Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 356
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 322(1): 16-22, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17400887

ABSTRACT

Autism is a neurodevelopmental disorder presenting before 3 years of age with deficits in communication and social skills and repetitive behaviors. In addition to genetic influences, recent studies suggest that prenatal drug or chemical exposures are risk factors for autism. Terbutaline, a beta2-adrenoceptor agonist used to arrest preterm labor, has been associated with increased concordance for autism in dizygotic twins. We studied the effects of terbutaline on microglial activation in different brain regions and behavioral outcomes in developing rats. Newborn rats were given terbutaline (10 mg/kg) daily on postnatal days (PN) 2 to 5 or PN 11 to 14 and examined 24 h after the last dose and at PN 30. Immunohistochemical studies showed that administration of terbutaline on PN 2 to 5 produced a robust increase in microglial activation on PN 30 in the cerebral cortex, as well as in cerebellar and cerebrocortical white matter. None of these effects occurred in animals given terbutaline on PN 11 to 14. In behavioral tests, animals treated with terbutaline on PN 2 to 5 showed consistent patterns of hyper-reactivity to novelty and aversive stimuli when assessed in a novel open field, as well as in the acoustic startle response test. Our findings indicate that beta2-adrenoceptor overstimulation during an early critical period results in microglial activation associated with innate neuroinflammatory pathways and behavioral abnormalities, similar to those described in autism. This study provides a useful animal model for understanding the neuropathological processes underlying autism spectrum disorders.


Subject(s)
Adrenergic beta-Agonists/toxicity , Autistic Disorder/chemically induced , Behavior, Animal/drug effects , Brain/drug effects , Microglia/drug effects , Terbutaline/toxicity , Animals , Animals, Newborn , Disease Models, Animal , Female , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-2/physiology , Reflex, Startle/drug effects
2.
Am J Physiol Regul Integr Comp Physiol ; 283(5): R1236-44, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12376418

ABSTRACT

Neonatal beta-adrenoceptors (beta-ARs) are resistant to agonist-induced desensitization. We examined the functioning of G(i) and G(s) after repeated administration of beta-AR agonists to newborn rats. Isoproterenol (beta(1)/beta(2) agonist) obtunded G(i) function in the heart but not the liver; in contrast, terbutaline, a beta(2)-selective agonist, enhanced G(i) function. Isoproterenol, but not terbutaline, increased membrane-associated G((s)alpha), which would enhance receptor function. In addition, isoproterenol increased and terbutaline maintained the proportion of the short-splice (S) variant of G((s)alpha) in the membrane fraction; G((s)alpha)S is functionally more active than the long-splice variant. Either isoproterenol or terbutaline treatment increased G((s)alpha) in the cytosolic fraction, a characteristic usually associated with desensitization in the adult. Decreased G(i) activity, coupled with increased membrane-associated G((s)alpha) concentrations and maintenance or increases in membrane G((s)alpha)S, provide strong evidence that unique effects on G protein function underlie the ability of the immature organism to sustain beta-AR cell signaling in the face of excessive or prolonged stimulation; these mechanisms also contribute to tissue selectivity of the effects of beta-agonists with divergent potencies toward different beta-AR subtypes.


Subject(s)
Animals, Newborn/physiology , GTP-Binding Proteins/physiology , Receptors, Adrenergic, beta/physiology , Adenylyl Cyclases/metabolism , Adrenergic beta-1 Receptor Agonists , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Animals , Blotting, Western , Female , GTP-Binding Protein alpha Subunits, Gs/isolation & purification , GTP-Binding Protein alpha Subunits, Gs/metabolism , Heart/drug effects , Heart/growth & development , Isoproterenol/pharmacology , Liver/drug effects , Liver/growth & development , Liver/metabolism , Male , Myocardium/metabolism , Pertussis Toxin/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-2/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Terbutaline/pharmacology
3.
Brain Res ; 951(2): 280-92, 2002 Oct 04.
Article in English | MEDLINE | ID: mdl-12270507

ABSTRACT

Nicotine is a neuroteratogen that targets synaptic function during critical developmental stages and recent studies indicate that CNS vulnerability extends into adolescence, the age at which smoking typically commences. We administered nicotine to adolescent rats via continuous minipump infusions from PN30 to PN47.5, using 6 mg/kg/day, a dose rate that replicates the plasma nicotine levels found in smokers, and examined 5HT receptors and related cell signaling during nicotine administration (PN45) and in the post-treatment period (PN50, 60, 75). Adolescent nicotine decreased 5HT(2) receptor binding in brain regions containing 5HT projections (hippocampus and cerebral cortex), with selectivity for females in the cerebral cortex; regions containing 5HT cell bodies showed either an increase (midbrain in males) or no change (brainstem). In contrast, there were no significant changes in 5HT(1A) receptors; however, the ability of the receptors to signal through adenylyl cyclase (AC) showed a switch from stimulatory to inhibitory effects in females during the post-treatment period. There were also transient alterations in AC responses to beta-adrenergic receptor stimulation, as well as pronounced induction of the AC response to the non-receptor-mediated stimulant, forskolin. Our results indicate that adolescent nicotine exposure alters the concentrations and functions of postsynaptic 5HT receptors in a manner commensurate with impaired 5HT synaptic function. The direction of change, emergence of defects after the cessation of nicotine administration, and sex-preference for effects in females, all support a relationship of impaired 5HT function to the higher incidence of depression seen in adolescent smokers.


Subject(s)
Adenylyl Cyclases/physiology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Serotonin/drug effects , Signal Transduction/drug effects , Adrenergic beta-Agonists/pharmacology , Aging/physiology , Animals , Brain Chemistry/drug effects , Colforsin/pharmacology , Female , GTP-Binding Proteins/metabolism , Isoproterenol/pharmacology , Male , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Serotonin/biosynthesis , Receptors, Serotonin/metabolism , Receptors, Serotonin, 5-HT1 , Serotonin/metabolism , Substance Withdrawal Syndrome/enzymology , Substance Withdrawal Syndrome/physiopathology , Up-Regulation/drug effects
4.
Am J Physiol Regul Integr Comp Physiol ; 283(1): R205-17, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12069947

ABSTRACT

During fetal and neonatal development, beta-adrenergic receptors (beta-ARs) appear to be resistant to desensitization by beta-agonist drugs. To determine the mechanisms underlying the regulatory differences between adults and neonates, we administered isoproterenol, a mixed beta(1)/beta(2)-AR agonist, and terbutaline, a beta(2)-selective agonist. Effects were examined in the ensuing 4 h after a single injection, or after the last of four daily injections. We prepared cell membranes from heart (predominantly beta(1)-ARs) and liver (predominantly beta(2)-ARs) and assessed signal transduction in the adenylyl cyclase (AC) pathway. In the first few hours after a single administration of isoproterenol to adult rats, cardiac beta-ARs showed activation of G proteins (elevated AC response to forskolin) and desensitization of beta-AR-mediated responses; after the fourth injection, heterologous desensitization emerged, characterized by a loss of signaling mediated either through beta-ARs or glucagon receptors. Terbutaline evoked an increase in the forskolin response but no desensitization of receptor-mediated responses. When we gave the same treatments to neonatal rats, we observed cardiac G protein activation, but there was neither homologous nor heterologous desensitization of beta-ARs or glucagon receptors. In the adult liver, isoproterenol and terbutaline both failed to evoke desensitization, regardless of whether the drugs were given once or for 4 days. In neonates, however, acute or chronic treatment elicited homologous desensitization of beta-AR-mediated AC signaling, while sensitizing the response to glucagon. These results show that neonatal beta-ARs are inherently capable of desensitization in some, but not all, cell types; cellular responses can be maintained through heterologous sensitization of signaling proteins downstream from the receptor. Differences from adult patterns of response are highly tissue selective and are likely to depend on ontogenetic differences in subtypes of beta-ARs and AC.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Animals, Newborn/physiology , Heart/drug effects , Isoproterenol/pharmacology , Liver/drug effects , Receptors, Adrenergic, beta/drug effects , Terbutaline/pharmacology , Adenylyl Cyclases/metabolism , Animals , Female , Liver/metabolism , Male , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/physiology , Sex Characteristics , Time Factors
5.
Am J Physiol Regul Integr Comp Physiol ; 282(5): R1356-63, 2002 May.
Article in English | MEDLINE | ID: mdl-11959676

ABSTRACT

Imbalances of beta-adrenoceptor (beta-AR) and muscarinic ACh receptor (mAChR) input are thought to underlie perinatal cardiovascular abnormalities in conditions such as sudden infant death syndrome. Administration of isoproterenol, a beta(1)/beta(2)-AR agonist, to neonatal rats on postnatal days (PN) 2-5 caused downregulation of cardiac m(2)AChRs and a corresponding decrement in their control of adenylyl cyclase activity. Terbutaline, a beta(2)-selective agonist that crosses the placenta and the blood-brain barrier, was also effective when given either on PN 2-5 or during gestational days 17-20. Terbutaline failed to downregulate brain m(2)AChRs, even though it downregulated beta-ARs; beta-ARs and m(2)AChRs are located on different cell populations in the brain, but they are on the same cells in the heart. Destruction of catecholaminergic neurons with neonatal 6-hydroxydopamine upregulated cardiac but not brain m(2)AChRs. These results suggest that perinatal beta-AR stimulation shifts cardiac receptor production away from the generation of m(2)AChRs so that the development of sympathetic innervation acts as a negative modulator of cholinergic function. Accordingly, tocolytic therapy with beta-AR agonists may compromise the perinatal balance of adrenergic and cholinergic inputs.


Subject(s)
Animals, Newborn/metabolism , Myocardium/metabolism , Receptor Cross-Talk , Receptors, Adrenergic, beta/physiology , Receptors, Muscarinic/physiology , Adenylyl Cyclases/metabolism , Adrenergic Agents/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Brain/metabolism , Down-Regulation , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Heart/embryology , Isoproterenol/pharmacology , Oxidopamine/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M2 , Receptors, Adrenergic, beta/drug effects , Receptors, Muscarinic/drug effects , Terbutaline/pharmacology
6.
Brain Res Dev Brain Res ; 133(2): 163-73, 2002 Feb 28.
Article in English | MEDLINE | ID: mdl-11882346

ABSTRACT

Chlorpyrifos (CPF), one of the most widely used pesticides, is a neurobehavioral teratogen in animals. We administered CPF to neonatal rats on postnatal days (PN) 1-4 (1 mg/kg) or PN11-14 (5 mg/kg), regimens devoid of overt systemic toxicity. We then examined the impact on catecholaminergic systems in adolescence (PN30) and adulthood (PN60), assessing basal neurotransmitter content and transmitter utilization rates (turnover) in brain regions comprising the major noradrenergic and dopaminergic projections. Although CPF had only sporadic effects on basal norepinephrine and dopamine content, it profoundly suppressed norepinephrine turnover across multiple regions, indicative of net reductions in presynaptic activity. Dopamine turnover showed less consistent effects, with subnormal turnover in some regions and activation in others. We also evaluated whether CPF exposure altered the ability of catecholamine systems to respond to acute cholinergic stimulation, elicited by administration of a single challenge dose of nicotine. In the normal brain, nicotine increases the utilization of norepinephrine and dopamine. With only a few exceptions, animals receiving neonatal CPF exposure showed lasting desensitization of the nicotine response; not only was the activation by nicotine blunted in the CPF group, but in some regions the nicotine response was reversed, eliciting a reduction in transmitter turnover. These results indicate that neonatal CPF exposure produces widespread deficiencies in catecholaminergic synaptic function that persist into adulthood, and that are best revealed by dynamic measures of synaptic activity and responsiveness, as opposed to static markers like basal transmitter levels. The effects seen here are likely to contribute to alterations in behavioral performance that persist or emerge long after the termination of CPF exposure.


Subject(s)
Aging/drug effects , Brain/drug effects , Catecholamines/metabolism , Chlorpyrifos/toxicity , Insecticides/toxicity , Neural Pathways/drug effects , Neurons/drug effects , Prenatal Exposure Delayed Effects , Aging/metabolism , Animals , Animals, Newborn/metabolism , Brain/growth & development , Brain/metabolism , Dopamine/metabolism , Dose-Response Relationship, Drug , Female , Male , Neural Pathways/growth & development , Neural Pathways/metabolism , Neurons/metabolism , Neurotoxins/toxicity , Nicotine/pharmacology , Norepinephrine/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Sex Factors , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
7.
Brain Res Dev Brain Res ; 133(2): 175-9, 2002 Feb 28.
Article in English | MEDLINE | ID: mdl-11882347

ABSTRACT

In humans, perinatal exposure to environmental tobacco smoke (ETS) is associated with neurobehavioral deficits. In the current study, we exposed Rhesus monkeys to ETS in late gestation and in the early neonatal period, and examined changes in neurotransmitter receptors in the brainstem and caudal portion of the cerebral cortex. Nicotinic acetylcholine receptors were markedly upregulated and the effect was selective in that there were no changes in m(2)-muscarinic acetylcholine receptors or in beta-adrenergic receptors. Nicotinic receptor upregulation is indicative of chronic cell stimulation by nicotine, and is a hallmark of nicotine-induced neuroteratogenesis. These results indicate that perinatal ETS exposes the fetus and neonate to quantities of nicotine that are sufficient to alter brain development.


Subject(s)
Brain/drug effects , Environmental Exposure , Macaca mulatta/embryology , Neurons/drug effects , Nicotiana/toxicity , Prenatal Exposure Delayed Effects , Receptors, Nicotinic/drug effects , Tobacco Smoke Pollution/adverse effects , Up-Regulation/drug effects , Animals , Brain/embryology , Brain/growth & development , Cell Membrane/drug effects , Cell Membrane/metabolism , Female , Macaca mulatta/growth & development , Macaca mulatta/metabolism , Neurons/metabolism , Pregnancy , Radioligand Assay , Receptor, Muscarinic M2 , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/metabolism , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Receptors, Nicotinic/metabolism , Up-Regulation/physiology
8.
Brain Res Dev Brain Res ; 132(1): 69-79, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11744108

ABSTRACT

Prenatal heroin exposure evokes neurochemical and behavioral deficits that in part, reflect disruption of septohippocampal cholinergic function. In earlier studies, we found that cholinergic synaptic defects involve changes in proteins, like protein kinase C, that are essential to receptor-mediated signaling. In the current study, we determined whether heroin targets another signaling protein, adenylyl cyclase (AC), which regulates the production of cAMP. Mice exposed to prenatal heroin showed subsequent postnatal elevations of AC activity that lasted into adulthood. The effect was most robust with stimulants that activate AC directly (forskolin, Mn(2+)), indicating increased expression of AC itself; we also identified shifts in catalytic properties suggestive of a change in the AC isoform. Superimposed on the overall induction of AC, there were transient deficits in the responses to stimulants working through G-proteins (NaF) or G-protein coupled receptors (isoproterenol, a beta-adrenoceptor agonist), indicating alterations at other steps in the signaling pathway. Effects on the regulation of AC activity were seen in brain regions with widely disparate maturational timetables and also occurred in regions, like the cerebellum, that are sparse in cholinergic input. These results suggest that the expression and/or function of signaling proteins distal to neurotransmitter receptors represent a major target for neurobehavioral teratogenesis by heroin; the fact that these targets are shared by otherwise unrelated neuroteratogens may account for a common set of neurochemical and behavioral anomalies in response to prenatal exposure to drugs or environmental chemicals.


Subject(s)
Adenylyl Cyclases/metabolism , Analgesics, Opioid/pharmacology , Brain/embryology , Brain/enzymology , Heroin/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Cerebellum/embryology , Cerebellum/enzymology , Cerebral Cortex/embryology , Cerebral Cortex/enzymology , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Hippocampus/embryology , Hippocampus/enzymology , Isoproterenol/pharmacology , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects , Teratogens/pharmacology
9.
Am J Physiol Regul Integr Comp Physiol ; 281(6): R1895-901, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11705775

ABSTRACT

Terbutaline, a beta(2)-adrenoceptor (beta(2)-AR) agonist, is a widely used tocolytic that also crosses the placenta to stimulate fetal beta-ARs. The current study examines the effects of terbutaline administered to neonatal rats. Terbutaline (10 mg/kg sc) given on postnatal day (PN) 2-5 or PN 11-14 elicited significant downregulation of both cardiac and hepatic beta-ARs, with a much greater effect in the liver. Despite the reduction in cardiac beta-ARs, receptor desensitization was absent as evidenced by the maintained ability of isoproterenol to stimulate adenylyl cyclase (AC) in membrane preparations. The underlying mechanism was dissected by using stimulants that operate at different points in the AC signaling pathway, NaF, forskolin, and Mn(2+). When administered in the early neonatal period, terbutaline failed to evoke any changes in cardiac AC activity; however, treatment on PN 11-14 evoked heterologous sensitization downstream from the receptor, evidenced by increases in the response to NaF and forskolin. In the liver, neonatal terbutaline administration elicited a small (approximately equal to 10%) decrease in the AC response to isoproterenol, an effect much smaller than the downregulation of beta-ARs (>40%). In this tissue, desensitization was again offset by heterologous sensitization of AC signaling. These results indicate that, in the developing organism, beta-AR-mediated cell signaling responses are maintained in the face of receptor downregulation through heterologous induction of downstream signaling elements. These unique responses serve to sustain beta-AR signaling in the perinatal period.


Subject(s)
Heart/physiology , Receptors, Adrenergic, beta/physiology , Signal Transduction/physiology , Terbutaline/pharmacology , Animals , Animals, Newborn , Body Weight/drug effects , Clonidine/pharmacology , Colforsin/pharmacology , Down-Regulation , Female , GTP-Binding Proteins/metabolism , Heart/drug effects , Isoproterenol/pharmacology , Liver/drug effects , Liver/physiology , Male , Membrane Proteins/metabolism , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/drug effects , Sodium Fluoride/pharmacology
10.
Brain Res Dev Brain Res ; 131(1-2): 113-25, 2001 Nov 26.
Article in English | MEDLINE | ID: mdl-11718842

ABSTRACT

Beta(2)-adrenoceptor agonists are commonly used to arrest preterm labor but they also penetrate the placenta to stimulate fetal beta-adrenergic receptors (betaAR), and have been implicated in subsequent neurobehavioral deficits. We administered terbutaline to pregnant rats on gestational days (GD) 17-20 and during two postnatal (PN) periods, PN2-5 and PN11-14, that correspond to third trimester human neurological development. We then examined betaAR binding sites and adenylyl cyclase (AC) signaling in fetal brain or neonatal brain regions. Although fetal terbutaline administration evoked betaAR downregulation, the ability of isoproterenol to stimulate AC was enhanced instead of desensitized. Sensitization occurred at post-receptor signaling proteins, as augmented responses were also seen for stimulants that bypass the receptors to work on G-proteins (NaF) or that stimulate AC directly (forskolin and Mn(2+)). When terbutaline was given on PN2-5, betaAR downregulation was obtained in brainstem, forebrain and cerebellum, but desensitization of the AC response was seen only in the forebrain; the desensitization was heterologous, reflecting decrements in total AC activity rather than specific loss of the betaAR response. With treatment on PN11-14, only the cerebellum showed betaAR downregulation and induction at the level of post-receptor signaling proteins maintained the betaAR-mediated AC response. Our results indicate that, unlike the adult, betaAR signaling in the fetus and neonate is resistant to homologous desensitization by beta-agonists, and in fact, displays heterologous sensitization that sustains or enhances the overall response. The inability to desensitize betaAR responses may lead to disruption of neural cell development as a consequence of tocolytic therapy.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Brain/growth & development , Brain/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction/drug effects , Terbutaline/pharmacology , Adaptation, Physiological/drug effects , Adenylyl Cyclases/metabolism , Animals , Brain/drug effects , Cyclic AMP/metabolism , Down-Regulation/drug effects , Female , Homeostasis/drug effects , Homeostasis/physiology , Male , Organ Size/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Tocolytic Agents/pharmacology
11.
Brain Res ; 914(1-2): 166-78, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11578609

ABSTRACT

Nicotine is a neuroteratogen that targets synaptic function during critical developmental stages and recent studies indicate that CNS vulnerability extends into adolescence, the time that smoking typically commences. We administered nicotine to pregnant or adolescent rats via continuous minipump infusions, using dose rates that replicate the plasma nicotine levels found in smokers. Fetal nicotine exposure (gestational days 4-21) decreased the cerebrocortical binding of paroxetine (PXT), a marker for the serotonin (5HT) transporter, likely indicative of a decrease in nerve terminals in that region; the effect lasted into adulthood. There was a corresponding increase in PXT binding in the midbrain/brainstem, the region containing the 5HT cell bodies that project to the cerebral cortex, a pattern typical of reactive sprouting in response to nerve terminal damage. After adolescent nicotine treatment (postnatal days 30-47), PXT binding was reduced in the hippocampus and striatum instead of the cerebral cortex, again accompanied by increased binding in the midbrain and brainstem; the patterns of effects within each region were gender-selective, although both males and females displayed abnormalities. Superimposed on this overall effect, there were transient increases in PXT binding, likely due to acute stimulant effects of nicotine. We also assessed 5HT presynaptic activity (5HIAA/5HT ratio). Withdrawal from adolescent nicotine treatment led to suppression of activity in the cerebral cortex and activation in the midbrain. These results indicate that both fetal and adolescent nicotine exposure elicit apparent damage to 5HT projections with reactive increases in regions containing 5HT cell bodies. Long-term changes in 5HT innervation and/or synaptic activity may play a role in the subsequent development of depression in the offspring of women who smoke during pregnancy or in adolescent smokers.


Subject(s)
Brain/drug effects , Membrane Transport Proteins , Nerve Degeneration/chemically induced , Nerve Tissue Proteins , Nicotine/toxicity , Prenatal Exposure Delayed Effects , Presynaptic Terminals/drug effects , Serotonin/metabolism , Tobacco Use Disorder/metabolism , Aging/drug effects , Aging/physiology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Brain/abnormalities , Brain/growth & development , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Hydroxyindoleacetic Acid/metabolism , Male , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Paroxetine/pharmacokinetics , Pregnancy , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Serotonin Plasma Membrane Transport Proteins , Sex Characteristics , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tobacco Use Disorder/pathology , Tobacco Use Disorder/physiopathology
12.
Brain Res Dev Brain Res ; 130(2): 249-52, 2001 Oct 24.
Article in English | MEDLINE | ID: mdl-11675126

ABSTRACT

Neural input plays a key role in the establishment of immune function, and environmental agents or drugs that interfere with the development of the nervous system elicit corresponding immunologic deficits. In the current study, we gave neonatal rats the widely used organophosphate pesticide, chlorpyrifos (CPF), and determined the immediate and long-term effects on T-lymphocyte function. Exposure of neonatal rats to 1 mg/kg of CPF daily on postnatal days (PN) 1-4 had no immediate effect (PN5) on T-cell mitogenic responses to concanavalin A challenge. However, once the animals reached adulthood, T-cell responses were significantly impaired. There were no deficits in basal T-cell replication rates, implying that the adverse effect of CPF exposure was specific to mitogenic activation. Treatment during a later neonatal period (PN11-14) elicited similar deficits in adulthood. CPF administration leads to inhibition of cholinesterase, and a cholinergic connection is supported by the fact that the results seen here correspond to those seen with a direct cholinergic stimulant (nicotine) administered during gestation or adolescence. These results indicate that exposure to CPF during a developmental period in which this organophosphate pesticide is known to produce lasting changes in neural function, elicits corresponding, long-term deficits in immune competence.


Subject(s)
Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Immune System/drug effects , Nervous System/drug effects , Neuroimmunomodulation/drug effects , Age Factors , Animals , Animals, Newborn , Cell Division/drug effects , Cell Division/immunology , Concanavalin A/pharmacology , Female , Immune System/cytology , Immune System/growth & development , Mitogens/pharmacology , Nervous System/growth & development , Nervous System/immunology , Pregnancy , Rats , Rats, Sprague-Dawley , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
13.
Brain Res Dev Brain Res ; 130(2): 253-6, 2001 Oct 24.
Article in English | MEDLINE | ID: mdl-11675127

ABSTRACT

Maternal cigarette smoking during pregnancy is known to alter immune function in the offspring and recent studies with animals indicate that prenatal nicotine exposure leads to lasting deficiencies in T-lymphocyte mitogenic responses, likely through excessive cholinergic stimulation during a critical stage of development. The current study was conducted to determine if the vulnerable period for nicotine-induced mis-programming of immune responses extends into adolescence, the stage at which most smokers begin tobacco use. Adolescent rats were given nicotine via osmotic minipump infusions on postnatal days (PN) 30-47.5, using a regimen that produces plasma levels (25 ng/ml) of nicotine similar to those in smokers or in users of transdermal nicotine patches. Toward the end of the infusion period (PN45) and 1 month after termination of nicotine exposure (PN80), we examined the mitogenic responses of splenocytes to Concanavalin A. Although no deficiencies were seen on PN45, there were robust decreases in mitogenic responses on PN80, with deficits apparent at both suboptimal and optimal concentrations of Concanavalin A. These results indicate that the adolescent immune system is vulnerable to nicotine-induced disruption of T-cell function.


Subject(s)
Immune System/drug effects , Immune System/growth & development , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Sexual Maturation/immunology , Age Factors , Animals , Concanavalin A/pharmacology , Female , Infusion Pumps, Implantable , Male , Mitogens/pharmacology , Rats , Rats, Sprague-Dawley , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
14.
Environ Health Perspect ; 109(9): 909-13, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11673119

ABSTRACT

The widely used organophosphate pesticide chlorpyrifos is a suspected neuroteratogen. In the current study, we compared the effects of chlorpyrifos and its major metabolites in two in vitro models, neuronotypic PC12 cells and gliotypic C6 cells. Chlorpyrifos inhibited DNA synthesis in both cell lines but had a greater effect on gliotypic cells. Chlorpyrifos oxon, the active metabolite that inhibits cholinesterase, also decreased DNA synthesis in PC12 and C6 cells with a preferential effect on the latter. Trichloropyridinol, the major catabolic product of chlorpyrifos, had a much smaller, but nevertheless statistically significant, effect that was equivalent in both cell lines. Diazinon, another organophosphate pesticide, also inhibited DNA synthesis with preference toward C6 cells, but was less effective than was chlorpyrifos. Physostigmine, a non-organophosphate cholinesterase inhibitor, was less effective than either chlorpyrifos or diazinon, but still caused significant inhibition of DNA synthesis in C6 cells. We also found that the addition of sera protected the cells from the adverse effects of chlorpyrifos and that the effect could be reproduced by addition of albumin. These results indicate that chlorpyrifos and other organophosphates such as diazinon have immediate, direct effects on neural cell replication, preferentially for gliotypic cells. In light of the protective effect of serum proteins, the fact that the fetus and newborn possess lower concentrations of these proteins suggests that greater neurotoxic effects may occur at blood levels of chlorpyrifos that are nontoxic to adults.


Subject(s)
Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , DNA/biosynthesis , Insecticides/toxicity , Models, Biological , Animals , Diazinon/toxicity , Neuroglia , PC12 Cells , Rats
15.
Brain Res Dev Brain Res ; 130(1): 65-72, 2001 Sep 23.
Article in English | MEDLINE | ID: mdl-11557094

ABSTRACT

Chlorpyrifos (CPF), one of the most widely-used organophosphate pesticides, is a suspected neuroteratogen. We administered CPF to neonatal rats on postnatal days (PN) 1-4 (1 mg/kg) or PN11-14 (5 mg/kg), treatments devoid of overt toxicity. At the end of the treatment period (PN5 and 15, respectively) and 5-7 days later, we then examined the effects on paroxetine (PXT) binding to the presynaptic 5HT high-affinity transporter, a marker for serotonin (5HT) projections. In males, we found a persistent decrease in PXT binding across the two different treatment regimens, with deficits apparent in a brain region containing 5HT terminal fields (forebrain) as well as in a region containing 5HT cell bodies (brainstem). In contrast, females given the early treatment regimen (PN1-4) showed deficits in the brainstem but transient elevations in the forebrain; the later treatment regimen (PN11-14) had no significant effect on PXT binding in females. These data are consistent with earlier work showing brainstem cell injury resulting from neonatal CPF exposure, and indicate specific damage to 5HT neurons, with a consequent loss of transporter expression in both terminal fields and perikarya. In females, the damage may be temporarily offset by initial trophic effects in the terminal region, consequent to the cholinergic stimulation evoked by cholinesterase inhibition via the active metabolite, CPF oxon. The gender-selective effects on 5HT systems are likely to contribute to similar gender dimorphism in behavioral performance. Because the CPF effects involve 5HT, a neurotransmitter intimately involved in the control of mood, we suggest the need to evaluate behaviors that typify animal models of depression.


Subject(s)
Brain Chemistry/drug effects , Carrier Proteins/biosynthesis , Chlorpyrifos/pharmacology , Cholinesterase Inhibitors/pharmacology , Membrane Glycoproteins/biosynthesis , Membrane Transport Proteins , Nerve Tissue Proteins , Animals , Animals, Newborn , Carrier Proteins/metabolism , Female , Male , Membrane Glycoproteins/metabolism , Paroxetine/metabolism , Paroxetine/pharmacology , Pregnancy , Rats , Serotonin Plasma Membrane Transport Proteins , Selective Serotonin Reuptake Inhibitors/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Tritium
16.
Brain Res Dev Brain Res ; 130(1): 83-9, 2001 Sep 23.
Article in English | MEDLINE | ID: mdl-11557096

ABSTRACT

Chlorpyrifos (CPF) is a widely used insecticides which has been shown to alter brain cell development. The current project was conducted to determine whether there are persistent behavioral effects of early [1 mg/kg/day postnatal days (PNDs) 1-4] or late (5 mg/kg/day PNDs 11-14) postnatal CPF exposure in rats. We tested spontaneous alternation in a T-maze, locomotor activity in the Figure-8 apparatus and learning in the 16-arm radial maze, throughout adolescence and into adulthood. Exposure during either neonatal period elicited significant long-term effects on cognitive behavior. In the radial-arm maze, as has been seen previously, control male performed more accurately than control females. Early postnatal CPF exposure reversed this effect. With exposure on PNDs 1-4, females in the CPF group showed a reduction in working and reference memory errors in the radial maze, reducing their error rate to that seen in control males; in contrast, CPF-exposed males exhibited an increase in errors during the initial stages of training. When animals were exposed on PNDs 11-14 and then tested in adolescence and adulthood, males showed a significant slowing of response latency in the T-maze and the rate of habituation in the Figure-8 apparatus was slowed in both sexes. When females were challenged acutely with the muscarinic antagonist, scopolamine, they did not show reference memory impairment, whereas controls did; these results suggest that adaptations occur after CPF exposure that lead to loss of muscarinic cholinergic control of reference memory. No such changes were seen with a nicotinic cholinergic antagonist (mecamylamine). These results indicate that early neonatal exposure to CPF induces long-term changes in cognitive performance that, in keeping with the neurochemical changes seen previously, are distinctly gender-selective. Additional defects may be revealed by similar strategies that subject the animals to acute challenges, thus uncovering the adaptive mechanisms that maintain basal performance.


Subject(s)
Behavior, Animal/drug effects , Chlorpyrifos/pharmacology , Insecticides/pharmacology , Maze Learning/drug effects , Animals , Animals, Newborn , Female , Male , Mecamylamine/pharmacology , Memory/drug effects , Motor Activity/drug effects , Muscarinic Antagonists/pharmacology , Nicotinic Antagonists/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley , Scopolamine/pharmacology
17.
Am J Physiol Regul Integr Comp Physiol ; 281(4): R1079-89, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11557613

ABSTRACT

Terbutaline (Ter), a beta(2)-adrenergic agonist used in preterm labor, stimulates fetal beta-adrenoceptors (beta-ARs). We administered Ter to pregnant rats on gestational days 17-20 and examined beta-ARs and adenylyl cyclase (AC) signaling in heart and liver. Ter produced less downregulation of cardiac beta-ARs than in adults, despite a higher proportion of the beta(2)-subtype, and failed to elicit desensitization of the receptor-mediated AC response. AC stimulants acting at different points indicated an offsetting of homologous desensitization at the level of the beta-AR by heterologous sensitization at the level of AC: induction of total AC catalytic activity and a shift in the catalytic profile or AC isoform. In fetal liver, Ter produced downregulation of beta-ARs, in keeping with the predominance of the beta(2)-subtype; hepatic receptor downregulation was equivalent in fetus and adult. Nevertheless, there was still no desensitization of beta-AR-mediated AC responses and again AC was induced. Our results indicate that, unlike in the adult, fetal beta-AR signaling is not desensitized by beta-agonists and, in fact, displays heterologous sensitization, thus sustaining responses during parturition. At the same time, the inability to desensitize beta-AR AC responses may lead to disruption of cardiac, hepatic, or neural cell development as a consequence of tocolytic therapy with beta-agonists.


Subject(s)
Adenylyl Cyclases/metabolism , Fetus/metabolism , Liver/metabolism , Myocardium/metabolism , Receptors, Adrenergic, beta/metabolism , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Down-Regulation/drug effects , Enzyme Induction/drug effects , Female , Fetal Weight/drug effects , Fetus/drug effects , GTP-Binding Proteins/metabolism , Male , Maternal-Fetal Exchange/physiology , Organ Size/drug effects , Pregnancy , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Terbutaline/pharmacology
18.
Environ Health Perspect ; 109(7): 651-61, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11485862

ABSTRACT

Chlorpyrifos targets mammalian brain development through a combination of effects directed at cholinergic receptors and intracellular signaling cascades that are involved in cell differentiation. We used sea urchin embryos as an invertebrate model system to explore the cellular mechanisms underlying the actions of chlorpyrifos and to delineate the critical period of developmental vulnerability. Sea urchin embryos and larvae were exposed to chlorpyrifos at different stages of development ranging from early cell cleavages through the prism stage. Although early cleavages were unaffected even at high chlorpyrifos concentrations, micromolar concentrations added at the mid-blastula stage evoked a prominent change in cell phenotype and overall larval structure, with appearance of pigmented cells followed by their accumulation in an extralarval cap that was extruded from the animal pole. At higher concentrations (20-40 microM), these abnormal cells constituted over 90% of the total cell number. Studies with cholinergic receptor blocking agents and protein kinase C inhibitors indicated two distinct types of effects, one mediated through stimulation of nicotinic cholinergic receptors and the other targeting intracellular signaling. The effects of chlorpyrifos were not mimicked by chlorpyrifos oxon, the active metabolite that inhibits cholinesterase, nor by nonorganophosphate cholinesterase inhibitors. Dieldrin, an organochlorine that targets GABA(A )receptors, was similarly ineffective. The effects of chlorpyrifos and its underlying cholinergic and signaling-related mechanisms parallel prior findings in mammalian embryonic central nervous system. Invertebrate test systems may thus provide both a screening procedure for potential neuroteratogenesis by organophosphate-related compounds, as well as a system with which to uncover novel mechanisms underlying developmental vulnerability.


Subject(s)
Brain/drug effects , Chlorpyrifos/toxicity , Dieldrin/toxicity , Insecticides/toxicity , Sea Urchins/embryology , Animals , Brain/growth & development , Dose-Response Relationship, Drug , Embryo, Nonmammalian/drug effects , Embryonic Development , Mammals/growth & development , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology
19.
Breast Cancer Res Treat ; 66(2): 143-6, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11437100

ABSTRACT

Theophylline reduces cell number in MDA-MB-231 cells through mechanisms over and above phosphodiesterase inhibition. In the current study, we used an intracellular fluorescent dye to show that theophylline and, to a much greater extent, 3-isobutyl-1-methylxanthine, evoke the generation of reactive oxygen species and also sensitize the cells to insult by other oxidants. Xanthine derivatives may therefore offer novel strategies for antitumor therapeutics.


Subject(s)
1-Methyl-3-isobutylxanthine/pharmacology , Breast Neoplasms/pathology , Phosphodiesterase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Theophylline/pharmacology , Female , Humans , Tumor Cells, Cultured/drug effects
20.
Brain Res ; 902(2): 229-43, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11384617

ABSTRACT

The commonly-used organophosphate insecticide, chlorpyrifos (CPF), impairs brain cell development, axonogenesis and synaptogenesis. In the current study, we administered CPF to neonatal rats on postnatal (PN) days 1-4 (1 mg/kg) or PN11-14 (5 mg/kg), treatments that were devoid of overt toxicity. We then examined two cholinergic synaptic markers, choline acetyltransferase activity (ChAT) and [3H]hemicholinium-3 binding (HC-3) in the hippocampus, midbrain, striatum, brainstem and cerebral cortex in the juvenile (PN30) and young adult (PN60). Across all brain regions, CPF exposure evoked significant reductions in both markers, with larger effects on HC-3 binding, which is responsive to neuronal impulse activity, than on ChAT, a constitutive marker. Superimposed on the deficits, there were gender-selective effects and distinct regional disparities in the critical exposure period for vulnerability. In the hippocampus, either the early or late treatment regimen evoked decreases in ChAT but the early regimen elicited a much larger decrease in HC-3; effects persisted into adulthood. In the midbrain, CPF administration on PN1-4 elicited deficits similar to those seen in the hippocampus; however, exposure on PN11-14 elicited changes preferentially in females. Gender selectivity was also apparent in the striatum, in this case reflecting deficits in females after CPF treatment on PN1-4. In contrast, the effects of CPF on the brainstem were relatively more robust in males; effects in the cerebral cortex were less notable than in other regions. These results indicate that neonatal CPF exposure produces widespread deficiencies in cholinergic synaptic function that persist into adulthood. The effects are likely to contribute to gender-selective alterations in behavioral performance that persist or emerge long after the termination of exposure and well after the restoration of cholinesterase activity.


Subject(s)
Acetylcholine/metabolism , Animals, Newborn/growth & development , Brain/drug effects , Chlorpyrifos/toxicity , Cholinergic Fibers/drug effects , Environmental Exposure/adverse effects , Insecticides/toxicity , Presynaptic Terminals/drug effects , Symporters , Age Factors , Animals , Animals, Newborn/metabolism , Biomarkers/analysis , Brain/growth & development , Brain/metabolism , Brain Stem/drug effects , Brain Stem/growth & development , Brain Stem/metabolism , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/metabolism , Dose-Response Relationship, Drug , Hemicholinium 3/pharmacokinetics , Hippocampus/drug effects , Hippocampus/growth & development , Hippocampus/metabolism , Mesencephalon/drug effects , Mesencephalon/growth & development , Mesencephalon/metabolism , Neostriatum/drug effects , Neostriatum/growth & development , Neostriatum/metabolism , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Plasma Membrane Neurotransmitter Transport Proteins , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Sex Factors , Tritium/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...