Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 360
Filter
1.
J Biol Chem ; 298(9): 102278, 2022 09.
Article in English | MEDLINE | ID: mdl-35863435

ABSTRACT

Immediate early genes (IEGs) are transcribed in response to neuronal activity from sensory stimulation during multiple adaptive processes in the brain. The transcriptional profile of IEGs is indicative of the duration of neuronal activity, but its sensitivity to the strength of depolarization remains unknown. Also unknown is whether activity history of graded potential changes influence future neuronal activity. In this work with dissociated rat cortical neurons, we found that mild depolarization-mediated by elevated extracellular potassium (K+)-induces a wide array of rapid IEGs and transiently depresses transcriptional and signaling responses to a successive stimulus. This latter effect was independent of de novo transcription, translation, and signaling via calcineurin or mitogen-activated protein kinase. Furthermore, as measured by multiple electrode arrays and calcium imaging, mild depolarization acutely subdues subsequent spontaneous and bicuculline-evoked activity via calcium- and N-methyl-d-aspartate receptor-dependent mechanisms. Collectively, this work suggests that a recent history of graded potential changes acutely depress neuronal intrinsic properties and subsequent responses. Such effects may have several potential downstream implications, including reducing signal-to-noise ratio during synaptic plasticity processes.


Subject(s)
Action Potentials , Calcineurin , Genes, Immediate-Early , Neurons , Transcription, Genetic , Action Potentials/drug effects , Action Potentials/physiology , Animals , Bicuculline/pharmacology , Calcineurin/genetics , Calcineurin/metabolism , Calcium/metabolism , GABA-A Receptor Antagonists/pharmacology , Genes, Immediate-Early/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Neurons/physiology , Potassium/metabolism , Potassium/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
2.
Neurochem Int ; 150: 105181, 2021 11.
Article in English | MEDLINE | ID: mdl-34509560

ABSTRACT

As the relationship among diet, brain aging and memory is complex, it provides ample opportunity for research in multiple directions including behaviour, epigenetics and neuroplasticity. Nutritional deficiencies together with genetic and environmental factors are the major cause of many age-associated pathologies including memory loss. A compromised vitamin B12-folate status in older people is highly prevalent worldwide. Researchers have established a close association between the adequate level of B12-folate and the maintenance of cognitive brain functions. One of the main reasons for age-associated memory loss is downregulation of neuronal immediate early genes (nIEGs). Therefore, we hypothesize here that vitamin B12-folic acid supplementation in old mice can improve memory by altering the expression status of nIEGs. To check this, 72-week-old male Swiss albino mice were orally administered with 2 µg of vitamin B12 and 22 µg of folic acid/mouse/day for eight weeks. Such supplementation improved recognition memory in old and altered the expression of nIEGs. The expression of nIEGs was further found to be regulated by changes in DNA methylation at their promoter regions and CREB phosphorylation (pCREB). In addition, Golgi-Cox staining showed significant improvement in dendritic length, number of branching points and spine density of hippocampal CA1 pyramidal neurons by B12-folic acid supplementation. Taken together, these findings suggest that vitamin B12-folic acid supplementation regulates nIEGs expression and improves dendritic arborization of hippocampal neurons and memory in old male mice.


Subject(s)
Aging/drug effects , Folic Acid/administration & dosage , Genes, Immediate-Early/drug effects , Memory Disorders/drug therapy , Neuronal Plasticity/drug effects , Vitamin B 12/administration & dosage , Aging/genetics , Aging/metabolism , Animals , Dietary Supplements , Genes, Immediate-Early/physiology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Male , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Neuronal Plasticity/physiology
3.
Gene ; 800: 145842, 2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34274479

ABSTRACT

Treatment of serum-starved quiescent human cells with fetal bovine serum (FBS), epidermal growth factor (EGF), or the phorbol ester (12-O-tetradecanoylphorbol-13-acetate, TPA) activates the RAS-MAPK pathway which initiates a transcriptional program which drives cells toward proliferation. Stimulation of the RAS-MAPK pathway activates mitogen- and stress-activated kinases (MSK) 1 and 2, which phosphorylate histone H3 at S10 (H3S10ph) or S28 (H3S28ph) (nucleosomal response) located at the regulatory regions of immediate-early genes, setting in motion a series of chromatin remodeling events that result in transcription initiation. To investigate immediate-early genes regulated by the MSK, we have completed transcriptome analyses (RNA sequencing) of human normal fibroblast cells (CCD-1070Sk) stimulated with EGF or TPA ± H89, a potent MSK/PKA inhibitor. The induction of many immediate-early genes was independent of MSK activity. However, the induction of immediate-early genes attenuated with H89 also had reduced induction with the PKA inhibitor, Rp-cAMPS. Several EGF-induced genes, coding for transcriptional repressors, were further upregulated with H89 but not with Rp-cAMPS, suggesting a role for MSK in modulating the induction level of these genes.


Subject(s)
Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Mitogens/pharmacology , Cell Line , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Epidermal Growth Factor/pharmacology , Fibroblasts/physiology , Gene Expression Profiling , Genes, Immediate-Early/drug effects , Humans , Isoquinolines/pharmacology , Reproducibility of Results , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Sulfonamides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thionucleotides/pharmacology
4.
BMC Vet Res ; 17(1): 247, 2021 Jul 18.
Article in English | MEDLINE | ID: mdl-34275451

ABSTRACT

BACKGROUND: Pseudorabies virus (PRV), a member of the Alphaherpesviruses, is one of the most important pathogens that harm the global pig industry. Accumulated evidence indicated that PRV could infect humans under certain circumstances, inducing severe clinical symptoms such as acute human encephalitis. Currently, there are no antiviral drugs to treat PRV infections, and vaccines available only for swine could not provide full protection. Thus, new control measures are urgently needed. RESULTS: In the present study, kaempferol exhibited anti-PRV activity in mice through improving survival rate by 22.22 %, which was higher than acyclovir (Positive control) with the survival rate of 16.67 % at 6 days post infection (dpi); meanwhile, the survival rate was 0 % at 6 dpi in the infected-untreated group. Kaempferol could inhibit the virus replication in the brain, lung, kidney, heart and spleen, especially the viral gene copies were reduced by over 700-fold in the brain, which was further confirmed by immunohistochemical examination. The pathogenic changes induced by PRV infection in these organs were also alleviated. The transcription of the only immediate-early gene IE180 in the brain was significantly inhibited by kaempferol, leading to the decreased transcriptional levels of the early genes (EPO and TK). The expression of latency-associated transcript (LAT) was also inhibited in the brain, which suggested that kaempferol could inhibit PRV latency. Kaempferol-treatment could induce higher levels of IL-1ß, IL-4, IL-6, TNF-α and IFN-γ in the serum at 3 dpi which were then declined to normal levels at 5 dpi. CONCLUSIONS: These results suggested that kaempferol was expected to be a new alternative control measure for PRV infection.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 1, Suid/drug effects , Kaempferols/pharmacology , Pseudorabies/drug therapy , Acyclovir/pharmacology , Animals , Brain , Gene Expression Regulation, Viral , Genes, Immediate-Early/drug effects , Herpesvirus 1, Suid/genetics , Male , Mice , Pseudorabies/mortality , Pseudorabies/pathology , Virus Replication/drug effects
5.
Biochem Pharmacol ; 192: 114696, 2021 10.
Article in English | MEDLINE | ID: mdl-34302794

ABSTRACT

Insulin binding to the insulin receptor triggers intracellular signaling cascades involving the activation of protein and lipid kinases. As a result, multiple biological functions of the cells are changed. Here, we analyzed the regulation and signaling cascades leading to insulin-induced activation of the stimulus-responsive transcription factors. For the analyses, we used chromatin-embedded reporter genes having a cellular nucleosomal organisation, and fibroblasts expressing human insulin receptors (HIRcB cells). The results show that stimulation of the insulin receptor induced the expression of the transcription factor Egr-1. Attenuation of Egr-1 promoter activation was observed following expression of a dominant-negative mutant of the ternary complex factor Elk-1. These data were corroborated by experiments showing that insulin receptor stimulation increased the transcriptional activation potential of Elk-1. In addition, the transcriptional activity of AP-1 was significantly elevated in insulin-stimulated HIRcB cells. Expression of the dominant-negative mutant of Elk-1 reduced insulin-induced activation of AP-1, indicating that Elk-1 controls both serum response element and AP-1-regulated transcription. Moreover, we show that stimulation of the insulin receptor activates cyclic AMP response element (CRE)-controlled transcription, involving the transcription factor CREB. Insulin-induced transcription of Elk-1 and CREB-controlled reporter genes was attenuated by overexpression of MAP kinase phosphatase-1 or a constitutively active mutant of calcineurin A, indicating that both phosphatases are part of a negative feedback loop for reducing insulin-mediated gene transcription. Finally, we show that expression of the adenoviral protein E1A selectively reduced CRE-mediated transcription following stimulation of the insulin receptor. These data indicate that insulin-regulated transcription of CRE-containing genes is under epigenetic control.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Genes, Immediate-Early/physiology , Insulin/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Transcription, Genetic/physiology , Cell Line , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Genes, Immediate-Early/drug effects , Humans , Insulin/pharmacology , Receptor, Insulin/agonists , Transcription, Genetic/drug effects
6.
Brain Res Bull ; 174: 260-267, 2021 09.
Article in English | MEDLINE | ID: mdl-34197938

ABSTRACT

Mu-opioid receptors (MOPs) mediate and modulate social reward and social interaction. However, few studies have examined the functionality of this system in rodent models of social impairment. Deficits in social motivation and cognition are observed in rodents following pre-natal exposure to the anti-epileptic valproic acid (VPA), however it is not known whether MOP functionality is altered in these animals. The present study examined the effects of acute administration of the prototypical MOP agonist morphine (1 mg/kg) on social behavioural responding in the 3-chamber test and immediate early gene expression in adolescent rats (postnatal day 28-43) prenatally exposed to VPA vs saline-exposed controls. Pharmacokinetic analysis of morphine concentration, MOP binding and expression were also examined. The data revealed that sociability and social novelty preference in the 3-chamber test were reduced in rats prenatally exposed to VPA compared to saline-exposed control counterparts. Morphine reduced both sociability and social novelty preference behaviour in saline-, but not VPA-, exposed rats. Analysis of immediate early gene expression revealed that morphine reduced the expression of cfos in the prefrontal cortex of both saline- and VPA-exposed rats and reduced expression of cfos and junb in the hippocampus of VPA-exposed rats only. Pharmacokinetic analysis revealed similar concentrations of morphine in the plasma and brain of both saline- and VPA-exposed rats and similar thalamic MOP occupancy levels. Gene and protein expression of MOP in prefrontal cortex and hippocampus did not differ between saline and VPA-exposed rats. These data indicate differential effects of morphine on social responding and immediate early gene expression in the hippocampus of VPA-exposed rats compared with saline-exposed controls. This study provides support for altered MOP functionality in rats prenatally exposed to VPA, which may underlie the social deficits observed in the model.


Subject(s)
Anticonvulsants/toxicity , Gene Expression/drug effects , Genes, Immediate-Early/drug effects , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/psychology , Receptors, Opioid, mu/agonists , Social Behavior , Valproic Acid/toxicity , Analgesics, Opioid/pharmacology , Animals , Female , Genes, fos/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Morphine/pharmacology , Pregnancy , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-jun/genetics , Rats , Rats, Sprague-Dawley
7.
J Neurochem ; 158(4): 912-927, 2021 08.
Article in English | MEDLINE | ID: mdl-34050937

ABSTRACT

Post-traumatic stress disorder (PTSD) is characterized by depression/anxiety and memory failure, primarily fear memory. According to the reports, neuroinflammation and synaptic plasticity can play a role in the neurophysiological mechanisms underlying PTSD. Bromodomain-containing protein 4 (Brd4) intriguingly affects regulating of inflammatory responses and learning and memory. This study aimed to explore the effect of inhibiting Brd4 on depression/anxiety-like behaviors, spatial and fear memory, and underlying mechanisms in a model of PTSD. Inescapable foot shocks (IFS) with a sound reminder in 6 days were used to induce PTSD-like behaviors which were tested using contextual and cue fear tests, sucrose preference test, open-field test, elevated plus maze test, and Y-maze test. Meanwhile, the Brd4 inhibitor JQ1 was used as an intervention. The results found that IFS induced PTSD-like behaviors and indicated obvious Brd4 expression in microglia of the prefrontal cortex (PFC), hippocampus, and amygdala, pro-inflammatory cytokines over-expression, microglial activation, and nuclear factor-kappa B over-expression in PFC and hippocampus but not in amygdala. Meanwhile, the alterations of immediate early genes (IEGs) were found in PFC, hippocampus, and amygdala. Besides, dendritic spine density was reduced in PFC and hippocampus but was elevated in amygdala of rats with IFS. In addition, treatment with JQ1 significantly reduced freezing time in the contextual and cue fear test, reversed the behavioral impairment, decreased the elevated neuroinflammation, and normalized the alteration in IEGs and dendritic spine densities. The results suggested that Brd4 was involved in IFS-induced PTSD-like behaviors through regulating neuroinflammation, dynamics of IEGs, and synaptic plasticity.


Subject(s)
Encephalitis/drug therapy , Fear/psychology , Gene Expression Regulation/drug effects , Genes, Immediate-Early/drug effects , Nuclear Proteins/antagonists & inhibitors , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Transcription Factors/antagonists & inhibitors , Animals , Anxiety/drug therapy , Anxiety/psychology , Azepines/pharmacology , Azepines/therapeutic use , Brain Chemistry/drug effects , Cues , Dendritic Spines/drug effects , Depression/drug therapy , Depression/psychology , Encephalitis/genetics , Male , Memory/drug effects , Motor Activity/drug effects , Rats , Rats, Wistar , Triazoles/pharmacology , Triazoles/therapeutic use
8.
Behav Brain Res ; 404: 113160, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33577880

ABSTRACT

BACKGROUND: Although extensively studied, the effect of antipsychotics is not completely understood at a network level. We tested the hypothesis that acute administration of haloperidol would modulate functional connectivity of brain regions relevant to schizophrenia pathophysiology. To assess putative changes in brain network properties and regional interactivity, we studied the expression of Homer1a, an Immediate Early Gene (IEG) demonstrated to be induced by antipsychotic administration and coding for a protein involved in glutamatergic synapses remodeling. METHODS: Sprague-Dawley rats (n = 26) assigned to vehicle (VEH; NaCl 0.9%) or haloperidol (HAL; 0.8 mg/kg) were included in the network analysis. Homer1a mRNA induction was evaluated by in situ hybridization. Signal intensity analysis was performed in 33 Regions of Interest (ROIs) in the cortex, the caudate putamen, and the nucleus accumbens. A signal correlation analysis was performed, computing all possible pairwise Pearson correlations among ROIs in the two groups. Two networks were generated for HAL and VEH groups, and their properties and topography were explored. RESULTS: VEH and HAL networks showed qualitative differences in global efficiency and clustering coefficient. The HAL network showed enhanced interactivity between cortical and striatal regions, and within caudate putamen subdivisions. On the other hand, it exhibited reduced inter-correlations between cingulate cortex and anterior insula and caudate putamen and nucleus accumbens. Moreover, haloperidol was able to modulate centrality of crucial functional hubs. These preclinical results corroborate and expand the clinical evidence that antipsychotics may modulate specific brain network properties and disease-related circuits' interactivity.


Subject(s)
Gene Regulatory Networks/drug effects , Genes, Immediate-Early/drug effects , Haloperidol/pharmacology , Nerve Net/drug effects , Post-Synaptic Density/drug effects , Receptors, Glutamate/drug effects , Animals , Antipsychotic Agents , Brain/drug effects , Brain/metabolism , In Situ Hybridization , Male , Neural Pathways/drug effects , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Transcriptome
9.
Psychopharmacology (Berl) ; 237(6): 1783-1793, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32296859

ABSTRACT

RATIONALE: Patients diagnosed with schizophrenia typically receive life-long treatments with antipsychotic drugs (APDs). However, the impact of chronic APDs treatment on neuroplastic mechanisms in the brain remains largely elusive. OBJECTIVE: Here, we focused on blonanserin, a second-generation antipsychotic (SGA) that acts as an antagonist at dopamine D2, D3, and serotonin 5-HT2A receptors, and represents an important tool for the treatment of schizophrenia. METHODS: We used rats to investigate the ability of chronic treatment blonanserin to modulate the activity of brain structures relevant for schizophrenia, under baseline conditions or in response to an acute forced swim session (FSS). We measured the expression of different immediate early genes (IEGs), including c-Fos, Arc/Arg 3.1, Zif268 and Npas4. RESULTS: Blonanserin per se produced limited changes in the expression of these genes under basal conditions, while, as expected, FSS produced a significant elevation of IEGs transcription in different brain regions. The response of blonanserin-treated rats to FSS show anatomical and gene-selective differences. Indeed, the upregulation of IEGs was greatly reduced in the striatum, a brain structure enriched in dopamine receptors, whereas the upregulation of some genes (Zif268, Npas4) was largely preserved in other regions, such as the prefrontal cortex and the ventral hippocampus. CONCLUSIONS: Taken together, our findings show that chronic exposure to blonanserin modulates selective IEGs with a specific anatomical profile. Moreover, the differential activation of specific brain regions under challenging conditions may contribute to specific clinical features of the drug.


Subject(s)
Antipsychotic Agents/administration & dosage , Brain/drug effects , Genes, Immediate-Early/drug effects , Piperazines/administration & dosage , Piperidines/administration & dosage , Stress, Psychological/drug therapy , Animals , Brain/physiology , Drug Administration Schedule , Genes, Immediate-Early/physiology , Male , Rats , Rats, Sprague-Dawley , Receptors, Dopamine/metabolism , Schizophrenia/drug therapy , Schizophrenia/genetics , Stress, Psychological/genetics , Stress, Psychological/psychology
10.
Mol Neurobiol ; 57(1): 11-22, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31512116

ABSTRACT

Large body of animal work and emerging clinical findings have suggested that early exposure to anesthetics may result in increased risk of learning disabilities and behavioral impairments. Recent studies have begun to investigate anesthesia-induced epigenetic modifications to elucidate their role in behavioral and neurodevelopmental abnormalities. Here we examine sevoflurane-induced transgenerational modifications of subicular neuronal DNA methylation and expression of immediate early genes (IEGs), arc and junB, crucial to synaptic plasticity and normal neuronal development. We show that 6 h sevoflurane exposure in postnatal day 7 rat pups resulted in decreased neuronal 5-methycytosine, indicating reduced DNA methylation. This effect is transgenerationally expressed in offspring born to exposed mothers which is of importance considering that decreased DNA methylation in the brain has been linked with functional decline in learning and memory. We further show that sevoflurane exposure induces upregulation of Arc and JunB mRNA expression, 42.7% and 35.2%, respectively. Transgenerational changes in Arc and JunB mRNA were sexually dimorphic only occurring in males born to exposed females, expressed as upregulation of Arc and JunB mRNA, 71.6% and 74.0%, respectively. We further investigated correlation between altered arc promoter methylation and observed upregulation of Arc mRNA and observed that sevoflurane reduced methylation in the 5-upstream promoter region of females exposed to sevoflurane. Transgenerational hypomethylation and modifications to IEGs crucial to synaptic plasticity, observed following neonatal sevoflurane exposure could contribute to morphological and cognitive deficits known to occur with neonatal sevoflurane exposure.


Subject(s)
Genes, Immediate-Early/drug effects , Memory/drug effects , Neuronal Plasticity/drug effects , Sevoflurane/pharmacology , Anesthetics, Inhalation/pharmacology , Animals , Animals, Newborn , Cognition Disorders/metabolism , Dendritic Spines/metabolism , Hippocampus/metabolism , Learning/drug effects , Methyl Ethers/pharmacology , Rats, Sprague-Dawley , Up-Regulation/drug effects
11.
Proc Natl Acad Sci U S A ; 117(38): 23304-23310, 2020 09 22.
Article in English | MEDLINE | ID: mdl-31636216

ABSTRACT

The induction of immediate-early gene (IEG) expression in brain nuclei in response to an experience is necessary for the formation of long-term memories. Additionally, the rapid dynamics of IEG induction and decay motivates the common use of IEG expression as markers for identification of neuronal assemblies ("ensembles") encoding recent experience. However, major gaps remain in understanding the rules governing the distribution of IEGs within neuronal assemblies. Thus, the extent of correlation between coexpressed IEGs, the cell specificity of IEG expression, and the spatial distribution of IEG expression have not been comprehensively studied. To address these gaps, we utilized quantitative multiplexed single-molecule fluorescence in situ hybridization (smFISH) and measured the expression of IEGs (Arc, Egr2, and Nr4a1) within spiny projection neurons (SPNs) in the dorsal striatum of mice following acute exposure to cocaine. Exploring the relevance of our observations to other brain structures and stimuli, we also analyzed data from a study of single-cell RNA sequencing of mouse cortical neurons. We found that while IEG expression is graded, the expression of multiple IEGs is tightly correlated at the level of individual neurons. Interestingly, we observed that region-specific rules govern the induction of IEGs in SPN subtypes within striatal subdomains. We further observed that IEG-expressing assemblies form spatially defined clusters within which the extent of IEG expression correlates with cluster size. Together, our results suggest the existence of IEG-expressing neuronal "superensembles," which are associated in spatial clusters and characterized by coherent and robust expression of multiple IEGs.


Subject(s)
Brain/metabolism , Genes, Immediate-Early , Neurons/metabolism , Animals , Behavior, Animal , Brain/drug effects , Brain/growth & development , Cocaine/pharmacology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Gene Expression , Genes, Immediate-Early/drug effects , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Single Molecule Imaging
12.
Neurochem Res ; 44(9): 2139-2146, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31376054

ABSTRACT

Morphine can influence immediate early genes (IEG) of activity-regulated cytoskeletal-associated protein (Arc) and brain-derived neurotrophic factor (BDNF) which are activated in response to physiological stimuli during learning, as well as the nerve growth factor (NGF) gene which increases the expression of several IEGs for memory formation. The purpose of the current study was first to evaluate the effect of acute (1-day) and subchronic (15-days) morphine administration on memory retrieval of rats and second to determine the hippocampal expression of NGF, BDNF and Arc genes as potential contributors in the observed effects in each setting. The effects of morphine (intraperitoneal, 10, 15 and 20 mg/kg) on memory function and gene expression were assessed using inhibitory avoidance test and real-time polymerase chain reaction, respectively. We found that a single dose of morphine at the highest dose of 20 mg/kg decreases the post-training step-through-latency, while repeated administration of the same dose for 15 successive days increases this indicator of memory retrieval. We did not detect a significant change in the hippocampal expression of Arc, BDNF or NGF genes after a single episode of morphine treatment. However, subchronic morphine administration (15 and 20 mg/kg) increased the expression of Arc and BDNF genes in a dose dependent manner. A higher mRNA expression for the NGF was observed at the higher dose of 20 mg/kg. We hypothesize that the subchronic effects were morphine-induced behavioral sensitization which may have been enhanced through increased hippocampal Arc expression.


Subject(s)
Gene Expression/drug effects , Hippocampus/metabolism , Learning/drug effects , Memory/drug effects , Morphine/pharmacology , RNA, Messenger/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Cytoskeletal Proteins/genetics , Genes, Immediate-Early/drug effects , Male , Nerve Growth Factor/genetics , Nerve Tissue Proteins/genetics , Rats, Wistar
13.
Psychopharmacology (Berl) ; 236(12): 3525-3539, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31280332

ABSTRACT

RATIONALE: Cocaine base paste (CBP) is an illegal drug of abuse usually consumed by adolescents in a socio-economically vulnerable situation. Repeated drug use targets key brain circuits disrupting the processes that underlie emotions and cognition. At the basis of such neuroadaptations lie changes in the expression of immediate-early genes (IEGs). Nevertheless, changes in transcriptional regulation associated with CBP consumption remain unknown. OBJECTIVES: We aimed to describe behavioral phenotype related to locomotion, anxiety-like behavior, and memory of CBP-injected mice and to study IEGs expression after an abstinence period. METHODS: Five-week-old female CF-1 mice were i.p. injected daily with vehicle or CBP (40 mg/kg) for 10 days and subjected to a 10-day period of abstinence. Open field and novel object recognition tests were used to evaluate locomotion and anxiety-like behaviors and recognition memory, respectively, during chronic administration and after abstinence. After abstinence, prefrontal cortex (mPFC) and nucleus accumbens (NAc) were isolated and gene expression analysis performed through real-time PCR. RESULTS: We found an increase in locomotion and anxiety-like behavior during CBP administration and after the abstinence period. Furthermore, the CBP group showed impaired recognition memory after abstinence. Egr1, FosB, ΔFosB, Arc, Bdnf, and TrkB expression was upregulated in CBP-injected mice in NAc and FosB, ΔFosB, Arc, and Npas4 expression was downregulated in mPFC. We generated an anxiety score and found positive and negative correlations with IEGs expression in NAc and mPFC, respectively. CONCLUSION: Our results suggest that chronic CBP exposure induced alterations in anxiety-like behavior and recognition memory. These changes were accompanied by altered IEGs expression.


Subject(s)
Anxiety/chemically induced , Anxiety/metabolism , Cocaine/administration & dosage , Genes, Immediate-Early/physiology , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Animals , Anxiety/psychology , Cocaine/toxicity , Dopamine Uptake Inhibitors/administration & dosage , Dopamine Uptake Inhibitors/toxicity , Female , Gene Expression Regulation , Genes, Immediate-Early/drug effects , Locomotion/drug effects , Locomotion/physiology , Mice , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects
14.
Neuroscience ; 415: 173-183, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31351142

ABSTRACT

Addiction to prescribed opioids including oxycodone has reached tragic levels. Herein, we investigated the relevance of fibroblast growth factors (FGFs) and immediate early genes (IEGs) to withdrawal-induced incubation of drug craving following escalated oxycodone self-administration (SA). Rats were trained to self-administer oxycodone for 4 weeks. Seeking tests were performed at various intervals during 1 month of drug withdrawal. Rats were euthanized 1 day after the last test and nucleus accumbens and dorsal striata were dissected for use in PCR analyses. Rats given long access (LgA, 9 h), but not short access (ShA, 3 h) to drug escalated their oxycodone intake and exhibited incubation of oxycodone seeking during withdrawal. These rats exhibited dose-dependent increases in fgf2 expression in the dorsal striatum. Fgfr2 expression was also significantly increased in the striatum in LgA, but not ShA groups. Similarly, striatal c-fos and junB mRNA levels showed greater increases in LgA rats. The observations that fgf mRNA levels were more altered in the dorsal striatum than in the NAc of LgA rats suggest that changes in striatal FGF expression may be more salient to incubation of oxycodone craving than alterations in the NAc. Targeting FGF signaling pathways might offer novel strategies against opioid addiction.


Subject(s)
Fibroblast Growth Factors/drug effects , Genes, Immediate-Early/drug effects , Opioid-Related Disorders/metabolism , Oxycodone/metabolism , Animals , Behavior, Addictive , Corpus Striatum/metabolism , Craving , Fibroblast Growth Factors/metabolism , Male , Models, Animal , Nucleus Accumbens/metabolism , Oxycodone/administration & dosage , RNA, Messenger , Rats , Rats, Sprague-Dawley , Self Administration , Substance Withdrawal Syndrome
15.
Neurobiol Learn Mem ; 163: 107030, 2019 09.
Article in English | MEDLINE | ID: mdl-31185278

ABSTRACT

Neonatal ethanol exposure during the third trimester equivalent of human pregnancy in the rat significantly impairs hippocampal and prefrontal neurobehavioral functioning. Postnatal day [PD] 4-9 ethanol exposure in rats disrupts long-term context memory formation, resulting in abolished post-shock and retention test freezing in a variant of contextual fear conditioning called the Context Preexposure Facilitation Effect (CPFE). This behavioral impairment is accompanied by disrupted medial prefrontal, but not dorsal hippocampal expression of the immediate early genes (IEGs) c-Fos, Arc, Egr-1, and Npas4 (Heroux, Robinson-Drummer, Kawan, Rosen, & Stanton, 2019). The current experiment examined if systemic administration of the acetylcholinesterase inhibitor physostigmine (PHY) prior to context learning would rescue prefrontal IEG expression and freezing in the CPFE. From PD4-9, Long-Evans rats received oral intubation of ethanol (EtOH; 5.25 g/kg/day) or sham-intubation (SI). Rats received a systemic injection of saline (SAL) or PHY (0.01 mg/kg) prior to all three phases (Experiment 1) or just context exposure (Experiment 2) in the CPFE from PD31-33. A subset of rats were sacrificed 30 min after context learning to assay changes in IEG expression in the medial prefrontal cortex (mPFC), dorsal hippocampus (dHPC), and ventral hippocampus (vHPC). Administration of PHY prior to all three phases or just context learning rescued both post-shock and retention test freezing in the CPFE in EtOH rats without altering performance in SI rats. EtOH-SAL rats had significantly reduced mPFC but not dHPC expression of c-Fos, Arc, Egr-1, and Npas4. EtOH-PHY treatment rescued mPFC expression of c-Fos in ethanol-exposed rats and increased Arc and Npas4 regardless of dosing condition. While there was no effect of PHY on dHPC or vHPC expression of Arc, Egr-1, or Npas4, this treatment significantly boosted hippocampal expression of c-Fos regardless of ethanol treatment. These findings implicate impaired cholinergic and prefrontal function in cognitive deficits arising from 3rd-trimester equivalent alcohol exposure.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Conditioning, Classical/drug effects , Ethanol/toxicity , Physostigmine/pharmacology , Prenatal Exposure Delayed Effects/chemically induced , Animals , Animals, Newborn , Female , Genes, Immediate-Early/drug effects , Hippocampus/drug effects , Hippocampus/physiology , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Pregnancy , Prenatal Exposure Delayed Effects/drug therapy , Rats , Rats, Long-Evans , Real-Time Polymerase Chain Reaction
16.
Neuropharmacology ; 153: 13-19, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30998946

ABSTRACT

Propensity to relapse following long periods of abstinence is a key feature of substance use disorder. Drugs of abuse, such as cocaine, cause long-term changes in the neural circuitry regulating reward, motivation, and memory processes through dysregulation of various molecular mechanisms, including epigenetic regulation of activity-dependent gene expression. Underlying drug-induced changes to neural circuit function are the molecular mechanisms regulating activity-dependent gene expression. Of note, histone acetyltransferases and histone deacetylases (HDACs), powerful epigenetic regulators of gene expression, are dysregulated following both acute and chronic cocaine exposure and are linked to cocaine-induced changes in neural circuit function. To better understand the effect of drug-induced changes on epigenetic function and behavior, we investigated HDAC3-mediated regulation of Nr4a2/Nurr1 in the medial habenula, an understudied pathway in cocaine-associated behaviors. Nr4a2, a transcription factor critical in cocaine-associated behaviors and necessary for MHb development, is enriched in the cholinergic cell-population of the MHb; yet, the role of NR4A2 within the MHb in the adult brain remains elusive. Here, we evaluated whether epigenetic regulation of Nr4a2 in the MHb has a role in reinstatement of cocaine-associated behaviors. We found that HDAC3 disengages from Nr4a2 in the MHb in response to cocaine-primed reinstatement. Whereas enhancing HDAC3 function in the MHb had no effect on reinstatement, we found, using a dominant-negative splice variant (NURR2C), that loss of NR4A2 function in the MHb blocked reinstatement behaviors. These results show for the first time that regulation of NR4A2 function in the MHb is critical in relapse-like behaviors.


Subject(s)
Cocaine/administration & dosage , Drug-Seeking Behavior/physiology , Epigenesis, Genetic/physiology , Genes, Immediate-Early/physiology , Habenula/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Animals , Epigenesis, Genetic/drug effects , Female , Genes, Immediate-Early/drug effects , Habenula/drug effects , Histone Deacetylases/metabolism , Male , Mice , Mice, Transgenic
17.
Horm Behav ; 108: 94-104, 2019 02.
Article in English | MEDLINE | ID: mdl-29499221

ABSTRACT

The peripartum period is associated with the onset of behaviors that shelter, feed and protect young offspring from harm. The neural pathway that regulates caregiving behaviors has been mapped in female rats and is conserved in mice. However, rats rely on late gestational hormones to shift their perception of infant cues from aversive to attractive, whereas laboratory mice are "spontaneously" maternal, but their level of responding depends on experience. For example, pup-naïve virgin female mice readily care for pups in the home cage, but avoid pups in a novel environment. In contrast, pup-experienced virgin mice care for pups in both contexts. Thus, virgin mice rely on experience to shift their perception of infant cues from aversive to attractive in a novel context. We hypothesize that alterations in immediate early gene activation may underlie the experience-driven shift in which neural pathways (fear/avoidance versus maternal/approach) are activated by pups to modulate context-dependent changes in maternal responding. Here we report that the effects of sodium butyrate, a drug that allows for an amplification of experience-induced histone acetylation and gene expression in virgins, are comparable to the natural onset of caregiving behaviors in postpartum mice and induce postpartum-like patterns of immediate early gene expression across brain regions. These data suggest that pups can activate a fear/defensive circuit in mice and experience-driven improvements in caregiving behavior could be regulated in part through decreased activation of this pathway.


Subject(s)
Behavior, Animal/drug effects , Genes, Immediate-Early/drug effects , Histone Deacetylase Inhibitors/pharmacology , Maternal Behavior/drug effects , Neural Pathways/drug effects , Postpartum Period/drug effects , Animals , Animals, Newborn , Cues , Female , Maternal Behavior/physiology , Mice , Mice, Inbred C57BL , Neural Pathways/metabolism , Parity/drug effects , Parity/genetics , Postpartum Period/physiology , Postpartum Period/psychology , Pregnancy , Up-Regulation/drug effects , Up-Regulation/genetics
18.
Neurochem Int ; 124: 10-18, 2019 03.
Article in English | MEDLINE | ID: mdl-30557593

ABSTRACT

Although some of the clinical manifestations of substance use disorders might be superficially similar, it is highly likely that different classes of abused drugs including opioids (heroin, morphine, and oxycodone, other opioids) and psychostimulants (cocaine and amphetamines) cause different neuroadaptations in various brain regions dependent in the distribution and concentration of their biochemical sites of actions. In fact, different molecular networks are indeed impacted by acute and chronic administration of addictive substances. Some of the genes whose expression is influenced by the administration of these substances are immediate-early genes (IEGs). IEGs include classes of low expression genes that can become very highly induced within seconds or minutes of activation by endogenous or exogenous stimuli. These IEGs might play important roles in activating target genes that regulate adaptations implicated in the behavioral manifestations diagnosed as addiction. Therefore, the purpose of this review is to provide an overview of recent data on the effects of psychostimulants and opioids on IEG expression in the brain. The review documents some contrasting effects of these classes of drugs on gene expression and indicates that further studies are necessary to identify the specific effects of each drug class when trying to predict clinical responses to therapeutic agents.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/metabolism , Central Nervous System Stimulants/pharmacology , Genes, Immediate-Early/physiology , Nerve Net/metabolism , Reward , Animals , Brain/drug effects , Gene Expression , Genes, Immediate-Early/drug effects , Humans , Nerve Net/drug effects
19.
Neurochem Res ; 43(12): 2460-2472, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30426349

ABSTRACT

Upon synaptic stimulation and glutamate release, glutamate receptors are activated to regulate several downstream effectors and signaling pathways resulting in synaptic modification. One downstream intracellular effect, in particular, is the expression of immediate-early genes (IEGs), which have been proposed to be important in synaptic plasticity because of their rapid expression following synaptic activation and key role in memory formation. In this study, we screened a natural compound library in order to find a compound that could induce the expression of IEGs in primary cortical neurons and discovered that psoralidin, a natural compound isolated from the seeds of Psoralea corylifolia, stimulated synaptic modulation. Psoralidin activated mitogen-activated protein kinase (MAPK) signaling, which in turn induced the expression of neuronal IEGs, particularly Arc, Egr-1, and c-fos. N-methyl-D-aspartate (NMDA) receptors activation and extracellular calcium influx were implicated in the psoralidin-induced intracellular changes. In glutamate dose-response curve, psoralidin shifted glutamate EC50 to lower values without enhancing maximum activity. Interestingly, psoralidin increased the density, area, and intensity of excitatory synapses in primary hippocampal neurons, which were mediated by NMDA receptor activation and MAPK signaling. These results suggest that psoralidin triggers synaptic remodeling through activating NMDA receptor and subsequent MAPK signaling cascades and therefore could possibly serve as an NMDA receptor modulator.


Subject(s)
Benzofurans/pharmacology , Cerebral Cortex/metabolism , Coumarins/pharmacology , Genes, Immediate-Early/physiology , Neuronal Plasticity/physiology , Neurons/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression , Genes, Immediate-Early/drug effects , Mice , Mice, Inbred ICR , Neuronal Plasticity/drug effects , Neurons/drug effects , Pregnancy , Synapses/drug effects
20.
J Neurosci ; 38(34): 7516-7528, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30030395

ABSTRACT

Epigenetic mechanisms have gained increasing attention as regulators of synaptic plasticity and responsiveness to drugs of abuse. In particular, it has been shown that the activity of the DNA methyltransferase 3a (Dnmt3a) mediates certain long-lasting effects of cocaine. Here we examined the role of the Dnmt isoforms, Dnmt3a1 and Dnmt3a2, within the nucleus accumbens (NAc) on transcriptional activity of immediate early genes (IEGs) and acute and long-lasting responsiveness to cocaine and cocaine conditioned cues. Using primary striatal cultures, we show that transcription of Dnmt3a2, but not that of Dnmt3a1, is activated by dopamine D1 receptor signaling and that knockdown of Dnmt3a2 using viral vector-mediated expression of Dnmt3a2-specific shRNAs impairs induction of the IEGs, Arc, FosB, and Egr2 Acute cocaine administration increases expression of Dnmt3a2 but not that of Dnmt3a1 in the NAc shell. In contrast, in the NAc core, expression of Dnmt3a1 and Dnmt3a2 was unaffected by cocaine administration. shRNA-mediated knockdown of Dnmt3a2 in vivo impairs the induction of IEGs, including Egr2 and FosB indicating that Dnmt3a2 regulates cocaine-dependent expression of plasticity genes in the rat NAc shell. Cocaine self-administration experiments in rats revealed that Dnmt3a2 regulates drug cue memories that drive reinstatement of cocaine seeking as well as incubation of this phenomenon within the NAc shell. Dnmt3a2 does not influence the primary reinforcing effects of cocaine. Thus, Dnmt3a2 mediates long-lasting cocaine cue memories within the NAc shell. Targeting Dnmt3a2 expression or function may interfere with cocaine craving and relapse.SIGNIFICANCE STATEMENT In humans, drug craving can occur in response to conditioned cues, even after extended periods of abstinence. In rats, cue-induced cocaine seeking has been shown to increase progressively during the first 2 months of abstinence from drug self-administration. This phenomenon, referred to as incubation of cocaine seeking, is consistent with the hypothesis that in humans craving increases over time and remains high following prolonged abstinence. Those long-lasting behavioral changes are likely to be mediated by epigenetic effects and neuroplastic changes within the mesolimbic brain reward system. Here we show that a specific isoform of DNA-methyltransferases in the NAc shell regulates drug cue memories that drive reinstatement of cocaine seeking after both early abstinence and incubation of cocaine craving.


Subject(s)
Cocaine-Related Disorders/enzymology , Craving/drug effects , DNA (Cytosine-5-)-Methyltransferases/physiology , Nerve Tissue Proteins/physiology , Nucleus Accumbens/enzymology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Benzazepines/pharmacology , Cocaine/administration & dosage , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/physiopathology , Conditioning, Operant/drug effects , Cues , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Enzyme Induction/drug effects , Extinction, Psychological/drug effects , Genes, Immediate-Early/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nucleus Accumbens/drug effects , Protein Isoforms/physiology , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/physiology , Self Administration , Substance Withdrawal Syndrome/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...