The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors

METH use causes neuroadaptations that negatively impact the prefrontal cortex (PFC) leading to addiction and associated cognitive decline in animals and humans. In contrast, modafinil enhances cognition by increasing PFC function. Accumulated evidence indicates that psychostimulant drugs, including...

Descripción completa

Guardado en:
Detalles Bibliográficos
Publicado: 2019
Materias:
Acceso en línea:https://bibliotecadigital.exactas.uba.ar/collection/paper/document/paper_02785846_v88_n_p222_Gonzalez
http://hdl.handle.net/20.500.12110/paper_02785846_v88_n_p222_Gonzalez
Aporte de:
id paper:paper_02785846_v88_n_p222_Gonzalez
record_format dspace
institution Universidad de Buenos Aires
institution_str I-28
repository_str R-134
collection Biblioteca Digital - Facultad de Ciencias Exactas y Naturales (UBA)
topic Dopamine receptors
Glutamate
Histone acetylation
Methamphetamine
Modafinil
Prefrontal cortex
8 chloro 2,3,4,5 tetrahydro 3 methyl 5 phenyl 1h 3 benzazepin 7 ol hydrogen maleate
biological marker
dopamine receptor
glutamic acid
histone deacetylase 1
histone deacetylase 2
histone H3
histone H4
messenger RNA
methamphetamine
modafinil
n methyl dextro aspartic acid
n methyl dextro aspartic acid GluN1
raclopride
unclassified drug
benzazepine derivative
central stimulant agent
dopamine receptor
dopamine receptor stimulating agent
histone
histone deacetylase 1
methamphetamine
modafinil
raclopride
receptor
Adra1a gene
Adra1b gene
animal cell
animal experiment
animal tissue
Article
controlled study
DA gene
Drd1 gene
Drd2 gene
epigenetics
excitatory postsynaptic potential
gene
gene expression
glutamate receptor gene
glutamatergic synapse
Grin1 gene
Hcrtr1 gene
Hcrtr2 gene
histamine gene
histone acetylation
Hrh1 gene
Hrh3 gene
male
medial prefrontal cortex
mouse
NE gene
neurotransmission
nonhuman
orexin gene
promoter region
protein analysis
protein expression
protein function
pyramidal nerve cell
synaptic transmission
whole cell patch clamp
animal
C57BL mouse
chromatin immunoprecipitation
cytology
drug effect
genetic epigenesis
genetics
metabolism
nerve cell
patch clamp technique
physiology
prefrontal cortex
transgenic mouse
Animals
Benzazepines
Central Nervous System Stimulants
Chromatin Immunoprecipitation
Dopamine Agents
Epigenesis, Genetic
Excitatory Postsynaptic Potentials
Histone Deacetylase 1
Histones
Male
Methamphetamine
Mice
Mice, Inbred C57BL
Mice, Transgenic
Modafinil
Neurons
Patch-Clamp Techniques
Prefrontal Cortex
Raclopride
Receptors, Biogenic Amine
Receptors, Dopamine
spellingShingle Dopamine receptors
Glutamate
Histone acetylation
Methamphetamine
Modafinil
Prefrontal cortex
8 chloro 2,3,4,5 tetrahydro 3 methyl 5 phenyl 1h 3 benzazepin 7 ol hydrogen maleate
biological marker
dopamine receptor
glutamic acid
histone deacetylase 1
histone deacetylase 2
histone H3
histone H4
messenger RNA
methamphetamine
modafinil
n methyl dextro aspartic acid
n methyl dextro aspartic acid GluN1
raclopride
unclassified drug
benzazepine derivative
central stimulant agent
dopamine receptor
dopamine receptor stimulating agent
histone
histone deacetylase 1
methamphetamine
modafinil
raclopride
receptor
Adra1a gene
Adra1b gene
animal cell
animal experiment
animal tissue
Article
controlled study
DA gene
Drd1 gene
Drd2 gene
epigenetics
excitatory postsynaptic potential
gene
gene expression
glutamate receptor gene
glutamatergic synapse
Grin1 gene
Hcrtr1 gene
Hcrtr2 gene
histamine gene
histone acetylation
Hrh1 gene
Hrh3 gene
male
medial prefrontal cortex
mouse
NE gene
neurotransmission
nonhuman
orexin gene
promoter region
protein analysis
protein expression
protein function
pyramidal nerve cell
synaptic transmission
whole cell patch clamp
animal
C57BL mouse
chromatin immunoprecipitation
cytology
drug effect
genetic epigenesis
genetics
metabolism
nerve cell
patch clamp technique
physiology
prefrontal cortex
transgenic mouse
Animals
Benzazepines
Central Nervous System Stimulants
Chromatin Immunoprecipitation
Dopamine Agents
Epigenesis, Genetic
Excitatory Postsynaptic Potentials
Histone Deacetylase 1
Histones
Male
Methamphetamine
Mice
Mice, Inbred C57BL
Mice, Transgenic
Modafinil
Neurons
Patch-Clamp Techniques
Prefrontal Cortex
Raclopride
Receptors, Biogenic Amine
Receptors, Dopamine
The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors
topic_facet Dopamine receptors
Glutamate
Histone acetylation
Methamphetamine
Modafinil
Prefrontal cortex
8 chloro 2,3,4,5 tetrahydro 3 methyl 5 phenyl 1h 3 benzazepin 7 ol hydrogen maleate
biological marker
dopamine receptor
glutamic acid
histone deacetylase 1
histone deacetylase 2
histone H3
histone H4
messenger RNA
methamphetamine
modafinil
n methyl dextro aspartic acid
n methyl dextro aspartic acid GluN1
raclopride
unclassified drug
benzazepine derivative
central stimulant agent
dopamine receptor
dopamine receptor stimulating agent
histone
histone deacetylase 1
methamphetamine
modafinil
raclopride
receptor
Adra1a gene
Adra1b gene
animal cell
animal experiment
animal tissue
Article
controlled study
DA gene
Drd1 gene
Drd2 gene
epigenetics
excitatory postsynaptic potential
gene
gene expression
glutamate receptor gene
glutamatergic synapse
Grin1 gene
Hcrtr1 gene
Hcrtr2 gene
histamine gene
histone acetylation
Hrh1 gene
Hrh3 gene
male
medial prefrontal cortex
mouse
NE gene
neurotransmission
nonhuman
orexin gene
promoter region
protein analysis
protein expression
protein function
pyramidal nerve cell
synaptic transmission
whole cell patch clamp
animal
C57BL mouse
chromatin immunoprecipitation
cytology
drug effect
genetic epigenesis
genetics
metabolism
nerve cell
patch clamp technique
physiology
prefrontal cortex
transgenic mouse
Animals
Benzazepines
Central Nervous System Stimulants
Chromatin Immunoprecipitation
Dopamine Agents
Epigenesis, Genetic
Excitatory Postsynaptic Potentials
Histone Deacetylase 1
Histones
Male
Methamphetamine
Mice
Mice, Inbred C57BL
Mice, Transgenic
Modafinil
Neurons
Patch-Clamp Techniques
Prefrontal Cortex
Raclopride
Receptors, Biogenic Amine
Receptors, Dopamine
description METH use causes neuroadaptations that negatively impact the prefrontal cortex (PFC) leading to addiction and associated cognitive decline in animals and humans. In contrast, modafinil enhances cognition by increasing PFC function. Accumulated evidence indicates that psychostimulant drugs, including modafinil and METH, regulate gene expression via epigenetic modifications. In this study, we measured the effects of single-dose injections of modafinil and METH on the protein levels of acetylated histone H3 (H3ac) and H4ac, deacetylases HDAC1 and HDAC2, and of the NMDA subunit GluN1 in the medial PFC (mPFC) of mice euthanized 1 h after drug administration. To test if dopamine (DA) receptors (DRs) participate in the biochemical effects of the two drugs, we injected the D1Rs antagonist, SCH23390, or the D2Rs antagonist, raclopride, 30 min before administration of METH and modafinil. We evaluated each drug effect on glutamate synaptic transmission in D1R-expressing layer V pyramidal neurons. We also measured the enrichment of H3ac and H4ac at the promoters of several genes including DA, NE, orexin, histamine, and glutamate receptors, and their mRNA expression, since they are responsive to chronic modafinil and METH treatment. Acute modafinil and METH injections caused similar effects on total histone acetylation, increasing H3ac and decreasing H4ac, and they also increased HDAC1, HDAC2 and GluN1 protein levels in the mouse mPFC. In addition, the effects of the drugs were prevented by pre-treatment with D1Rs and D2Rs antagonists. Specifically, the changes in H4ac, HDAC2, and GluN1 were responsive to SCH23390, whereas those of H3ac and GluN1 were responsive to raclopride. Whole-cell patch clamp in transgenic BAC-Drd1a-tdTomato mice showed that METH, but not modafinil, induced paired-pulse facilitation of EPSCs, suggesting reduced presynaptic probability of glutamate release onto layer V pyramidal neurons. Analysis of histone 3/4 enrichment at specific promoters revealed: i) distinct effects of the drugs on histone 3 acetylation, with modafinil increasing H3ac at Drd1 and Adra1b promoters, but METH increasing H3ac at Adra1a; ii) distinct effects on histone 4 acetylation enrichment, with modafinil increasing H4ac at the Drd2 promoter and decreasing it at Hrh1, but METH increasing H4ac at Drd1; iii) comparable effects of both psychostimulants, increasing H3ac at Drd2, Hcrtr1, and Hrh1 promoters, decreasing H3ac at Hrh3, increasing H4ac at Hcrtr1, and decreasing H4ac at Hcrtr2, Hrh3, and Grin1 promoters. Interestingly, only METH altered mRNA levels of genes with altered histone acetylation status, inducing increased expression of Drd1a, Adra1a, Hcrtr1, and Hrh1, and decreasing Grin1. Our study suggests that although acute METH and modafinil can both increase DA neurotransmission in the mPFC, there are similar and contrasting epigenetic and transcriptional consequences that may account for their divergent clinical effects. © 2018
title The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors
title_short The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors
title_full The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors
title_fullStr The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors
title_full_unstemmed The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors
title_sort effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors
publishDate 2019
url https://bibliotecadigital.exactas.uba.ar/collection/paper/document/paper_02785846_v88_n_p222_Gonzalez
http://hdl.handle.net/20.500.12110/paper_02785846_v88_n_p222_Gonzalez
_version_ 1768542737688166400
spelling paper:paper_02785846_v88_n_p222_Gonzalez2023-06-08T15:26:51Z The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors Dopamine receptors Glutamate Histone acetylation Methamphetamine Modafinil Prefrontal cortex 8 chloro 2,3,4,5 tetrahydro 3 methyl 5 phenyl 1h 3 benzazepin 7 ol hydrogen maleate biological marker dopamine receptor glutamic acid histone deacetylase 1 histone deacetylase 2 histone H3 histone H4 messenger RNA methamphetamine modafinil n methyl dextro aspartic acid n methyl dextro aspartic acid GluN1 raclopride unclassified drug benzazepine derivative central stimulant agent dopamine receptor dopamine receptor stimulating agent histone histone deacetylase 1 methamphetamine modafinil raclopride receptor Adra1a gene Adra1b gene animal cell animal experiment animal tissue Article controlled study DA gene Drd1 gene Drd2 gene epigenetics excitatory postsynaptic potential gene gene expression glutamate receptor gene glutamatergic synapse Grin1 gene Hcrtr1 gene Hcrtr2 gene histamine gene histone acetylation Hrh1 gene Hrh3 gene male medial prefrontal cortex mouse NE gene neurotransmission nonhuman orexin gene promoter region protein analysis protein expression protein function pyramidal nerve cell synaptic transmission whole cell patch clamp animal C57BL mouse chromatin immunoprecipitation cytology drug effect genetic epigenesis genetics metabolism nerve cell patch clamp technique physiology prefrontal cortex transgenic mouse Animals Benzazepines Central Nervous System Stimulants Chromatin Immunoprecipitation Dopamine Agents Epigenesis, Genetic Excitatory Postsynaptic Potentials Histone Deacetylase 1 Histones Male Methamphetamine Mice Mice, Inbred C57BL Mice, Transgenic Modafinil Neurons Patch-Clamp Techniques Prefrontal Cortex Raclopride Receptors, Biogenic Amine Receptors, Dopamine METH use causes neuroadaptations that negatively impact the prefrontal cortex (PFC) leading to addiction and associated cognitive decline in animals and humans. In contrast, modafinil enhances cognition by increasing PFC function. Accumulated evidence indicates that psychostimulant drugs, including modafinil and METH, regulate gene expression via epigenetic modifications. In this study, we measured the effects of single-dose injections of modafinil and METH on the protein levels of acetylated histone H3 (H3ac) and H4ac, deacetylases HDAC1 and HDAC2, and of the NMDA subunit GluN1 in the medial PFC (mPFC) of mice euthanized 1 h after drug administration. To test if dopamine (DA) receptors (DRs) participate in the biochemical effects of the two drugs, we injected the D1Rs antagonist, SCH23390, or the D2Rs antagonist, raclopride, 30 min before administration of METH and modafinil. We evaluated each drug effect on glutamate synaptic transmission in D1R-expressing layer V pyramidal neurons. We also measured the enrichment of H3ac and H4ac at the promoters of several genes including DA, NE, orexin, histamine, and glutamate receptors, and their mRNA expression, since they are responsive to chronic modafinil and METH treatment. Acute modafinil and METH injections caused similar effects on total histone acetylation, increasing H3ac and decreasing H4ac, and they also increased HDAC1, HDAC2 and GluN1 protein levels in the mouse mPFC. In addition, the effects of the drugs were prevented by pre-treatment with D1Rs and D2Rs antagonists. Specifically, the changes in H4ac, HDAC2, and GluN1 were responsive to SCH23390, whereas those of H3ac and GluN1 were responsive to raclopride. Whole-cell patch clamp in transgenic BAC-Drd1a-tdTomato mice showed that METH, but not modafinil, induced paired-pulse facilitation of EPSCs, suggesting reduced presynaptic probability of glutamate release onto layer V pyramidal neurons. Analysis of histone 3/4 enrichment at specific promoters revealed: i) distinct effects of the drugs on histone 3 acetylation, with modafinil increasing H3ac at Drd1 and Adra1b promoters, but METH increasing H3ac at Adra1a; ii) distinct effects on histone 4 acetylation enrichment, with modafinil increasing H4ac at the Drd2 promoter and decreasing it at Hrh1, but METH increasing H4ac at Drd1; iii) comparable effects of both psychostimulants, increasing H3ac at Drd2, Hcrtr1, and Hrh1 promoters, decreasing H3ac at Hrh3, increasing H4ac at Hcrtr1, and decreasing H4ac at Hcrtr2, Hrh3, and Grin1 promoters. Interestingly, only METH altered mRNA levels of genes with altered histone acetylation status, inducing increased expression of Drd1a, Adra1a, Hcrtr1, and Hrh1, and decreasing Grin1. Our study suggests that although acute METH and modafinil can both increase DA neurotransmission in the mPFC, there are similar and contrasting epigenetic and transcriptional consequences that may account for their divergent clinical effects. © 2018 2019 https://bibliotecadigital.exactas.uba.ar/collection/paper/document/paper_02785846_v88_n_p222_Gonzalez http://hdl.handle.net/20.500.12110/paper_02785846_v88_n_p222_Gonzalez