2 weeks and 3 months after DSP4. These changes observed in forebrain regions occurred without a loss in LC noradrenergic neurons. Expression of synthesizing enzymes or NET did not change in amount of expression/neuron despite the reduction in NE tissue content and NET binding site concentrations at early time points, suggesting no compensatory response. In addition, DSP4 did not affect basal activity of LC at any time point in anesthetized animals, but 2 weeks after DSP4 there is a significant increase in irregular firing of noradrenergic neurons. These data indicate that DSP4 is not a selective LC noradrenergic neurotoxin, but does affect noradrenergic neuron terminals locally, as evident by the changes in transmitter and markers at terminal regions. However, since DSP4 did not result in a loss of noradrenergic neurons, it is not considered an adequate model for noradrenergic neuronal loss observed in AD and PD.


Subject(s)
Benzylamines/toxicity , Locus Coeruleus/drug effects , Locus Coeruleus/pathology , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Norepinephrine/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Locus Coeruleus/metabolism , Male , Nerve Degeneration/metabolism , Neurotoxins/toxicity , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/metabolism , Time , Time Factors
5.
Neuroscience ; 146(1): 471-80, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17324522

ABSTRACT

In Alzheimer's disease (AD) there is a significant loss of locus coeruleus (LC) noradrenergic neurons. However, recent work has shown the surviving noradrenergic neurons to display many compensatory changes, including axonal sprouting to the hippocampus. The prefrontal cortex (PFC) is a forebrain region that is affected in dementia, and receives innervation from the LC noradrenergic neurons. Reduced PFC function can reduce cognition and disrupt behavior. Because the PFC is an important area in AD, we determined if noradrenergic innervation from the LC noradrenergic neurons is maintained and if adrenoreceptors are altered postsynaptically. Presynaptic PFC alpha2-adrenoreceptor (AR) binding site density, as determined by 3H-RX821002, suggests that axons from surviving noradrenergic neurons in the LC are sprouting to the PFC of subjects with dementia. Changes in postsynaptic alpha1-AR in the PFC of subjects with dementia indicate normal to elevated levels of binding sites. Expression of alpha1-AR subtypes (alpha1A- and alpha1D-AR) and alpha2C-AR subtype mRNA in the PFC of subjects with dementia is similar to what was observed in the hippocampus with one exception, the expression of alpha1A-AR mRNA. The expression of the alpha1A-AR mRNA subtype is significantly reduced in specific layers of the PFC in subjects with dementia. The loss of alpha1A-, alpha1D- and alpha2C-AR mRNA subtype expression in the PFC may be attributed to neuronal loss observed in dementia. These changes in postsynaptic AR would suggest a reduced function of the PFC. Consequence of this reduced function of the PFC in dementia is still unknown but it may affect memory and behavior.


Subject(s)
Dementia/pathology , Gene Expression Regulation/physiology , Prefrontal Cortex/metabolism , Receptors, Adrenergic/metabolism , Adrenergic alpha-Antagonists/pharmacokinetics , Adult , Aged , Aged, 80 and over , Female , Humans , Idazoxan/analogs & derivatives , Idazoxan/pharmacokinetics , Male , Middle Aged , Postmortem Changes , Prazosin/pharmacokinetics , Prefrontal Cortex/pathology , RNA, Messenger/metabolism , Receptors, Adrenergic/classification , Receptors, Adrenergic/genetics
6.
Neuroscience ; 126(3): 795-803, 2004.
Article in English | MEDLINE | ID: mdl-15183527

ABSTRACT

The alpha2-adrenoreceptor (AR) is the most investigated noradrenergic receptor with regard to modulation of seizure activity. However, because of the complexity of multiple alpha2-AR subtypes and their distribution, the exact role of this receptor in modulating seizure activity is not clear. alpha2A- and alpha2C-ARs function as both autoreceptors (presynaptic) on noradrenergic neurons, where they regulate norepinephrine (NE) release, and as postsynaptic receptors on neurons that receive noradrenergic innervation, where they regulate the release of other neurotransmitters (heteroreceptor). The nonselective alpha2-AR agonist clonidine produced a proconvulsant effect on seizure susceptibility, while the selective alpha2A-AR agonist guanfacine was anticonvulsant. The effects of both alpha2-AR agonists were absent in alpha2a knockout mice, suggesting that the alpha2A-AR mediates the proconvulsant and anticonvulsant effect of alpha2-AR agonists on seizure susceptibility. To determine whether the alpha2-AR agonists were acting on inhibitory presynaptic autoreceptors to decrease NE release or on postsynaptic receptors on NE target neurons, the effects of clonidine and guanfacine were determined in dopamine beta-hydroxylase knockout (Dbh -/-) mice that lack NE. The anticonvulsant effect of guanfacine persisted in Dbh -/- mice, suggesting that guanfacine may act preferentially on alpha2A-postsynaptic receptors that regulate the action of NE on target neurons. In contrast, the proconvulsant effect of clonidine was lost in Dbh -/- mice, suggesting that clonidine may act on presynaptic autoreceptors to decrease NE release. We hypothesize that the alpha2A-presynaptic autoreceptor is responsible for the proconvulsant effect of alpha2-AR agonists, while the alpha2A-postsynaptic receptor is responsible for the anticonvulsant effect of alpha2-AR agonists. These data help to clarify the inconsistent effects of alpha2-AR agonists on seizure activity.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Anticonvulsants/pharmacology , Convulsants/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Animals , Autoreceptors/drug effects , Autoreceptors/metabolism , Clonidine/pharmacology , Guanfacine/pharmacology , Mice , Mice, Knockout , Norepinephrine/deficiency , Norepinephrine/metabolism , Pentylenetetrazole/pharmacology , Receptors, Adrenergic, alpha-2/deficiency , Receptors, Adrenergic, alpha-2/drug effects , Seizures/chemically induced , Seizures/physiopathology
7.
Neuroscience ; 126(1): 241-6, 2004.
Article in English | MEDLINE | ID: mdl-15145089

ABSTRACT

The effects of chronic elevations in circulating glucocorticoids on the expression of peptides and peptide receptors of the hypothalamic-pituitary-adrenal (HPA) axis have been studied extensively in rodents, but they have not been examined in primates. To determine the responses of the HPA axis in primates to elevated cortisol, hypothalamic and pituitary tissue from normal older pigtailed macaques (Macaca nemestrina) that had received daily oral administration of cortisol or placebo for 1 year were studied. Pro-opiomelanocortin in the anterior pituitary and corticotropin-releasing factor (CRF) mRNA expression in the hypothalamic paraventricular nucleus (PVN) were significantly reduced in cortisol-treated monkeys in comparison with controls. CRF receptor 1 (CRF-R1) expression in the anterior pituitary and arginine vasopressin mRNA expression in the PVN were unchanged by chronic cortisol administration. Sustained elevation of circulating glucocorticoids results in suppression of HPA peptide and peptide receptor expression in the PVN and anterior pituitary similar to those found in rodents. Chronic therapeutic administration of glucocorticoids in humans may have unintended consequences for hypothalamic and pituitary function.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hydrocortisone/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiology , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/physiology , Animals , Arginine Vasopressin/genetics , Corticotropin-Releasing Hormone/genetics , Female , Gene Expression/drug effects , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Macaca nemestrina , Male , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Receptors, Corticotropin-Releasing Hormone/genetics
8.
J Neurosci ; 21(19): 7764-9, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567066

ABSTRACT

Epilepsy is a disease of neuronal hyperexcitability, and pharmacological and genetic studies have identified norepinephrine (NE) and neuropeptide Y (NPY) as important endogenous regulators of neuronal excitability. Both transmitters signal through G-protein-coupled receptors, are expressed either together or separately, and are abundant in brain regions implicated in seizure generation. NPY knock-out (NPY KO) and dopamine beta-hydroxylase knock-out (DBH KO) mice that lack NE are susceptible to seizures, and agonists of NE and NPY receptors protect against seizures. To examine the relative contributions of NE and NPY to neuronal excitability, we tested Dbh;Npy double knock-out (DKO) mice for seizure sensitivity. In general, DBH KO mice were much more seizure-sensitive than NPY KO mice and had normal NPY expression, demonstrating that an NPY deficiency did not contribute to the DBH KO seizure phenotype. DKO mice were only slightly more sensitive than DBH KO mice to seizures induced by kainic acid, pentylenetetrazole, or flurothyl, although DKO mice were uniquely prone to handling-induced seizures. NPY contributed to the seizure phenotype of DKO mice at high doses of convulsant agents and advanced stages of seizures. These data suggest that NE is a more potent endogenous anticonvulsant than NPY, and that NPY has the greatest contribution under conditions of extreme neuronal excitability.


Subject(s)
Genetic Predisposition to Disease , Neuropeptide Y/metabolism , Norepinephrine/metabolism , Seizures/physiopathology , Animals , Dopamine beta-Hydroxylase/deficiency , Dopamine beta-Hydroxylase/genetics , Exercise Test , Flurothyl , Handling, Psychological , In Situ Hybridization , Kainic Acid , Male , Mice , Mice, Knockout , Neuropeptide Y/deficiency , Neuropeptide Y/pharmacology , Norepinephrine/deficiency , Norepinephrine/pharmacology , Pentylenetetrazole , Phenotype , Seizures/chemically induced , Seizures/prevention & control
9.
J Pharmacol Exp Ther ; 298(3): 1042-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11504801

ABSTRACT

It has been known for many years that norepinephrine (NE) is a potent endogenous anticonvulsant, yet there is confusion as to which receptor(s) mediate this effect. This is probably due to multiple factors, including the importance of distinct signaling pathways for different seizure paradigms, a lack of comprehensive pharmacological studies, and difficulty in interpreting existing pharmacological results due to the presence of endogenous NE. We sought to circumvent these problems by testing the anticonvulsant activity of selective agonists for most known adrenoreceptors (ARs) in dopamine beta-hydroxylase knockout (Dbh -/-) mice that lack endogenous NE. Dbh -/- mice are hypersensitive to pentylenetetrazole (PTZ)-induced seizures, demonstrating that endogenous NE inhibits PTZ-induced seizures in the wild type. Pretreatment of Dbh -/- mice with an alpha(1)AR or beta(2)AR, but not an alpha(2)AR or beta(1)AR agonist significantly protected against PTZ-induced seizures. In contrast, only the beta(2)AR agonist showed anticonvulsant activity in heterozygous controls. Furthermore, an alpha(1)AR antagonist exacerbated PTZ-induced seizures in control mice, whereas a beta(2)AR antagonist had no effect. We conclude that activation of the alpha(1)AR is primarily responsible for the anticonvulsant activity of endogenous NE in the murine PTZ model of epilepsy. Endogenous NE probably does not activate the beta(2)AR under these conditions, but exogenous activation of the beta(2)AR produces an anticonvulsant effect.


Subject(s)
Adrenergic alpha-1 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Anticonvulsants/pharmacology , Norepinephrine/physiology , Seizures/prevention & control , Animals , Autoreceptors/drug effects , Convulsants , Mice , Mice, Knockout , Norepinephrine/deficiency , Norepinephrine/genetics , Pentylenetetrazole , Seizures/chemically induced
10.
Brain Res Dev Brain Res ; 129(2): 211-4, 2001 Aug 23.
Article in English | MEDLINE | ID: mdl-11506865

ABSTRACT

Ketogenic diet (KD) is a high fat, low carbohydrate diet used to treat children with epilepsy that are refractory to conventional antiepileptic drugs (AEDs). The anticonvulsant mechanism of the KD is unknown. To determine if the noradrenergic system has a role in mediating the anticonvulsant action of the KD, dopamine beta-hydroxylase knockout (Dbh -/-) mice that lack norepinephrine (NE) and Dbh +/- littermates that have normal NE content were fed either a standard rodent chow or the KD. When exposed to the convulsant flurothyl, Dbh +/- mice fed the KD had significantly longer latencies to myoclonic jerk (MJ) and generalized clonic-tonic (CT) seizures than Dbh +/- mice fed normal chow. In contrast, Dbh -/- mice fed the KD had seizure latencies to both MJ and CT comparable to Dbh -/- mice fed normal chow. These results suggest that an intact, functional noradrenergic nervous system is required for the KD to exert an anticonvulsant effect.


Subject(s)
Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Epilepsy/diet therapy , Ketone Bodies/biosynthesis , Norepinephrine/physiology , Animals , Convulsants , Dietary Carbohydrates/therapeutic use , Dietary Fats/therapeutic use , Dopamine beta-Hydroxylase/deficiency , Dopamine beta-Hydroxylase/genetics , Dopamine beta-Hydroxylase/metabolism , Epilepsy/chemically induced , Flurothyl , Mice , Mice, Knockout/genetics , Reaction Time/physiology
11.
Epilepsy Res ; 46(3): 191-203, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11518622

ABSTRACT

Seizure incidence varies significantly with age, with seizure susceptibility particularly high during the first few years of life. Of significant concern is what effects do brief, repetitive seizures have on the developing brain. We approached this issue by examining the change in seizure threshold, and related markers of neuronal activity and metabolic activity (c-fos mRNA and 2-deoxyglucose [2DG]), as a function of repetitive seizure episodes in immature and mature rats. Starting on postnatal day 15 (P15) (immature) or P60 (adult) rats were given two flurothyl seizures a day for 5 days (nine or ten seizures). The seizure latency profile, our measure of threshold, in immature versus adult rats across the 5-day testing period was different. In immature rats, threshold for the second seizure on each day was significantly lower than for the first seizure, suggesting that there was little refractoriness after the first seizure of the day. In contrast, the mature animal had a significantly longer threshold latency to the second seizure for the first 3 days of testing. The immature animal was also more likely than the adult to exhibit tonic extension as a feature of the first seizure of the day. Following repetitive seizures, more regions of the CNS showed c-fos mRNA expression in the immature animal than adults, suggesting that repetitive seizures in the immature animal activated a greater percentage of the brain. Compared with the effects of a single seizure, repetitive seizures resulted in less 2DG labeling in most regions of the brain (except the hippocampus); in the immature brain this difference was more distinct than in adults. The consequences of repetitive seizures in the immature animal results in distinctly different seizure behavior and neuronal activity pattern (c-fos expression) than that observed in the mature animal.


Subject(s)
Aging/metabolism , Behavior, Animal/physiology , Seizures/metabolism , Aging/physiology , Animals , Animals, Newborn , Antimetabolites/pharmacokinetics , Brain/metabolism , Deoxyglucose/pharmacokinetics , Female , Male , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Seizures/physiopathology
12.
Brain Res Mol Brain Res ; 84(1-2): 135-40, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11113540

ABSTRACT

Despite the loss of locus coeruleus (LC) noradrenergic neurons in Alzheimer's disease (AD), cerebrospinal fluid norepinephrine (NE) levels are normal or increased in AD. This paradox suggests compensatory upregulation of NE synthetic capacity or downregulation of the NE transporter (NET) in the remaining LC neurons. LC tyrosine hydroxylase (TH) mRNA expression in the LC was measured in AD subjects (n=5) and in age and gender comparable non-demented subjects (n=6). When AD subjects were divided into those still ambulatory prior to death (CDR 3/4) and those in a prolonged 'vegetative' state prior to death (CDR 5), differences among groups became apparent at specific levels of the LC. In CDR 3/4 AD subjects there was increased TH mRNA expression per neuron compared to non-demented subjects in the caudal half of the LC. However, expression of NET mRNA in the same subjects was not significantly different at any level of the LC. These preliminary results suggest an upregulation of NE biosynthetic capacity in at least some LC neurons in AD prior to the very late stage of the disease.


Subject(s)
Alzheimer Disease/genetics , Carrier Proteins/genetics , Gene Expression , Locus Coeruleus/metabolism , Locus Coeruleus/pathology , Symporters , Tyrosine 3-Monooxygenase/genetics , Aged , Aged, 80 and over , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Female , Humans , In Situ Hybridization , Male , Neurons/metabolism , Neurons/pathology , Norepinephrine Plasma Membrane Transport Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Epilepsia ; 41 Suppl 6: S70-5, 2000.
Article in English | MEDLINE | ID: mdl-10999523

ABSTRACT

PURPOSE/METHODS: Seizures in early life are thought to contribute to the development of human temporal lobe epilepsy. To examine the consequences of early seizures, we elicited status epilepticus in immature, 5.5- to 7.0-month-old pigtailed macaques by unilateral microinfusion of bicuculline methiodide into the entorhinal cortex. RESULTS: This report focuses on neuropathological changes in the hippocampus. Bicuculline infusion consistently elicited limbic-like seizures with prolonged, relatively localized electrographic activity. Magnetic resonance imaging revealed enhanced signal intensity in the ipsilateral hippocampus after seizures; in some cases, there was also progressive hippocampal atrophy. Histological changes were variable; in two of five monkeys, there was significant hippocampal neuron loss, gliosis, granule cell dispersion, and mossy fiber reorganization. CONCLUSIONS: The histopathological findings and associated magnetic resonance imaging abnormalities after bicuculline-induced status epilepticus in infant monkeys mimic common aspects of human temporal lobe epilepsy.


Subject(s)
Bicuculline/analogs & derivatives , Disease Models, Animal , Epilepsy, Temporal Lobe/pathology , Hippocampus/pathology , Neuronal Plasticity/physiology , Animals , Animals, Newborn/growth & development , Bicuculline/pharmacology , Entorhinal Cortex/drug effects , Entorhinal Cortex/pathology , Hippocampus/drug effects , Macaca nemestrina , Magnetic Resonance Imaging , Mossy Fibers, Hippocampal/pathology , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Temporal Lobe/pathology
14.
Brain Res Mol Brain Res ; 79(1-2): 163-8, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10925155

ABSTRACT

We examined 5-HT(7) receptor mRNA expression with in situ hybridization histochemistry in the brains of young (3 months), middle-aged (12 months) and old rats (24 months). In the ventral CA3 area of the hippocampus 5-HT(7) mRNA expression is reduced by approximately 30% between young and middle age without further decline between middle and old age. In other brain areas 5-HT(7) mRNA expression is unaffected by age.


Subject(s)
Aging/genetics , Brain/metabolism , Receptors, Serotonin/genetics , Transcription, Genetic , Animals , Brain/growth & development , Male , Organ Specificity , Pyramidal Cells/physiology , RNA, Messenger/genetics , Rats , Rats, Inbred BN , Receptors, Serotonin/analysis , Receptors, Serotonin/metabolism
15.
J Neurosci ; 19(24): 10985-92, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10594079

ABSTRACT

Several lines of evidence suggest that norepinephrine (NE) can modulate seizure activity. However, the experimental methods used in the past cannot exclude the possible role of other neurotransmitters coreleased with NE from noradrenergic terminals. We have assessed the seizure susceptibility of genetically engineered mice that lack NE. Seizure susceptibility was determined in the dopamine beta-hydroxylase null mutant (Dbh -/-) mouse using four different convulsant stimuli: 2,2,2-trifluroethyl ether (flurothyl), pentylenetetrazol (PTZ), kainic acid, and high-decibel sound. Dbh -/- mice demonstrated enhanced susceptibility (i.e., lower threshold) compared with littermate heterozygous (Dbh +/-) controls to flurothyl, PTZ, kainic acid, and audiogenic seizures and enhanced sensitivity (i.e., seizure severity and mortality) to flurothyl, PTZ, and kainic acid. c-Fos mRNA expression in the cortex, hippocampus (CA1 and CA3), and amygdala was increased in Dbh -/- mice in association with flurothyl-induced seizures. Enhanced seizure susceptibility to flurothyl and increased seizure-induced c-fos mRNA expression were reversed by pretreatment with L-threo-3, 4-dihydroxyphenylserine, which partially restores the NE content in Dbh -/- mice. These genetically engineered mice confirm unambiguously the potent effects of the noradrenergic system in modulating epileptogenicity and illustrate the unique opportunity offered by Dbh -/- mice for elucidating the pathways through which NE can regulate seizure activity.


Subject(s)
Norepinephrine/deficiency , Seizures/chemically induced , Seizures/etiology , Acoustic Stimulation , Animals , Convulsants , Disease Susceptibility , Flurothyl , Mice , Mice, Knockout/genetics , Norepinephrine/genetics , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/metabolism
16.
Alcohol Clin Exp Res ; 23(10): 1643-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10549997

ABSTRACT

BACKGROUND: Prenatal ethanol exposure affects brain dopaminergic neuronal systems, and many of these alterations are permanent. METHODS: The primary objective of this study was to determine the effects of prenatal ethanol exposure on adult mRNA expression for two key regulatory proteins in the mesolimbic and nigrostriatal dopaminergic cell groups which mediate behavioral responses to alcohol and other drugs of abuse: tyrosine hydroxylase (TH) and dopamine transporter (DAT) in the substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA). To also address the effects on noradrenergic regulation, we quantitated mRNA expression for TH and norepinephrine transporter (NET) in the noradrenergic loci of the locus coeruleus (LC). RESULTS: Daily dietary ethanol consumption by female Sprague-Dawley rats for 3 weeks before, and continuing throughout, pregnancy decreased both DAT (approximately 68%,p < 0.002) and TH (approximately 45%,p < 0.002) mRNA expression in the VTA of adult male offspring. This prenatal exposure also suppressed DAT mRNA expression in the SNpc (approximately 81 %;p < 0.03), although TH mRNA expression in this region was not significantly altered. Prenatal ethanol exposure did not alter significantly either TH or NET mRNA expression in the LC of adult male offspring, which suggests that this brain catecholaminergic response may be limited to DA neurons. CONCLUSION: These results demonstrated that prenatal maternal ethanol consumption suppresses mRNA expression for important regulatory proteins in the mesolimbic and nigrostriatal dopaminergic systems of adult male rat offspring. These persistent prenatal ethanol-induced changes in mRNA expression may thus contribute to the persistent effects of fetal ethanol exposure on the diverse behavioral and/or metabolic responses mediated by the mesolimbic and nigrostriatal dopaminergic systems in the adult.


Subject(s)
Carrier Proteins/drug effects , Central Nervous System Depressants/administration & dosage , Dopamine/biosynthesis , Ethanol/administration & dosage , Gene Expression Regulation/drug effects , Membrane Glycoproteins , Membrane Transport Proteins , Nerve Tissue Proteins , Prenatal Exposure Delayed Effects , Tyrosine 3-Monooxygenase/drug effects , Animals , Carrier Proteins/metabolism , Dopamine Plasma Membrane Transport Proteins , Female , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Mesencephalon/drug effects , Mesencephalon/metabolism , Norepinephrine/metabolism , Pregnancy , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
17.
Dev Neurosci ; 21(3-5): 320-7, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10575255

ABSTRACT

Potassium channels play a critical role in limiting neuronal excitability. Mutations in certain voltage-gated potassium channels have been associated with hyperexcitable phenotypes in both humans and animals. However, only recently have mutations in potassium channel genes (i.e. KCNQ2 and KCNQ3) been discovered in a human epilepsy, benign familial neonatal convulsions. Recently, it has been reported that mice lacking the voltage-gated Shaker-like potassium channel Kv1.1 alpha-subunit develop recurrent spontaneous seizures early in postnatal development. The clinical relevance of the Kv1.1 knockout mouse has been underscored by a recent report of epilepsy occurring in a family affected by mutations in the KCNA1 locus (the human homologue of Kv1.1) which typically cause episodic ataxia and myokymia. Here we summarize preliminary studies characterizing the developmental changes in seizure susceptibility and neuronal activation in the three genotypes of Kv1.1 mice (-/-, +/-, +/+). Using behavioral and immediate-early gene indicators of regional brain excitability, we have found that a seizure-sensitive predisposition exists in Kv1.1 -/- animals at a very young age (P10), before either spontaneous seizure activity or changes in c-fos mRNA expression can be demonstrated. Kv1.1 +/- mice, although behaviorally indistinguishable from wild types, also have an increased susceptibility to seizures at a similar early age. The Kv1. 1 knockout mouse possesses many features desirable in a developmental animal epilepsy model and represents a clinically relevant model of early-onset epilepsies.


Subject(s)
Animals, Newborn/growth & development , Mice, Knockout/genetics , Mice, Knockout/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Seizures/etiology , Animals , Disease Susceptibility , Kv1.1 Potassium Channel , Mice , Mice, Knockout/growth & development
18.
Dev Neurosci ; 21(3-5): 352-64, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10575259

ABSTRACT

Seizures, particularly multiple episodes and/or status epilepticus (SE) are prevalent in pediatric patients. Pediatric SE is associated with brain changes that have been hypothesized to contribute to the onset of temporal lobe epilepsy (TLE). In order to gain insight into the effects of seizures on the immature brain and the risk for later TLE, we have developed a model of limbic SE in the pigtailed macaque monkey. In separate studies, bicuculline methiodide or a bicuculline 'cocktail' was infused into three regions of the brain (area tempestas, hippocampus, entorhinal cortex) to induce seizures. Measures included MRI, electrophysiology, behavior and morphology. Our results suggest that monkey models of SE may provide useful tools for understanding the effects of prolonged seizures during infancy and the origins of TLE in humans.


Subject(s)
Status Epilepticus/physiopathology , Animals , Animals, Newborn , Behavior, Animal , Bicuculline/administration & dosage , Bicuculline/analogs & derivatives , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Electric Stimulation/methods , Electroencephalography , Electrophysiology , Entorhinal Cortex/pathology , Entorhinal Cortex/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Injections , Macaca nemestrina , Magnetic Resonance Imaging , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Status Epilepticus/psychology
19.
Horm Metab Res ; 31(5): 335-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10422731

ABSTRACT

Aminergic signaling in the CNS is terminated by clearance from the synapse via high-affinity transporter molecules in the presynaptic membrane. Relatively recent sequence identification of these molecules has now permitted the initiation of studies of regulation of transporter function at the cellular and systems levels. In vitro studies provide evidence that the transporters for dopamine, serotonin, and gamma-aminobutyric acid are substrates for regulation by protein kinase C signaling. In vivo studies provide evidence that insulin and adrenal and gonadal steroid hormones may regulate the synthesis and activity of the transporters. Future directions should permit evaluation of the role of endocrine regulation in neurotransmitter clearance, and thus in the maintenance of normal CNS aminergic signaling.


Subject(s)
Homeostasis , Hormones/physiology , Receptors, Neurotransmitter/physiology , Animals , Central Nervous System Diseases , Humans , Receptors, Neurotransmitter/drug effects , Signal Transduction
20.
Brain Res ; 826(1): 143-7, 1999 Apr 24.
Article in English | MEDLINE | ID: mdl-10216207

ABSTRACT

In normal aging, cell loss occurs in the locus coeruleus (LC), the major noradrenergic nucleus in the brain. This study examined changes in the LC of aged rats by measuring mRNA expression for tyrosine hydroxylase (TH) and the norepinephrine uptake transporter (NET). TH and NET mRNA expression were measured by in situ hybridization in young, middle-aged and aged rats. It appears that in middle age, the transporter system responds initially to LC cell loss by decreasing NET mRNA expression. Then, with further aging and cell loss, TH mRNA expression increases which may potentially increase NE synthesis in the remaining neurons. These findings suggest that multiple regulatory components are used to maintain stable noradrenergic synaptic levels despite neuronal loss. Published by Elsevier Science B.V.


Subject(s)
Aging/genetics , Carrier Proteins/genetics , Gene Expression Regulation, Enzymologic/physiology , Locus Coeruleus/metabolism , Symporters , Tyrosine 3-Monooxygenase/genetics , Animals , Brain Chemistry/physiology , In Situ Hybridization , Locus Coeruleus/chemistry , Locus Coeruleus/cytology , Neurons/chemistry , Neurons/enzymology , Norepinephrine Plasma Membrane Transport Proteins , Oligonucleotide Probes , Phosphorus Radioisotopes , RNA, Messenger/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...