Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Mol Pharmacol ; 98(4): 445-453, 2020 10.
Article in English | MEDLINE | ID: mdl-32198209

ABSTRACT

Millions of Americans suffer from opiate use disorder, and over 100 die every day from opioid overdoses. Opioid use often progresses into a vicious cycle of abuse and withdrawal, resulting in very high rates of relapse. Although the physical and psychologic symptoms of opiate withdrawal are well-documented, sleep disturbances caused by chronic opioid exposure and withdrawal are less well-understood. These substances can significantly disrupt sleep acutely and in the long term. Yet poor sleep may influence opiate use, suggesting a bidirectional feed-forward interaction between poor sleep and opioid use. The neurobiology of how opioids affect sleep and how disrupted sleep affects opioid use is not well-understood. Here, we will summarize what is known about the effects of opioids on electroencephalographic sleep in humans and in animal models. We then discuss the neurobiology interface between reward-related brain regions that mediate arousal and wakefulness as well as the effect of opioids in sleep-related brain regions and neurotransmitter systems. Finally, we summarize what is known of the mechanisms underlying opioid exposure and sleep. A critical review of such studies, as well as recommendations of studies that evaluate the impact of manipulating sleep during withdrawal, will further our understanding of the cyclical feedback between sleep and opioid use. SIGNIFICANCE STATEMENT: We review recent studies on the mechanisms linking opioids and sleep. Opioids affect sleep, and sleep affects opioid use; however, the biology underlying this relationship is not understood. This review compiles recent studies in this area that fill this gap in knowledge.


Subject(s)
Analgesics, Opioid/adverse effects , Opioid-Related Disorders/physiopathology , Sleep/drug effects , Analgesics, Opioid/pharmacology , Animals , Brain/drug effects , Brain/physiopathology , Electroencephalography , Humans , Sleep/physiology
2.
Genes Brain Behav ; 18(7): e12493, 2019 09.
Article in English | MEDLINE | ID: mdl-29896789

ABSTRACT

Adolescent stress can impact health and well-being not only during adulthood of the exposed individual but even in future generations. To investigate the molecular mechanisms underlying these long-term effects, we exposed adolescent males to stress and measured anxiety behaviors and gene expression in the amygdala-a critical region in the control of emotional states-in their progeny for two generations, offspring and grandoffspring. Male C57BL/6 mice underwent chronic unpredictable stress (CUS) for 2 weeks during adolescence and were used to produce two generations of offspring. Male and female offspring and grandoffspring were tested in behavioral assays to measure affective behavior and stress reactivity. Remarkably, transgenerational inheritance of paternal stress exposure produced a protective phenotype in the male, but not the female lineage. RNA-seq analysis of the amygdala from male offspring and grandoffspring identified differentially expressed genes (DEGs) in mice derived from fathers exposed to CUS. The DEGSs clustered into numerous pathways, and the "notch signaling" pathway was the most significantly altered in male grandoffspring. Therefore, we show that paternal stress exposure impacts future generations which manifest in behavioral changes and molecular adaptations.


Subject(s)
Amygdala/metabolism , Stress, Psychological/genetics , Transcriptome , Amygdala/growth & development , Animals , Epigenesis, Genetic , Male , Mice , Mice, Inbred C57BL , Paternal Inheritance , Phenotype
3.
Mol Psychiatry ; 19(7): 801-10, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23999525

ABSTRACT

Addiction to nicotine and the ability to quit smoking are influenced by genetic factors. We used functional genomic approaches (chromatin immunoprecipitation (ChIP) and whole-genome sequencing) to identify cAMP response element-binding protein (CREB) targets following chronic nicotine administration and withdrawal (WD) in rodents. We found that chronic nicotine and WD differentially modulate CREB binding to the gene for neuregulin 3 (NRG3). Quantitative analysis of saline, nicotine and nicotine WD in two biological replicates corroborate this finding, with NRG3 increases in both mRNA and protein following WD from chronic nicotine treatment. To translate these data for human relevance, single-nucleotide polymorphisms (SNPs) across NRG3 were examined for association with prospective smoking cessation among smokers of European ancestry treated with transdermal nicotine in two independent cohorts. Individual SNP and haplotype analysis support the association of NRG3 SNPs and smoking cessation success. NRG3 is a neural-enriched member of the epidermal growth factor family, and a specific ligand for the receptor tyrosine kinase ErbB4, which is also upregulated following nicotine treatment and WD. Mice with significantly reduced levels of NRG3 or pharmacological inhibition of ErbB4 show similar reductions in anxiety following nicotine WD compared with control animals, suggesting a role for NRG3 in nicotine dependence. Although the function of the SNP in NRG3 in humans is not known, these data suggest that Nrg3/ErbB4 signaling may be an important factor in nicotine dependence.


Subject(s)
Genetic Predisposition to Disease/genetics , Intracellular Signaling Peptides and Proteins/genetics , Neuregulins/genetics , Tobacco Use Disorder/genetics , Adolescent , Adult , Afatinib , Aged , Animals , Behavior, Animal/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , DNA/metabolism , Female , Genome-Wide Association Study , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Middle Aged , Mutation , Neuregulins/metabolism , Nicotine/pharmacology , Polymorphism, Single Nucleotide/genetics , Quinazolines/pharmacology , Rats , Receptor, ErbB-4/antagonists & inhibitors , Smoking/drug therapy , Smoking/genetics , Substance Withdrawal Syndrome/genetics , Tobacco Use Cessation Devices , White People/genetics , Young Adult
4.
Genes Brain Behav ; 11(5): 529-38, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22471438

ABSTRACT

Exercise, specifically voluntary wheel running, is a potent stimulator of hippocampal neurogenesis in adult mice. In addition, exercise induces behavioral changes in numerous measures of anxiety in rodents. However, the physiological underpinnings of these changes are poorly understood. To investigate the role of neurogenesis in exercise-mediated anxiety, we examined the cellular and behavioral effects of voluntary wheel running in mice with a reduction in hippocampal neurogenesis, achieved through conditional deletion of ataxia telangiectasia-mutated and rad-3-related protein (ATR), a cell cycle checkpoint kinase necessary for normal levels of neurogenesis. Following hippocampal microinjection of an adeno-associated virus expressing Cre recombinase to delete ATR, mice were exposed to 4 weeks of voluntary wheel running and subsequently evaluated for anxiety-like behavior. Wheel running resulted in increased cell proliferation and neurogenesis, as measured by bromodeoxyuridine and doublecortin, respectively. Wheel running also resulted in heightened anxiety in the novelty-induced hypophagia, open field and light-dark box tests. However, both the neurogenic and anxiogenic effects of wheel running were attenuated following hippocampal ATR deletion, suggesting that increased neurogenesis is an important mediator of exercise-induced anxiety.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Hippocampus/physiopathology , Neurogenesis/physiology , Physical Conditioning, Animal/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Count , Cell Cycle Proteins/genetics , Cell Proliferation , Mice , Mice, Transgenic , Neurons/physiology , Protein Serine-Threonine Kinases/genetics , Running/physiology
5.
Neuroscience ; 205: 178-84, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22240251

ABSTRACT

OPRM1 A118G is a common single nucleotide polymorphism (SNP) in the coding region of the human mu opioid receptor (MOPR) gene OPRM1. This SNP is associated with higher morphine doses required for postoperative analgesia as well as a variety of drug addiction phenotypes. A mouse model possessing the equivalent substitution (A112G) in the Oprm1 gene was generated to facilitate mechanistic studies. Mice homozygous for the G112 allele (G/G) displayed lower antinociception to morphine compared with those homozygous for A112 allele (A/A), similar to humans, suggesting that the mice are a good model to further characterize underlying factors contributing to phenotypes associated with this SNP. Here, we compared [³H]DAMGO binding to the MOPR in the brains of A/A and G/G mice using quantitative in vitro autoradiography. A/A mice exhibited higher [³H]DAMGO binding than G/G in the cingulate, motor, and insular cortices, nucleus accumbens core and shell, hypothalamus, thalamus, amygdala, periaqueductal gray, superficial gray of superior colliculus, and ventral tegmental area. No genotype differences were observed in somatosensory cortex, caudate putamen, and hippocampus. When males and females were examined separately, A/A mice showed higher [³H]DAMGO binding than G/G mice in more brain regions in males than in females. Radioligand binding using brain membranes also showed higher [³H]DAMGO binding in the cortex and thalamus in A/A mice than G/G mice but no genotype differences in the caudate putamen or hippocampus. Thus, the A112G SNP is associated with reduced MOPR expression in some, but not all, brain regions, and appears to have some sex differences. The elevated MOPR expression in periaqueductal gray and thalamus in A/A mice are consistent with their higher antinociceptive responses to morphine. The higher MOPR levels in nucleus accumbens and/or ventral tegmental area of A/A mice is consistent with the higher morphine-induced hyperactivity and locomotor sensitization observed in these mice. Thus, these results provide some insights into the observed decreased clinical opioid potency in humans with the A118G SNP.


Subject(s)
Down-Regulation/genetics , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/genetics , Alanine/genetics , Amino Acid Substitution/drug effects , Amino Acid Substitution/genetics , Analgesics, Opioid/pharmacology , Animals , Disease Models, Animal , Down-Regulation/drug effects , Female , Gene Knock-In Techniques , Glycine/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Polymorphism, Single Nucleotide/drug effects , Polymorphism, Single Nucleotide/genetics , Receptors, Opioid, mu/biosynthesis , Transgenes/drug effects , Transgenes/genetics
6.
Genes Brain Behav ; 8(7): 661-75, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19563516

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) play a pivotal role in excitatory neurotransmission, synaptic plasticity and brain development. Clinical and experimental evidence suggests a dysregulation of NMDAR function and glutamatergic pathways in the pathophysiology of schizophrenia. We evaluated electrophysiological and behavioral properties of NMDAR deficiency utilizing mice that express only 5-10% of the normal level of NMDAR NR1 subunit. Auditory and visual event related potentials yielded significantly increased amplitudes for the P20 and N40 components in NMDAR deficient (NR1(neo)-/-) mice suggesting decreased inhibitory tone. Compared to wild types, NR1(neo)-/- mice spent less time in social interactions and showed reduced nest building. NR1(neo)-/- mice displayed a preference for open arms of a zero maze and central zone of an open field, possibly reflecting decreased anxiety-related behavioral inhibition. However, locomotor activity did not differ between groups in either home cage environment or during behavioral testing. NR1(neo)-/- mice displayed hyperactivity only when placed in a large unfamiliar environment, suggesting that neither increased anxiety nor non-specific motor activation accounts for differential behavioral patterns. Data suggest that NMDAR NR1 deficiency causes disinhibition in sensory processing as well as reduced behavioral inhibition and impaired social interactions. The behavioral signature in NR1(neo)-/- mice supports the impact of impaired NMDAR function in a mouse model with possible relevance to negative symptoms in schizophrenia.


Subject(s)
Brain Chemistry/genetics , Brain/metabolism , Genetic Predisposition to Disease/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics , Schizophrenia/metabolism , Animals , Anxiety/genetics , Auditory Diseases, Central/genetics , Auditory Diseases, Central/metabolism , Auditory Diseases, Central/physiopathology , Behavior, Animal/physiology , Brain/physiopathology , Disease Models, Animal , Evoked Potentials/genetics , Female , Genotype , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neural Inhibition/genetics , Perceptual Disorders/genetics , Perceptual Disorders/metabolism , Perceptual Disorders/physiopathology , Phenotype , Schizophrenia/physiopathology , Social Behavior , Visual Pathways/metabolism , Visual Pathways/physiopathology
7.
Neuroscience ; 152(3): 829-36, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18295410

ABSTRACT

Alterations in the brain that contribute to the development of epilepsy, also called epileptogenesis, are not well understood, which makes it difficult to develop strategies for preventing epilepsy. Here we have studied the role of the CRE binding transcription factors, cyclic-AMP responsive element modulator (CREM) and inducible cyclic-AMP early repressor (ICER), in the development of epilepsy following pilocarpine induced status epilepticus (SE) in mice. Following SE, ICER mRNA and protein are increased in neurons. The increase in ICER, however, is not necessary for neuronal injury following SE as pilocarpine treatment induces equivalent neuronal injury in pyramidal neurons of wild type and CREM/ICER null mice. Following SE, the CREM/ICER null mice develop a more severe epileptic phenotype experiencing approximately threefold more frequent spontaneous seizures. Together these data suggest that the increase in ICER mRNA following SE may have a role in suppressing the severity of epilepsy.


Subject(s)
Cyclic AMP Response Element Modulator/genetics , Cyclic AMP/metabolism , Epilepsy/genetics , Epilepsy/metabolism , Hippocampus/metabolism , Animals , Cyclic AMP Response Element Modulator/metabolism , Disease Progression , Epilepsy/physiopathology , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Pyramidal Cells/metabolism , RNA, Messenger/metabolism , Seizures/genetics , Seizures/metabolism , Seizures/physiopathology , Status Epilepticus/genetics , Status Epilepticus/metabolism , Status Epilepticus/physiopathology , Up-Regulation/genetics
8.
Neuroscience ; 133(3): 647-55, 2005.
Article in English | MEDLINE | ID: mdl-15893884

ABSTRACT

Analysis of mice with targeted disruptions of fosB or the gene encoding dopamine beta-hydroxylase suggests that FosB and adrenergic signaling play critical roles in maternal nurturing behavior. The majority of neonates born to null females from either mutation fail to thrive, and virgin mutant females of both lines exhibit impaired pup retrieval. Considering whether FosB and adrenergic signaling might share a signaling pathway important for maternal behavior, we examined the role of a potential intermediary, cyclic AMP response element-binding protein (CREB). Here we report that approximately 40% of neonates (all heterozygous) born to mice lacking the major isoforms of CREB (Creb-alphaDelta-/-) died within several days of birth. In contrast, heterozygotes born to Creb-alphaDelta+/- females thrived. Cross-fostering demonstrated that neonates born to Creb-alphaDelta(-/dagger/-) females thrived when reared by wild-type females, and that Creb-alphaDelta-/- females were capable of rearing neonates whose maternal care was initiated by wild-type females. Further, virgin Creb-alphaDelta-/- females were deficient in pup retrieval despite exhibiting normal investigation of pups and of novel objects. No maternal behavior phenotype was present in mice with a null mutation of the cyclic AMP response element modulator (Crem) gene. Interestingly, the number of cells immunostaining for phospho-CREB (on Ser(133)) in the medial preoptic area of the hypothalamus, a key region for the expression of maternal behavior, increased nearly three-fold in wild-type mice following exposure to pups but not to novel objects. On the other hand, basal expression and induction of FosB in response to pup exposure appeared to be independent of CREB because levels were equivalent between wild-type and Creb-alphaDelta-/- females. These results implicate CREB in maternal nurturing behavior and suggest that CREB is not critical for expression or induction of FosB in adult virgin female mice.


Subject(s)
Behavior, Animal/physiology , Cyclic AMP Response Element-Binding Protein/genetics , Maternal Behavior/physiology , Preoptic Area/physiology , Animals , Animals, Newborn , Cyclic AMP Response Element Modulator , DNA-Binding Proteins/genetics , Exploratory Behavior/physiology , Female , Genotype , Male , Mice , Mice, Mutant Strains , Phosphorylation , Pregnancy , Proto-Oncogene Proteins c-fos/genetics , Survival Rate , Transcription Factors/genetics
9.
Neuroscience ; 121(2): 355-61, 2003.
Article in English | MEDLINE | ID: mdl-14521994

ABSTRACT

NAC1 cDNA was identified as a novel transcript induced in the nucleus accumbens from rats chronically treated with cocaine. NAC1 is a member of the Bric-a-brac Tramtrac Broad complex/Pox virus and Zinc finger family of transcription factors and has been shown by overexpression studies to prevent the development of behavioral sensitization resulting from repeated cocaine treatment. This paper reports the cloning and characterization of the corresponding gene. The mouse Nac1 gene consist of six exons, with exon 2 containing an alternative splice donor, providing a molecular explanation of the splice variants observed in mouse and rat. Transcripts of Nac1 were ubiquitously detected in different mouse tissues with prominent expression in the brain. The mouse Nac1 gene was localized to chromosome 8, suggesting a highly plausible candidate gene to explain differences in cocaine-induced behaviors between C57BL6/J and DBA/2J mice that had previously been mapped to the area. In addition, a functional AP1 binding site has been identified in an intron 1 enhancer of the Nac1 gene that plays an essential role in the activation of the gene in differentiation of neuroblastoma cells. Co-transfection with c-jun and c-fos expression plasmids, which encode the two subunits of AP1, activated the wild type Nac1 intron 1 enhancer two-fold over basal, nearly at the level of NAC1 enhancer activity seen in differentiated N2A cells. Mutation of the AP1 site completely abrogated all activation of the NAC1 enhancer in differentiated N2A cells. Activation of immediate early genes such as c-fos and c-jun following chronic drug treatments has been well characterized. The present data describe one potential regulatory cascade involving these transcription factors and activation of NAC1. Identification of drug induced alterations in gene expression is key to understanding the types of molecular adaptations underlying addiction.


Subject(s)
DNA-Binding Proteins/genetics , Nerve Tissue Proteins/metabolism , Transcription Factor AP-1/physiology , Zinc Fingers/physiology , Animals , Blotting, Northern , Blotting, Southern , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 8 , Cloning, Molecular , Colforsin/pharmacology , DNA Probes , Drosophila , Drosophila Proteins , Exons , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mutagenesis , Nerve Tissue Proteins/genetics , Neuroblastoma , Promoter Regions, Genetic/physiology , RNA, Messenger/biosynthesis , Rats , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis , Transcription Factor AP-1/metabolism , Transcription Factors , Transfection
10.
Mol Psychiatry ; 8(7): 664-72, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12874602

ABSTRACT

alpha 1 Adrenergic receptors mediate a variety of physiological responses and have been well studied in the cardiovascular and peripheral nervous system. However, their role in the central nervous system remains ill defined because of the lack of highly specific ligands to the alpha1 receptor subtypes. Here, we have employed gene targeting to elucidate the role of alpha 1d receptors in vivo. In addition to disrupting function, the insertion of the lacZ gene into the alpha 1d receptor locus enabled the specific identification of cells expressing the alpha 1d gene. These cells are localized in the cortex, hippocampus, olfactory bulb, dorsal geniculate and ventral posterolateral nuclei of the thalamus. Behaviorally, the alpha 1d(-/-) mice show normal locomotor activity during the subjective day, or resting phase of their cycle. However, during subjective night, or active phase, wheel-running activity is significantly reduced in mutant mice. Furthermore, these mice show a reduction in exploratory rearing behavior in a novel cage environment. Lastly, alpha 1d(-/-) mice show reduced hyperlocomotion after acute amphetamine administration. Together, these data reveal the functional importance of alpha 1d adrenoceptors in mediating a variety of stimulus-induced changes in locomotor behaviors. While the sensitivity of noradrenergic neurons to environmental stimuli has been well documented, our data demonstrate that at least some of these post-synaptic responses are mediated by alpha 1d adrenergic receptors.


Subject(s)
Motor Activity/genetics , Nerve Tissue Proteins/physiology , Receptors, Adrenergic, alpha-1/physiology , Alleles , Animals , Avoidance Learning/drug effects , Brain/physiology , Brain Mapping , Chimera/genetics , Circadian Rhythm , Cocaine/pharmacology , Dextroamphetamine/pharmacology , Environment , Exploratory Behavior/drug effects , Female , Gene Targeting , Lac Operon , Locomotion , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Mutagenesis, Insertional , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , RNA, Messenger/analysis , Reaction Time/genetics , Receptors, Adrenergic, alpha-1/deficiency , Receptors, Adrenergic, alpha-1/genetics , Rotation
11.
Hippocampus ; 12(1): 18-26, 2002.
Article in English | MEDLINE | ID: mdl-11918283

ABSTRACT

The cyclic AMP (cAMP)-response element binding protein (CREB) is an activity-dependent transcription factor that plays a role in synaptic plasticity and memory storage in Aplysia, Drosophila, and rodents. Mice with targeted deletions of two CREB isoforms (alpha and delta; CREB alphadelta mice) have been characterized on a mixed genetic background of C57BL/6 (B6) and 129/SvEv (129), as well as on a defined F1 hybrid of B6 and FVB/N, and these results suggest that the phenotype of CREB alphadelta mice depends critically on genetic background. In an examination of the hypothesis that the role of CREB in learning and memory can be influenced by strain differences, we analyzed mice with the CREB alphadelta mutation on an F1 hybrid background of B6 and 129 strains. CREB alphadelta mice on this background had impaired short-term and long-term cued and contextual fear conditioning and normal spatial learning in the Morris water maze. Our results suggest that at least some aspects of hippocampal function are normal in CREB alphadelta mice, and that CREB alphadelta mice on the B6/129 F1 background have alterations in amygdala function. These studies underscore the importance of controlling for genetic background in the behavioral analysis of knockout and transgenic mice.


Subject(s)
Behavior, Animal/physiology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/physiology , Hippocampus/physiology , Mutation , Animals , Conditioning, Psychological/physiology , Fear/physiology , Hybridization, Genetic , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
12.
J Neurosci ; 21(23): 9438-44, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11717377

ABSTRACT

Addiction is a complex process that relies on the ability of an organism to integrate positive and negative properties of drugs of abuse. Therefore, studying the reinforcing as well as aversive components of drugs of abuse in a single model system will enable us to understand the role of final common mediators, such as cAMP response element-binding protein (CREB), in the addiction process. To this end, we analyzed mice with a mutation in the alpha and Delta isoforms of the CREB gene. Previously we have shown that CREB(alphaDelta) mutant mice in a mixed genetic background show attenuated signs of physical dependence, as measured by the classic signs of withdrawal. We have generated a uniform genetically stable F1 hybrid (129SvEv/C57BL/6) mouse line harboring the CREB mutation. We have found the functional activity of CREB in these F1 hybrid mice to be dramatically reduced compared with their wild-type littermates. These mice maintain a reduced withdrawal phenotype after chronic morphine. We are now poised to examine a number of complex behavioral phenotypes related to addiction in a well defined CREB-deficient mouse model. We demonstrate that the aversive properties of morphine are still present in CREB mutant mice despite a reduction of physical withdrawal. On the other hand, these mice do not respond to the reinforcing properties of morphine in a conditioned place preference paradigm. In contrast, CREB mutant mice demonstrate an enhanced response to the reinforcing properties of cocaine compared with their wild-type controls in both conditioned place preference and sensitization behaviors. These data may provide the first paradigm for differential vulnerability to various drugs of abuse.


Subject(s)
Cocaine-Related Disorders/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Morphine Dependence/metabolism , Reinforcement, Psychology , Alleles , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/metabolism , Cocaine/administration & dosage , Cocaine-Related Disorders/genetics , Conditioning, Psychological/drug effects , Crosses, Genetic , Disease Models, Animal , Disease Susceptibility , Drug Implants , Injections, Intraperitoneal , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Morphine/administration & dosage , Morphine/antagonists & inhibitors , Morphine Dependence/genetics , Motivation , Motor Activity/drug effects , Narcotic Antagonists/pharmacology , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , Response Elements/physiology , Spatial Behavior/drug effects , Spatial Behavior/physiology
13.
J Pharmacol Exp Ther ; 298(3): 1092-100, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11504806

ABSTRACT

Plasticity in serotonergic transmission in serotonin or 5-hydroxytryptamine (5-HT) receptor mutants was examined by measuring the regulation of extracellular 5-HT levels in the striatum and ventral hippocampus of 5-HT(1A) and 5-HT(1B) receptor knockout mice using in vivo microdialysis. The efficacy of genetic deletion was verified by showing blunted regulation of extracellular 5-HT with selective 5-HT receptor agonists. 5-HT(1A) receptor knockout mice failed to demonstrate reduction of extracellular 5-HT in response to systemic administration of the 5-HT(1A) receptor agonist R-8-hydroxydipropylaminotetralin (R-8-OH-DPAT) and 5-HT(1B) receptor knockout mice failed to demonstrate reduction of extracellular 5-HT in response to systemic administration of the 5-HT(1B) receptor agonist CP 94,253. Plasticity also developed to deletion of the complementary autoreceptor. 5-HT(1A) receptor knockout mice demonstrated a significantly greater response to CP 94,253 in the striatum, but not the ventral hippocampus, suggesting the development of enhanced sensitivity of striatal 5-HT(1B) receptors. In 5-HT(1B) receptor knockout mice, R-8-OH-DPAT evoked a significantly diminished response in the ventral hippocampus, but not the striatum, suggesting the potential desensitization of 5-HT(1A) receptors in the median raphe nucleus. The pattern of regional compensations between somatodendritic and terminal autoreceptors was confirmed by pharmacological challenges using the selective serotonin reuptake inhibitor fluoxetine combined with either a 5-HT(1A) (WAY 100635) or a 5-HT(1B/1D) (GR 127935) receptor antagonist. The regional pattern of compensation may be determined by the preferential role of 5-HT(1A) or 5-HT(1B) receptors in regulating 5-HT release. Taken together, these results demonstrate the development of regional plasticity between complementary somatodendritic and terminal autoreceptors after the genetic deletion of 5-HT(1A) or 5-HT(1B) receptors.


Subject(s)
Gene Deletion , Receptors, Serotonin/deficiency , Receptors, Serotonin/genetics , Serotonin/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Area Under Curve , Fluoxetine/pharmacology , Genotype , Male , Mice , Mice, Knockout , Microdialysis , Piperazines/pharmacology , Pyridines/pharmacology , Receptor, Serotonin, 5-HT1B , Receptors, Serotonin, 5-HT1 , Reverse Transcriptase Polymerase Chain Reaction , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology
14.
Proc Natl Acad Sci U S A ; 97(18): 9984-9, 2000 Aug 29.
Article in English | MEDLINE | ID: mdl-10954748

ABSTRACT

Heterotrimeric G proteins mediate the earliest step in cell responses to external events by linking cell surface receptors to intracellular signaling pathways. G(z) is a member of the G(i) family of G proteins that is prominently expressed in platelets and brain. Here, we show that deletion of the alpha subunit of G(z) in mice: (i) impairs platelet aggregation by preventing the inhibition of cAMP formation normally seen at physiologic concentrations of epinephrine, and (ii) causes the mice to be more resistant to fatal thromboembolism. Loss of G(zalpha) also results in greatly exaggerated responses to cocaine, reduces the analgesic effects of morphine, and abolishes the effects of widely used antidepressant drugs that act as catecholamine reuptake inhibitors. These changes occur despite the presence of other G(ialpha) family members in the same cells and are not accompanied by detectable compensatory changes in the level of expression of other G protein subunits. Therefore, these results provide insights into receptor selectivity among G proteins and a model for understanding platelet function and the effects of psychoactive drugs.


Subject(s)
Blood Platelets/physiology , Brain/physiology , GTP-Binding Protein alpha Subunits , Heterotrimeric GTP-Binding Proteins/metabolism , Platelet Activation/physiology , Psychotropic Drugs/pharmacology , Animals , Blood Platelets/drug effects , Brain/drug effects , Cocaine/pharmacology , Collagen/pharmacology , Cyclic AMP/blood , Epinephrine/pharmacology , Heterotrimeric GTP-Binding Proteins/deficiency , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Lung/drug effects , Lung/physiopathology , Male , Mice , Mice, Knockout , Morphine/pharmacology , Motor Activity , Platelet Aggregation , Signal Transduction , Thromboembolism/genetics , Thromboembolism/physiopathology , Thromboembolism/prevention & control
15.
Science ; 286(5448): 2358-61, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10600750

ABSTRACT

Nerve growth factor (NGF) and other neurotrophins support survival of neurons through processes that are incompletely understood. The transcription factor CREB is a critical mediator of NGF-dependent gene expression, but whether CREB family transcription factors regulate expression of genes that contribute to NGF-dependent survival of sympathetic neurons is unknown. CREB-mediated gene expression was both necessary for NGF-dependent survival and sufficient on its own to promote survival of sympathetic neurons. Moreover, expression of Bcl-2 was activated by NGF and other neurotrophins by a CREB-dependent transcriptional mechanism. Overexpression of Bcl-2 reduced the death-promoting effects of CREB inhibition. Together, these data support a model in which neurotrophins promote survival of neurons, in part through a mechanism involving CREB family transcription factor-dependent expression of genes encoding prosurvival factors.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Nerve Growth Factor/pharmacology , Neurons/cytology , Sympathetic Nervous System/cytology , Animals , Apoptosis , Axons/drug effects , Axons/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cell Nucleus/metabolism , Cell Survival , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Genes, bcl-2 , Genetic Vectors , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Signal Transduction , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Transfection
16.
J Biol Chem ; 273(16): 9971-5, 1998 Apr 17.
Article in English | MEDLINE | ID: mdl-9545342

ABSTRACT

The angiotensin-converting enzyme (ACE) gene produces two mRNA species from tissue-specific promoters. The transcription start site of the mRNA for the smaller testicular isozyme (ACET) is located within an intron of the larger transcription unit that encodes the pulmonary isozyme (ACEP).We have previously demonstrated that a 298-base pair DNA fragment, 5' to the rabbit ACET mRNA transcription initiation site, can activate the testicular expression of a transgenic reporter gene. In the current study, using the same transgenic reporter system, we identified a putative cyclic AMP response element present within this DNA fragment to be absolutely essential for transcriptional activation. Moreover, we observed that ACET mRNA was not expressed in the testes of mice homozygous for a null mutation in the transcription factor CREM. However, in the same mice, ACEP mRNA was abundantly expressed in the lung. Our observations indicate that ACET mRNA expression in the testes is regulated by the putative cyclic AMP response element present 5' to the transcription start site and the corresponding transcription factor CREM.


Subject(s)
DNA-Binding Proteins/biosynthesis , Isoenzymes/biosynthesis , Peptidyl-Dipeptidase A/biosynthesis , Testis/enzymology , Transcription, Genetic , Animals , Brain/enzymology , Chloramphenicol O-Acetyltransferase/biosynthesis , Cyclic AMP Response Element Modulator , DNA-Binding Proteins/genetics , Isoenzymes/genetics , Kidney/enzymology , Lung/enzymology , Male , Mice , Mice, Knockout , Mice, Transgenic , Organ Specificity , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/biosynthesis , Rabbits , Recombinant Fusion Proteins/biosynthesis , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
17.
J Mol Med (Berl) ; 76(2): 104-10, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9500675

ABSTRACT

Many drugs of abuse, administered repeatedly over time, cause physical dependence which is expressed by a withdrawal syndrome when the drug is removed from the system. These processes can be thought of as adaptations of the neuronal system to an altered pharmacological state. The molecular mechanisms underlying these adaptations are still not known. A considerable amount of evidence is accumulating which implicates alterations in several components of the cAMP signal transduction cascade in these drug-induced processes. The transcription factor cAMP response element binding protein (CREB) in particular has been shown both in vitro and in vivo to be altered in response to several drugs of abuse, including opiates. This review discusses in detail this transcription factor and demonstrates its importance in the signal transduction cascades involving abused substances.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Substance-Related Disorders/genetics , Animals , Brain/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Forecasting , Gene Expression Regulation , Genes, Immediate-Early , Mice , Morphine/administration & dosage , Substance Withdrawal Syndrome
18.
Curr Biol ; 7(1): 1-11, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-8999994

ABSTRACT

BACKGROUND: The cAMP responsive element binding protein (CREB) is a transcription factor the activity of which is modulated by increases in the intracellular levels of cAMP and calcium. Results from studies with Aplysia, Drosophila and mice indicate that CREB-activated transcription is required for long-term memory. Furthermore, a recent study found that long-term memory for olfactory conditioning can be induced with a single trial in transgenic Drosophila expressing a CREB activator, whereas in normal flies, with presumably lower CREB-mediated transcription levels, conditioning requires multiple spaced trials. This suggests that CREB-mediated transcription is important in determining the type of training required for long-term memory of olfactory conditioning in Drosophila. Interestingly, studies with cultured Aplysia neurons indicated that removing a CREB repressor promoted the formation of long-term facilitation, a cellular model of non-associative memory. RESULTS: Here, we have confirmed that mice lacking the alpha and Delta CREB proteins (CREBalphaDelta-) have abnormal long-term, but not short-term, memory, as tested in an ethologically meaningful task. Importantly, additional spaced training can overcome the profound memory deficits of CREBalphaDelta- mutants. Increasing the intertrial interval from 1 to 60 minutes overcame the memory deficits of the CREBalphaDelta- mice in three distinct behavioral tasks: contextual fear conditioning, spatial learning and socially transmitted food preferences. CONCLUSIONS: Previous findings and results presented here demonstrate that CREB mutant mice have profound long-term memory deficits. Importantly, our findings indicate that manipulations of CREB function can affect the number of trials and the intertrial interval required for committing information to long-term memory. Remarkably, this effect of CREB function is not restricted to simple conditioning tasks, but also affects complex behaviours such as spatial memory and memory for socially transmitted food preferences.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Memory/physiology , Animals , Conditioning, Psychological/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Female , Food Preferences/physiology , Male , Maze Learning/physiology , Mice , Mice, Mutant Strains , Time Factors
19.
Science ; 273(5275): 657-9, 1996 Aug 02.
Article in English | MEDLINE | ID: mdl-8662559

ABSTRACT

Chronic morphine administration induces an up-regulation of several components of the cyclic adenosine 5'-monophosphate (cAMP) signal transduction cascade. The behavioral and biochemical consequences of opiate withdrawal were investigated in mice with a genetic disruption of the alpha and Delta isoforms of the cAMP-responsive element-binding protein (CREB). In CREBalphadelta mutant mice the main symptoms of morphine withdrawal were strongly attenuated. No change in opioid binding sites or in morphine-induced analgesia was observed in these mutant mice, and the increase of adenylyl cyclase activity and immediate early gene expression after morphine withdrawal was normal. Thus, CREB-dependent gene transcription is a factor in the onset of behavioral manifestations of opiate dependence.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/physiology , Morphine Dependence/etiology , Morphine/administration & dosage , Substance Withdrawal Syndrome/physiopathology , Adenylyl Cyclases/metabolism , Analgesia , Animals , Behavior, Animal , Cerebral Cortex/enzymology , Drug Tolerance , Gene Expression Regulation , Gene Targeting , Genes, Immediate-Early , Locus Coeruleus/metabolism , Mice , Morphine/adverse effects , Morphine/pharmacology , Morphine Dependence/metabolism , Mutation , Naloxone/pharmacology , Receptors, Opioid/metabolism , Signal Transduction
20.
Nature ; 380(6570): 162-5, 1996 Mar 14.
Article in English | MEDLINE | ID: mdl-8600391

ABSTRACT

Spermatogenesis is a complex developmental process that occurs in several phases. A large number of genes have been identified that are expressed during spermatogenesis, but the biological significance of many of these is not yet known. We have used gene targeting to selectively eliminate the transcription factor CREM (cyclic AMP- responsive element modulator), which is thought to be important for mammalian spermatogenesis. Male mice deficient for all CREM proteins are sterile, as their developing spermatids fail to differentiate into sperm, and postmeiotic gene expression in the testis declines dramatically. The cessation of sperm development is not accompanied by decreases in the levels of follicle-stimulating hormone or testosterone. Our findings indicate that the CREM gene is essential for spermatogenesis, and mice deficient for this transcription factor could serve as a model system for the study of idiopathic infertility in men.


Subject(s)
DNA-Binding Proteins/physiology , Repressor Proteins , Spermatogenesis/physiology , Androgens/metabolism , Animals , Cell Line , Cyclic AMP Response Element Modulator , DNA-Binding Proteins/genetics , Epididymis/pathology , Follicle Stimulating Hormone/blood , Gene Targeting , Infertility/genetics , Leucine Zippers/genetics , Male , Mice , Mice, Inbred C57BL , Organ Size , Proteins/metabolism , RNA/metabolism , Spermatogenesis/genetics , Testis/metabolism , Testis/pathology , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...