UB Vice President for Research and Economic Development markup
David Dietz UB Profile
Pharmacology and Toxicology
Professor
Chair
955 Main Street
Room 3102G
Buffalo NY 14203
716-829-2071
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Ilaria Ceglia, Ko-Woon Lee, Michael E Cahill, Steven M Graves, David Dietz, Dalton J Surmeier, Eric J Nestler, Angus C Nairn, Paul Greengard, Yong Kim
Keywords: Cdk5 WAVE1 cocaine medium spiny projection neurons striatum
Abstract: Wiskott-Aldrich syndrome protein (WASP) family verprolin homologous protein 1 (WAVE1) regulates actin-related protein 2/3 (Arp2/3) complex-mediated actin polymerization. Our previous studies have found WAVE1 to be inhibited by Cdk5-mediated phosphorylation in brain and to play a role in the regulation of dendritic spine morphology. Here we report that mice in which WAVE1 was knocked out (KO) in neurons expressing the D1 dopamine receptor (D1-KO), but not mice where WAVE1 was knocked out in neurons expressing the D2 dopamine receptor (D2-KO), exhibited a significant decrease in place preference associated with cocaine. In contrast to wild-type (WT) and WAVE1 D2-KO mice, cocaine-induced sensitized locomotor behavior was not maintained in WAVE1 D1-KO mice. After chronic cocaine administration and following withdrawal, an acute cocaine challenge induced WAVE1 activation in striatum, which was assessed by dephosphorylation. The cocaine-induced WAVE1 dephosphorylation was attenuated by coadministration of either a D1 dopamine receptor or NMDA glutamate receptor antagonist. Upon an acute challenge of cocaine following chronic cocaine exposure and withdrawal, we also observed in WT, but not in WAVE1 D1-KO mice, a decrease in dendritic spine density and a decrease in the frequency of excitatory postsynaptic AMPA receptor currents in medium spiny projection neurons expressing the D1 dopamine receptor (D1-MSNs) in the nucleus accumbens. These results suggest that WAVE1 is involved selectively in D1-MSNs in cocaine-evoked neuronal activity-mediated feedback regulation of glutamatergic synapses.
PubMedId: 28115704
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 114, 2017
Authors: Jennifer A Martin, Aaron Caccamise, Craig T Werner, Rathipriya Viswanathan, Jessie J Polanco, Andrew F Stewart, Shruthi A Thomas, Fraser J Sim, David M Dietz
Abstract: Opiate abuse and addiction have become a worldwide epidemic with great societal and financial burdens, highlighting a critical need to understand the neurobiology of opiate addiction. While several studies have focused on drug-dependent changes in neurons, the role of glia in opiate addiction remains largely unstudied. RNA sequencing pathway analysis from the prefrontal cortex of male rats revealed changes in several genes associated with oligodendrocyte differentiation and maturation following heroin self-administration. Among these genes changed was Sox10, which is regulated, in part, by the chromatin remodeler BRG1/SMARCA4. To directly test the functional role of Sox10 in mediating heroin-induced behavioral plasticity, we selectively overexpressed Sox10 and BRG1 in the prefrontal cortex. Overexpression of either Sox10 or BRG1 decreased the motivation to obtain heroin infusions in a progressive ratio test without altering the acquisition or maintenance of heroin self-administration. These data demonstrate a critical, and perhaps compensatory, role of Sox10 and BRG1 in oligodendrocytes in regulating the motivation for heroin.Neuropsychopharmacology accepted article preview online, 20 December 2017. doi:10.1038/npp.2017.303.
PubMedId: 29260792
Journal-article: Neuropsychopharmacology, 2017
Authors: M E Cahill, D M Walker, A M Gancarz, Z J Wang, C K Lardner, R C Bagot, R L Neve, D M Dietz, E J Nestler
Abstract: The nucleus accumbens (NAc) is a primary brain reward region composed predominantly of medium spiny neurons (MSNs). In response to early withdrawal from repeated cocaine administration, de novo dendritic spine formation occurs in NAc MSNs. Much evidence indicates that this new spine formation facilitates the rewarding properties of cocaine. Early withdrawal from repeated cocaine also produces dramatic alterations in the transcriptome of NAc MSNs, but how such alterations influence cocaine's effects on dendritic spine formation remain unclear. Studies in non-neuronal cells indicate that actin cytoskeletal regulatory pathways in nuclei have a direct role in the regulation of gene transcription in part by controlling the access of co-activators to their transcription factor partners. In particular, actin state dictates the interaction between the serum response factor (SRF) transcription factor and one of its principal co-activators, MAL. Here we show that cocaine induces alterations in nuclear F-actin signaling pathways in the NAc with associated changes in the nuclear subcellular localization of SRF and MAL. Using in vivo optogenetics, the brain region-specific inputs to the NAc that mediate these nuclear changes are investigated. Finally, we demonstrate that regulated SRF expression, in turn, is critical for the effects of cocaine on dendritic spine formation and for cocaine-mediated behavioral sensitization. Collectively, these findings reveal a mechanism by which nuclear-based changes influence the structure of NAc MSNs in response to cocaine.Molecular Psychiatry advance online publication, 30 May 2017; doi:10.1038/mp.2017.116.
PubMedId: 28555077
Journal-article: Molecular Psychiatry, 2017
Authors: Diane M Damez-Werno, HaoSheng Sun, Kimberly N Scobie, Ningyi Shao, Jaclyn Rabkin, Caroline Dias, Erin S Calipari, Ian Maze, Catherine J Pena, Deena M Walker, Michael E Cahill, Ramesh Chandra, Amy Gancarz, Ezekiell Mouzon, Joseph A Landry, Hannah Cates, Mary-Kay Lobo, David Dietz, C David Allis, Ernesto Guccione, Gustavo Turecki, Paola Defilippi, Rachael L Neve, Yasmin L Hurd, Li Shen, Eric J Nestler
Keywords: ChIP-seq Src drug addiction histone arginine (R) methylation medium spiny neurons
Abstract: Repeated cocaine exposure regulates transcriptional regulation within the nucleus accumbens (NAc), and epigenetic mechanisms-such as histone acetylation and methylation on Lys residues-have been linked to these lasting actions of cocaine. In contrast to Lys methylation, the role of histone Arg (R) methylation remains underexplored in addiction models. Here we show that protein-R-methyltransferase-6 (PRMT6) and its associated histone mark, asymmetric dimethylation of R2 on histone H3 (H3R2me2a), are decreased in the NAc of mice and rats after repeated cocaine exposure, including self-administration, and in the NAc of cocaine-addicted humans. Such PRMT6 down-regulation occurs selectively in NAc medium spiny neurons (MSNs) expressing dopamine D2 receptors (D2-MSNs), with opposite regulation occurring in D1-MSNs, and serves to protect against cocaine-induced addictive-like behavioral abnormalities. Using ChIP-seq, we identified Src kinase signaling inhibitor 1 (Srcin1; also referred to as p140Cap) as a key gene target for reduced H3R2me2a binding, and found that consequent Srcin1 induction in the NAc decreases Src signaling, cocaine reward, and the motivation to self-administer cocaine. Taken together, these findings suggest that suppression of Src signaling in NAc D2-MSNs, via PRMT6 and H3R2me2a down-regulation, functions as a homeostatic brake to restrain cocaine action, and provide novel candidates for the development of treatments for cocaine addiction.
PubMedId: 27506785
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 113, 2016
Authors: Zi-Jun Wang, Jennifer A Martin, Lauren E Mueller, Aaron Caccamise, Craig T Werner, Rachael L Neve, Amy M Gancarz, Jun-Xu Li, David M Dietz
Keywords: BRG1 Chromatin remodeling Cocaine Cue-induced reinstatement SMAD3 Self-administration
Abstract: Drug addiction is defined as a chronic disease characterized by compulsive drug seeking and episodes of relapse despite prolonged periods of drug abstinence. Neurobiological adaptations, including transcriptional and epigenetic alterations in the nucleus accumbens, are thought to contribute to this life-long disease state. We previously demonstrated that the transcription factor SMAD3 is increased after 7 days of withdrawal from cocaine self-administration. However, it is still unknown which additional factors participate in the process of chromatin remodeling and facilitate the binding of SMAD3 to promoter regions of target genes. Here, we examined the possible interaction of BRG1-also known as SMARCA4, an adenosine triphosphatase-containing chromatin remodeler-and SMAD3 in response to cocaine exposure. The expression of BRG1, as well as its binding to SMAD3 and target gene promoter regions, was evaluated in the nucleus accumbens and dorsal striatum of rats using western blotting, co-immunoprecipitation, and chromatin immunoprecipitation following abstinence from cocaine self-administration. Rats were assessed for cocaine-seeking behaviors after either intra-accumbal injections of the BRG1 inhibitor PFI3 or viral-mediated overexpression of BRG1. After withdrawal from cocaine self-administration, BRG1 expression and complex formation with SMAD3 are increased in the nucleus accumbens, resulting in increased binding of BRG1 to the promoter regions of Ctnnb1, Mef2d, and Dbn1. Intra-accumbal infusion of PFI3 attenuated, whereas viral overexpression of Brg1 enhanced, cocaine-reinstatement behavior. BRG1 is a key mediator of the SMAD3-dependent regulation of cellular and behavioral plasticity that mediates cocaine seeking after a period of withdrawal.
PubMedId: 27422367
Journal-article: Biological psychiatry, vol. 80, 2016
Authors: Michael??E. Cahill, Rosemary??C. Bagot, Amy??M. Gancarz, Deena??M. Walker, HaoSheng Sun, Zi-Jun Wang, Elizabeth??A. Heller, Jian Feng, Pamela??J. Kennedy, Ja??Wook Koo, Hannah??M. Cates, Rachael??L. Neve, Li Shen, David??M. Dietz, Eric??J. Nestler
Abstract: Dendritic spines are the sites of most excitatory synapses in the CNS, and opposing alterations in the synaptic structure of medium spiny neurons (MSNs) of the nucleus accumbens (NAc), a primary brain reward region, are seen at early versus late time points after cocaine administration. Here we investigate the time-dependent molecular and biochemical processes that regulate this bidirectional synaptic structural plasticity of NAc MSNs and associated changes in cocaine reward in response to chronic cocaine exposure. Our findings reveal key roles for the bidirectional synaptic expression of the Rap1b small GTPase and an associated local synaptic protein translation network in this process. The transcriptional mechanisms and pathway-specific inputs to NAc that regulate Rap1b expression are also characterized. Collectively, these findings provide a precise mechanism by which nuclear to synaptic interactions induce "metaplasticity" in NAc MSNs, and we reveal the specific effects of this plasticity on reward behavior in a brain circuit-specific manner.
PubMedId: 26844834
Journal-article: Neuron, vol. 89, pp. 566-582, 2016
Authors: Allyson K. Friedman, Barbara Juarez, Stacy M. Ku, Hongxing Zhang, Rhodora C. Calizo, Jessica J. Walsh, Dipesh Chaudhury, Song Zhang, Angel Hawkins, David M. Dietz, James W. Murrough, Maria Ribadeneira, Erik H. Wong, Rachael L. Neve, Ming-Hu Han
Abstract: Less than half of patients suffering from major depressive disorder, a leading cause of disability worldwide, achieve remission with current antidepressants, making it imperative to develop more effective treatment. A new therapeutic direction is emerging from the increased understanding of natural resilience as an active stress-coping process. It is known that potassium (K(+)) channels in the ventral tegmental area (VTA) are an active mediator of resilience. However, no druggable targets have been identified to potentiate active resilience mechanisms. In the chronic social defeat stress model of depression, we report that KCNQ-type K(+) channel openers, including FDA-approved drug retigabine (ezogabine), show antidepressant efficacy. We demonstrate that overexpression of KCNQ channels in the VTA dopaminergic neurons and either local infusion or systemic administration of retigabine normalized neuronal hyperactivity and depressive behaviours. These findings identify KCNQ as a target for conceptually novel antidepressants that function through the potentiation of active resilience mechanisms.
PubMedId: 27216573
Journal-article: Nature Communications, vol. 7, pp. 11671, 2016
Authors: H. Sun, J. A. Martin, C. T. Werner, Z.-J. Wang, D. M. Damez-Werno, K. N. Scobie, N.-Y. Shao, C. Dias, J. Rabkin, J. W. Koo, A. M. Gancarz, E. A. Mouzon, R. L. Neve, L. Shen, D. M. Dietz, E. J. Nestler
Keywords: Addiction Baz1b chromatin depression epigenetics remodeler
Abstract: ATP-dependent chromatin remodeling proteins are being implicated increasingly in the regulation of complex behaviors, including models of several psychiatric disorders. Here, we demonstrate that Baz1b, an accessory subunit of the ISWI family of chromatin remodeling complexes, is upregulated in the nucleus accumbens (NAc), a key brain reward region, in both chronic cocaine-treated mice and mice that are resilient to chronic social defeat stress. In contrast, no regulation is seen in mice that are susceptible to this chronic stress. Viral-mediated overexpression of Baz1b, along with its associated subunit Smarca5, in mouse NAc is sufficient to potentiate both rewarding responses to cocaine, including cocaine self-administration, and resilience to chronic social defeat stress. However, despite these similar, proreward behavioral effects, genome-wide mapping of BAZ1B in NAc revealed mostly distinct subsets of genes regulated by these chromatin remodeling proteins after chronic exposure to either cocaine or social stress. Together, these findings suggest important roles for BAZ1B and its associated chromatin remodeling complexes in NAc in the regulation of reward behaviors to distinct emotional stimuli and highlight the stimulus-specific nature of the actions of these regulatory proteins. We show that BAZ1B, a component of chromatin remodeling complexes, in the nucleus accumbens regulates reward-related behaviors in response to chronic exposure to both rewarding and aversive stimuli by regulating largely distinct subsets of genes.
PubMedId: 27053203
Journal-article: Journal of Neuroscience, vol. 36, pp. 3954-3961, 2016
Authors: Zi-Jun Wang, Jennifer A Martin, Amy M Gancarz, Danielle N Adank, Fraser J Sim, David M Dietz
Abstract: Drug addiction is a long-lasting disease characterized by compulsive drug intake mediated in part by neuronal and biological adaptations in key brain areas, such as the nucleus accumbens (NAc). While we previously demonstrated involvement of the activin 2a receptor in drug taking, the role of its ligand, activin A, in cocaine relapse is unknown. Activin A levels in the NAc were assessed via ELISA and immunohistochemistry (in neurons, astrocytes, and microglia) following a cocaine binge paradigm. Cocaine exposure significantly increased the levels of activin A in the NAc of animals that had self-administered cocaine prior to the 14-day withdrawal compared with levels in saline controls. This was accompanied by an increase in the proportion of IBA1+ microglia in the NAc that were immunopositive for activin A. In contrast, the proportions of NeuN+ neurons and GFAP+ astrocytes that were immunopositive for activin A remained unaltered. In conclusion, these data suggest that increased secretion of activin A, particularly from microglia, in the NAc represents a novel potential target for the treatment of cocaine relapse.
PubMedId: 28272550
Journal-article: Scientific reports, vol. 7, 2016
Authors: Ramesh Chandra, Michel Engeln, Christopher Schiefer, Mary H Patton, Jennifer A Martin, Craig T Werner, Lace M Riggs, T Chase Francis, Madeleine McGlincy, Brianna Evans, Hyungwoo Nam, Shweta Das, Kasey Girven, Prasad Konkalmatt, Amy M Gancarz, Sam A Golden, Sergio D I?iguez, Scott J Russo, Gustavo Turecki, Brian N Mathur, Meaghan Creed, David M Dietz, Mary Kay Lobo
PubMedId: 29268097
Journal Article: Neuron, vol. 96, 2016
Authors: HaoSheng Sun, Diane M Damez-Werno, Kimberly N Scobie, Ning-Yi Shao, Caroline Dias, Jacqui Rabkin, Ja Wook Koo, Erica Korb, Rosemary C Bagot, Francisca H Ahn, Michael E Cahill, Benoit Labont??, Ezekiell Mouzon, Elizabeth A Heller, Hannah Cates, Sam A Golden, Kelly Gleason, Scott J Russo, Simon Andrews, Rachael Neve, Pamela J Kennedy, Ian Maze, David M Dietz, C David Allis, Gustavo Turecki, Patrick Varga-Weisz, Carol Tamminga, Li Shen, Eric J Nestler
Abstract: Improved treatment for major depressive disorder (MDD) remains elusive because of the limited understanding of its underlying biological mechanisms. It is likely that stress-induced maladaptive transcriptional regulation in limbic neural circuits contributes to the development of MDD, possibly through epigenetic factors that regulate chromatin structure. We establish that persistent upregulation of the ACF (ATP-utilizing chromatin assembly and remodeling factor) ATP-dependent chromatin-remodeling complex, occurring in the nucleus accumbens of stress-susceptible mice and depressed humans, is necessary for stress-induced depressive-like behaviors. We found that altered ACF binding after chronic stress was correlated with altered nucleosome positioning, particularly around the transcription start sites of affected genes. These alterations in ACF binding and nucleosome positioning were associated with repressed expression of genes implicated in susceptibility to stress. Together, our findings identify the ACF chromatin-remodeling complex as a critical component in the development of susceptibility to depression and in regulating stress-related behaviors.
PubMedId: 26390241
Journal-article: Nature Medicine, vol. 21, pp. 1146-1153, 2015
Authors: Ja Wook Koo, Michelle S Mazei-Robison, Quincey LaPlant, Gabor Egervari, Kevin M Braunscheidel, Danielle N Adank, Deveroux Ferguson, Jian Feng, Haosheng Sun, Kimberly N Scobie, Diane M Damez-Werno, Efrain Ribeiro, Catherine Jensen Pe??a, Deena Walker, Rosemary C Bagot, Michael E Cahill, Sarah Ann R Anderson, Benoit Labont??, Georgia E Hodes, Heidi Browne, Benjamin Chadwick, Alfred J Robison, Vincent F Vialou, Caroline Dias, Zachary Lorsch, Ezekiell Mouzon, Mary Kay Lobo, David M Dietz, Scott J Russo, Rachael L Neve, Yasmin L Hurd, Eric J Nestler
Abstract: Brain-derived neurotrophic factor (BDNF) has a crucial role in modulating neural and behavioral plasticity to drugs of abuse. We found a persistent downregulation of exon-specific Bdnf expression in the ventral tegmental area (VTA) in response to chronic opiate exposure, which was mediated by specific epigenetic modifications at the corresponding Bdnf gene promoters. Exposure to chronic morphine increased stalling of RNA polymerase II at these Bdnf promoters in VTA and altered permissive and repressive histone modifications and occupancy of their regulatory proteins at the specific promoters. Furthermore, we found that morphine suppressed binding of phospho-CREB (cAMP response element binding protein) to Bdnf promoters in VTA, which resulted from enrichment of trimethylated H3K27 at the promoters, and that decreased NURR1 (nuclear receptor related-1) expression also contributed to Bdnf repression and associated behavioral plasticity to morphine. Our findings suggest previously unknown epigenetic mechanisms of morphine-induced molecular and behavioral neuroadaptations.
PubMedId: 25643298
Journal-article: Nature Neuroscience, 2015
Authors: Lara??J. Duffney, Ping Zhong, Jing Wei, Emmanuel Matas, Jia Cheng, Luye Qin, Kaijie Ma, David??M. Dietz, Yuji Kajiwara, Joseph??D. Buxbaum, Zhen Yan
Abstract: Haploinsufficiency of the Shank3 gene, which encodes a scaffolding protein at glutamatergic synapses, is a highly prevalent and penetrant risk factor for autism. Using combined behavioral, electrophysiological, biochemical, imaging, and molecular approaches, we find that Shank3-deficient mice exhibit autism-like social deficits and repetitive behaviors, as well as the significantly diminished NMDA receptor (NMDAR) synaptic function and synaptic distribution in prefrontal cortex. Concomitantly, Shank3-deficient mice have a marked loss of cortical actin filaments, which is associated with the reduced Rac1/PAK activity and increased activity of cofilin, the major actin depolymerizing factor. The social deficits and NMDAR hypofunction are rescued by inhibiting cofilin or activating Rac1 in Shank3-deficient mice and are induced by inhibiting PAK or Rac1 in wild-type mice. These results indicate that the aberrant regulation of synaptic actin filaments and loss of synaptic NMDARs contribute to the manifestation of autism-like phenotypes. Thus, targeting actin regulators provides a strategy for autism treatment.
PubMedId: 26027926
Journal-article: Cell Reports, vol. 11, pp. 1400-1413, 2015
Authors: Katherine N Wright, Fiona Hollis, Florian Duclot, Amanda M Dossat, Caroline E Strong, T Chase Francis, Roger Mercer, Jian Feng, David M Dietz, Mary Kay Lobo, Eric J Nestler, Mohamed Kabbaj
Keywords: addiction c-Fos cocaine epigenetics methionine
Abstract: Epigenetic mechanisms, such as histone modifications, regulate responsiveness to drugs of abuse, such as cocaine, but relatively little is known about the regulation of addictive-like behaviors by DNA methylation. To investigate the influence of DNA methylation on the locomotor-activating effects of cocaine and on drug-seeking behavior, rats receiving methyl supplementation via chronic l-methionine (MET) underwent either a sensitization regimen of intermittent cocaine injections or intravenous self-administration of cocaine, followed by cue-induced and drug-primed reinstatement. MET blocked sensitization to the locomotor-activating effects of cocaine and attenuated drug-primed reinstatement, with no effect on cue-induced reinstatement or sucrose self-administration and reinstatement. Furthermore, upregulation of DNA methyltransferase 3a and 3b and global DNA hypomethylation were observed in the nucleus accumbens core (NAc), but not in the medial prefrontal cortex (mPFC), of cocaine-pretreated rats. Glutamatergic projections from the mPFC to the NAc are critically involved in the regulation of cocaine-primed reinstatement, and activation of both brain regions is seen in human addicts when reexposed to the drug. When compared with vehicle-pretreated rats, the immediate early gene c-Fos (a marker of neuronal activation) was upregulated in the NAc and mPFC of cocaine-pretreated rats after cocaine-primed reinstatement, and chronic MET treatment blocked its induction in both regions. Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. Overall, these data suggest that drug-seeking behaviors are, in part, attributable to a DNA methylation-dependent process, likely occurring at specific gene loci (e.g., c-Fos) in the reward pathway.
PubMedId: 26063926
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 35, 2015
Authors: Caroline Dias, David Dietz, Michelle Mazei-Robison, Haosheng Sun, Diane Damez-Werno, Deveroux Ferguson, Matthew Wilkinson, Jane Magida, Virginia Gao, Rachael Neve, Eric J Nestler
Keywords: Dendritic spines Medium spiny neurons Morphological plasticity Wnt signaling
Abstract: Chronic cocaine exposure increases the density of dendritic spines on medium spiny neurons (MSNs), the predominant neuronal cell type of the nucleus accumbens (NAc), a key brain reward region. We recently showed that suppression of Rac1, a small GTPase, is a critical mediator of this structural plasticity, but the upstream determinants of Rac1 activity in this context remain to be elucidated. In this study we examined whether isoforms of Dishevelled, a key hub protein of multiple branches of Wnt signaling, including Rac1, are regulated in the NAc by chronic cocaine, and whether these Dishevelled isoforms control Rac1 activity in this brain region in vivo. We found that chronic cocaine administration decreased expression of Dishevelled-2, and several other Wnt signaling components, in the NAc, and that overexpression of Dishevelled-2, but not Dishevelled-1, conversely upregulated Rac1 activity and prevented the cocaine induction of dendritic spines on NAc MSNs. We posit that the cocaine-induced downregulation of Dishevelled-2 in the NAc is an upstream regulator of Rac1 activity and plays an important role in the dynamic structural plasticity of NAc MSNs seen in response to chronic cocaine exposure.
PubMedId: 25957559
Journal-article: Neuroscience letters, vol. 598, 2015
Authors: Ramesh Chandra, T Chase Francis, Prasad Konkalmatt, Ariunzaya Amgalan, Amy M Gancarz, David M Dietz, Mary Kay Lobo
Keywords: Egr3 RiboTag cocaine medium spiny neurons nucleus accumbens transcription
Abstract: An imbalance in molecular signaling cascades and transcriptional regulation in nucleus accumbens (NAc) medium spiny neuron (MSN) subtypes, those enriched in dopamine D1 versus D2 receptors, is implicated in the behavioral responses to psychostimulants. To provide further insight into the molecular mechanisms occurring in MSN subtypes by cocaine, we examined the transcription factor early growth response 3 (Egr3). We evaluated Egr3 because it is a target of critical cocaine-mediated signaling pathways and because Egr3-binding sites are found on promoters of key cocaine-associated molecules. We first used a RiboTag approach to obtain ribosome-associated transcriptomes from each MSN subtype and found that repeated cocaine administration induced Egr3 ribosome-associated mRNA in NAc D1-MSNs while reducing Egr3 in D2-MSNs. Using Cre-inducible adeno-associated viruses combined with D1-Cre and D2-Cre mouse lines, we observed that Egr3 overexpression in D1-MSNs enhances rewarding and locomotor responses to cocaine, whereas overexpression in D2-MSNs blunts these behaviors. miRNA knock-down of Egr3 in MSN subtypes produced opposite behavioral responses from those observed with overexpression. Finally, we found that repeated cocaine administration altered Egr3 binding to promoters of genes that are important for cocaine-mediated cellular and behavioral plasticity. Genes with increased Egr3 binding to promoters, Camk2, CREB, FosB, Nr4a2, and Sirt1, displayed increased mRNA in D1-MSNs and, in some cases, a reduction in D2-MSNs. Histone and the DNA methylation enzymes G9a and Dnmt3a displayed reduced Egr3 binding to their promoters and reduced mRNA in D1-MSNs. Our study provides novel insight into an opposing role of Egr3 in select NAc MSN subtypes in cocaine action.
PubMedId: 25995477
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 35, 2015
Authors: Amy Gancarz, Yan Jouroukhin, Atsushi Saito, Alexey Shevelkin, Lauren E Mueller, Atsushi Kamiya, David M Dietz, Mikhail V Pletnikov
Keywords: Co-morbidity Cocaine DISC1 Drug abuse Psychiatric disorders
Abstract: Substance abuse and other psychiatric diseases may share molecular pathology. In order to test this hypothesis, we examined the role of Disrupted In Schizophrenia 1 (DISC1), a psychiatric risk factor, in cocaine self-administration (SA). Cocaine SA significantly increased expression of DISC1 in the nucleus accumbens (NAc); while knockdown of DISC1 in NAc significantly increased cocaine SA and decreased phosphorylation of GSK-3 at Ser9 compared to scrambled shRNA. Our study provides the first mechanistic evidence of a critical role of DISC1 in drug-induced behavioral neuroadaptations and sheds more light at the shared molecular pathology of drug abuse and other major psychiatric disorders.
PubMedId: 26385055
Journal-article: Neuroscience research, vol. 105, 2015
Authors: Amy M Gancarz, Zi-Jun Wang, Gabrielle L Schroeder, Diane Damez-Werno, Kevin M Braunscheidel, Lauren E Mueller, Monica S Humby, Aaron Caccamise, Jennifer A Martin, Karen C Dietz, Rachael L Neve, David M Dietz
Abstract: Activin receptor signaling, including the transcription factor Smad3, was upregulated in the rat nucleus accumbens (NAc) shell following withdrawal from cocaine. Direct genetic and pharmacological manipulations of this pathway bidirectionally altered cocaine seeking while governing morphological plasticity in NAc neurons. Thus, Activin/Smad3 signaling is induced following withdrawal from cocaine, and such regulation may be a key molecular mechanism underlying behavioral and cellular plasticity in the brain following cocaine self-administration.
PubMedId: 26030849
Journal-article: Nature neuroscience, vol. 18, 2015
Authors: Kimberly N Scobie, Diane Damez-Werno, HaoSheng Sun, NingYi Shao, Amy Gancarz, Clarisse H Panganiban, Caroline Dias, JaWook Koo, Paola Caiafa, Lewis Kaufman, Rachael L Neve, David M Dietz, Li Shen, Eric J Nestler
Keywords: drug addiction histone PARylation medium spiny neurons
Abstract: Many of the long-term effects of cocaine on the brain's reward circuitry have been shown to be mediated by alterations in gene expression. Several chromatin modifications, including histone acetylation and methylation, have been implicated in this regulation, but the effect of other histone modifications remains poorly understood. Poly(ADP-ribose) polymerase-1 (PARP-1), a ubiquitous and abundant nuclear protein, catalyzes the synthesis of a negatively charged polymer called poly(ADP-ribose) or PAR on histones and other substrate proteins and forms transcriptional regulatory complexes with several other chromatin proteins. Here, we identify an essential role for PARP-1 in cocaine-induced molecular, neural, and behavioral plasticity. Repeated cocaine administration, including self-administration, increased global levels of PARP-1 and its mark PAR in mouse nucleus accumbens (NAc), a key brain reward region. Using PARP-1 inhibitors and viral-mediated gene transfer, we established that PARP-1 induction in NAc mediates enhanced behavioral responses to cocaine, including increased self-administration of the drug. Using chromatin immunoprecipitation sequencing, we demonstrated a global, genome-wide enrichment of PARP-1 in NAc of cocaine-exposed mice and identified several PARP-1 target genes that could contribute to the lasting effects of cocaine. Specifically, we identified sidekick-1--important for synaptic connections during development--as a critical PARP-1 target gene involved in cocaine's behavioral effects as well as in its ability to induce dendritic spines on NAc neurons. These findings establish the involvement of PARP-1 and PARylation in the long-term actions of cocaine.
PubMedId: 24449909
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 111, 2014
Authors: Amy M Gancarz-Kausch, Danielle N Adank, David M Dietz
Abstract: Drug addiction is characterized by compulsive drug-taking behaviors and a high propensity to relapse following drug cessation. Drug craving and seeking can increase during a period of abstinence, but this phenomenon is not observed in drug-induced reinstatement models. To investigate the effect of withdrawal on cocaine relapse, rats were exposed to extended-access cocaine self-administration and subjected to either 1 or 30 d of withdrawal. When tested during 12 h unlimited access to cocaine (binge), the duration of the withdrawal did not influence cocaine intake. However, using a histamine punishment procedure that greatly suppresses drug-taking behavior, we demonstrate that longer periods of abstinence from cocaine induce a greater persistence in responding for drug in the face of negative consequences.
PubMedId: 25363133
Journal-article: Scientific reports, vol. 4, 2014
Authors: David A Thorn, Li Jing, Yanyan Qiu, Amy M Gancarz-Kausch, Chad M Galuska, David M Dietz, Yanan Zhang, Jun-Xu Li
Abstract: Animal knockout studies suggest that trace amine-associated receptor (TAAR) 1 is involved in behavioral effects of psychostimulants such as cocaine. Recently, several highly selective TAAR 1 agonists have been discovered. However, little is known of the impact of TAAR 1 agonists on abuse-related effects of cocaine. Here, we report the effects of a TAAR 1 agonist RO5263397 on several abuse-related behavioral effects of cocaine in rats. RO5263397 was evaluated for its effects on cocaine-induced behavioral sensitization, conditioned place preference (CPP), cue- and cocaine prime-induced reinstatement of cocaine-seeking behavior, and cocaine self-administration using behavioral economic analysis. RO5263397 reduced the expression of cocaine behavioral sensitization, cue- and cocaine prime-induced reinstatement of cocaine seeking, and expression but not development of cocaine CPP. Behavioral economic analysis showed that RO5263397 increased the elasticity of the cocaine demand curve, but did not change cocaine consumption at minimal prices. Taken together, this is the first systematic assessment of a TAAR 1 agonist on a range of behavioral effects of cocaine, showing that RO5263397 was efficacious in reducing cocaine-mediated behaviors. Collectively, these data uncover essential neuromodulatory roles of TAAR 1 on cocaine abuse, and suggest that TAAR 1 may represent a novel drug target for the treatment of cocaine addiction.
PubMedId: 24743376
Journal-article: Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, vol. 39, 2014
Authors: V. Vialou, R. C. Bagot, M. E. Cahill, D. Ferguson, A. J. Robison, D. M. Dietz, B. Fallon, M. Mazei-Robison, S. M. Ku, E. Harrigan, C. A. Winstanley, T. Joshi, J. Feng, O. Berton, E. J. Nestler
Keywords: CCK accumbens amygdala anxiety depression mPFC
Abstract: Decreased medial prefrontal cortex (mPFC) neuronal activity is associated with social defeat-induced depression- and anxiety-like behaviors in mice. However, the molecular mechanisms underlying the decreased mPFC activity and its prodepressant role remain unknown. We show here that induction of the transcription factor FosB in mPFC, specifically in the prelimbic (PrL) area, mediates susceptibility to stress. FosB induction in PrL occurred selectively in susceptible mice after chronic social defeat stress, and overexpression of FosB in this region, but not in the nearby infralimbic (IL) area, enhanced stress susceptibility. FosB produced these effects partly through induction of the cholecystokinin (CCK)-B receptor: CCKB blockade in mPFC induces a resilient phenotype, whereas CCK administration into mPFC mimics the anxiogenic- and depressant-like effects of social stress. We previously found that optogenetic stimulation of mPFC neurons in susceptible mice reverses several behavioral abnormalities seen after chronic social defeat stress. Therefore, we hypothesized that optogenetic stimulation of cortical projections would rescue the pathological effects of CCK in mPFC. After CCK infusion in mPFC, we optogenetically stimulated mPFC projections to basolateral amygdala or nucleus accumbens, two subcortical structures involved in mood regulation. Stimulation of corticoamygdala projections blocked the anxiogenic effect of CCK, although no effect was observed on other symptoms of social defeat. Conversely, stimulation of corticoaccumbens projections reversed CCK-induced social avoidance and sucrose preference deficits but not anxiogenic-like effects. Together, these results indicate that social stress-induced behavioral deficits are mediated partly by molecular adaptations in mPFC involving FosB and CCK through cortical projections to distinct subcortical targets.
PubMedId: 24623766
Journal-article: Journal of Neuroscience, vol. 34, pp. 3878-3887, 2014
Authors: Ian Maze, Dipesh Chaudhury, David M Dietz, Melanie Von Schimmelmann, Pamela J Kennedy, Mary Kay Lobo, Stephanie E Sillivan, Michael L Miller, Rosemary C Bagot, HaoSheng Sun, Gustavo Turecki, Rachael L Neve, Yasmin L Hurd, Li Shen, Ming-Hu Han, Anne Schaefer, Eric J Nestler
Abstract: Cocaine-mediated repression of the histone methyltransferase (HMT) G9a has recently been implicated in transcriptional, morphological and behavioral responses to chronic cocaine administration. Here, using a ribosomal affinity purification approach, we found that G9a repression by cocaine occurred in both Drd1-expressing (striatonigral) and Drd2-expressing (striatopallidal) medium spiny neurons. Conditional knockout and overexpression of G9a within these distinct cell types, however, revealed divergent behavioral phenotypes in response to repeated cocaine treatment. Our studies further indicated that such developmental deletion of G9a selectively in Drd2 neurons resulted in the unsilencing of transcriptional programs normally specific to striatonigral neurons and in the acquisition of Drd1-associated projection and electrophysiological properties. This partial striatopallidal to striatonigral 'switching' phenotype in mice indicates a new role for G9a in contributing to neuronal subtype identity and suggests a critical function for cell type-specific histone methylation patterns in the regulation of behavioral responses to environmental stimuli.
PubMedId: 24584053
Journal-article: Nature Neuroscience, vol. 17, pp. 533-539, 2014
Authors: Allyson K Friedman, Jessica J Walsh, Barbara Juarez, Stacy M Ku, Dipesh Chaudhury, Jing Wang, Xianting Li, David M Dietz, Nina Pan, Vincent F Vialou, Rachael L Neve, Zhenyu Yue, Ming-Hu Han
Abstract: Typical therapies try to reverse pathogenic mechanisms. Here, we describe treatment effects achieved by enhancing depression-causing mechanisms in ventral tegmental area (VTA) dopamine (DA) neurons. In a social defeat stress model of depression, depressed (susceptible) mice display hyperactivity of VTA DA neurons, caused by an up-regulated hyperpolarization-activated current (I(h)). Mice resilient to social defeat stress, however, exhibit stable normal firing of these neurons. Unexpectedly, resilient mice had an even larger I(h), which was observed in parallel with increased potassium (K(+)) channel currents. Experimentally further enhancing Ih or optogenetically increasing the hyperactivity of VTA DA neurons in susceptible mice completely reversed depression-related behaviors, an antidepressant effect achieved through resilience-like, projection-specific homeostatic plasticity. These results indicate a potential therapeutic path of promoting natural resilience for depression treatment.
PubMedId: 24744379
Journal-article: Science (New York, N.Y.), vol. 344, 2014
Authors: L E Mueller, M A Kausch, T Markovic, D A A MacLaren, D M Dietz, J Park, S D Clark
Keywords: Cocaine Conditioned place preference Laterodorsal tegmentum Pedunculopontine tegmentum Urotensin II
Abstract: Although the peptide urotensin II (UII) has well studied direct actions on the cardiovascular system, the UII receptor (UIIR) is expressed by neurons of the hindbrain. Specifically, the UIIR is expressed by the cholinergic neurons of the laterodorsal tegmentum (LDTg) and the pedunculopontine tegmentum (PPTg). These neurons send axons to the ventral tegmental area (VTA), for which the PPTg and LDTg are the sole source of acetylcholine. Therefore, it was hypothesized that UIIR activation within the VTA would modulate reward-related behaviors, such as cocaine-induced drug seeking. Intra-VTA microinjections of UII at high concentrations (1 nmole) established conditioned place preference (CPP), but also blocked cocaine-mediated CPP (10 mg/kg). When rats received systemic sub-effectual doses of cocaine (7.5 mg/kg) with intra-VTA injections of 1 or 10 pmole of UII CPP was formed. Furthermore, the second endogenous ligand for the UIIR, urotensin II-related peptide, had the same effect at the 10 pmole dose. The effects of low doses of UII were blocked by pretreatment with the UIIR antagonist SB657510. Furthermore, it was found that intra-VTA UII (10 pmole) further increased cocaine-mediated (7.5 mg/kg) rises in electrically evoked dopamine in the nucleus accumbens. Our study has found that activation of VTA-resident UIIR produces observable behavioral changes in rats, and that UIIR is able to modulate the effects of cocaine. In addition, it was found that UIIR activation within the VTA can potentiate cocaine-mediated neurochemical effects. Therefore, the coincident activation of the UII-system and cocaine administration may increase the liability for drug taking behavior.
PubMedId: 25264578
Journal-article: Behavioural brain research, vol. 278, 2014
Authors: Amy M Gancarz-Kausch, Gabrielle L Schroeder, Clarisse Panganiban, Danielle Adank, Monica S Humby, Michael A Kausch, Stewart D Clark, David M Dietz
Abstract: The addicted phenotype is characterized as a long-lasting, chronically relapsing disorder that persists following long periods of abstinence, suggesting that the underlying molecular changes are stable and endure for long periods even in the absence of drug. Here, we investigated Transforming Growth Factor-Beta Type I receptor (TGF- R1) expression in the nucleus accumbens (NAc) following periods of withdrawal from cocaine self-administration (SA) and a sensitizing regimen of non-contingent cocaine. Rats were exposed to either (i) repeated systemic injections (cocaine or saline), or (ii) self-administration (cocaine or saline) and underwent a period of forced abstinence (either 1 or 7 days of drug cessation). Withdrawal from cocaine self-administration resulted in an increase in TGF- R1 protein expression in the NAc compared to saline controls. This increase was specific for volitional cocaine intake as no change in expression was observed following a sensitizing regimen of experimenter-administered cocaine. These findings implicate TGF- signaling as a novel potential therapeutic target for treating drug addiction.
PubMedId: 24386286
Journal-article: PloS one, vol. 8, 2013
Authors: David M Dietz, Pamela J Kennedy, HaoSheng Sun, Ian Maze, Amy M Gancarz, Vincent Vialou, Ja Wook Koo, Ezekiell Mouzon, Subroto Ghose, Carol A Tamminga, Eric J Nestler
Abstract: FosB, a FosB gene product, is induced in the prefrontal cortex (PFC) by repeated exposure to several stimuli including antipsychotic drugs such as haloperidol. However, the functional consequences of increased FosB expression following antipsychotic treatment have not been explored. Here, we assessed whether FosB induction by haloperidol mediates the positive or negative consequences or clinical-related actions of antipsychotic treatment. We show that individuals with schizophrenia who were medicated with antipsychotic drugs at their time of death display increased FosB levels in the PFC, an effect that is replicated in rats treated chronically with haloperidol. In contrast, individuals with schizophrenia who were medication-free did not exhibit this effect. Viral-mediated overexpression of FosB in the PFC of rodents induced cognitive deficits as measured by inhibitory avoidance, increased startle responses in prepulse inhibition tasks, and increased MK-801-induced anxiety-like behaviors. Together, these results suggest that FosB induction in the PFC by antipsychotic treatment contributes to the deleterious effects of these drugs and not to their therapeutic actions.
PubMedId: 24067299
Journal-article: Neuropsychopharmacology, vol. 39, pp. 538-544, 2013
Authors: Ramesh Chandra, Jeffrey D Lenz, Amy M Gancarz, Dipesh Chaudhury, Gabrielle L Schroeder, Ming-Hu Han, Joseph F Cheer, David M Dietz, Mary Kay Lobo
Keywords: Tiam1 cocaine medium spiny neurons nucleus accumbens optogenetics
Abstract: Exposure to psychostimulants results in structural and synaptic plasticity in striatal medium spiny neurons (MSNs). These cellular adaptations arise from alterations in genes that are highly implicated in the rearrangement of the actin-cytoskeleton, such as T-lymphoma invasion and metastasis 1 (Tiam1). Previous studies have demonstrated a crucial role for dopamine receptor 1 (D1)-containing striatal MSNs in mediating psychostimulant induced plasticity changes. These D1-MSNs in the nucleus accumbens (NAc) positively regulate drug seeking, reward, and locomotor behavioral effects as well as the morphological adaptations of psychostimulant drugs. Here, we demonstrate that rats that actively self-administer cocaine display reduced levels of Tiam1 in the NAc. To further examine the cell type-specific contribution to these changes in Tiam1 we used optogenetics to selectively manipulate NAc D1-MSNs or dopamine receptor 2 (D2) expressing MSNs. We find that repeated channelrhodopsin-2 activation of D1-MSNs but not D2-MSNs caused a down-regulation of Tiam1 levels similar to the effects of cocaine. Further, activation of D2-MSNs, which caused a late blunted cocaine-mediated locomotor behavioral response, did not alter Tiam1 levels. We then examined the contribution of D1-MSNs to the cocaine-mediated decrease of Tiam1. Using the light activated chloride pump, eNpHR3.0 (enhanced Natronomonas pharaonis halorhodopsin 3.0), we selectively inhibited D1-MSNs during cocaine exposure, which resulted in a behavioral blockade of cocaine-induced locomotor sensitization. Moreover, inhibiting these NAc D1-MSNs during cocaine exposure reversed the down-regulation of Tiam1 gene expression and protein levels. These data demonstrate that altering activity in specific neural circuits with optogenetics can impact the underlying molecular substrates of psychostimulant-mediated behavior and function.
PubMedId: 23745104
Journal-article: Frontiers in molecular neuroscience, vol. 6, 2013
Authors: David M Dietz, Haosheng Sun, Mary Kay Lobo, Michael E Cahill, Benjamin Chadwick, Virginia Gao, Ja Wook Koo, Michelle S Mazei-Robison, Caroline Dias, Ian Maze, Diane Damez-Werno, Karen C Dietz, Kimberly N Scobie, Deveroux Ferguson, Daniel Christoffel, Yoko Ohnishi, Georgia E Hodes, Yi Zheng, Rachael L Neve, Klaus M Hahn, Scott J Russo, Eric J Nestler
Abstract: Repeated cocaine administration increases the dendritic arborization of nucleus accumbens neurons, but the underlying signaling events remain unknown. Here we show that repeated exposure to cocaine negatively regulates the active form of Rac1, a small GTPase that controls actin remodeling in other systems. Further, we show, using viral-mediated gene transfer, that overexpression of a dominant negative mutant of Rac1 or local knockout of Rac1 is sufficient to increase the density of immature dendritic spines on nucleus accumbens neurons, whereas overexpression of a constitutively active Rac1 or light activation of a photoactivatable form of Rac1 blocks the ability of repeated cocaine exposure to produce this effect. Downregulation of Rac1 activity likewise promotes behavioral responses to cocaine exposure, with activation of Rac1 producing the opposite effect. These findings establish that Rac1 signaling mediates structural and behavioral plasticity in response to cocaine exposure.
PubMedId: 22522400
Journal-article: Nature neuroscience, vol. 15, 2012
Authors: Diane Damez-Werno, Quincey LaPlant, HaoSheng Sun, Kimberly N Scobie, David M Dietz, Ian M Walker, Ja Wook Koo, Vincent F Vialou, Ezekiell Mouzon, Scott J Russo, Eric J Nestler
Abstract: FosB, a Fosb gene product, is induced in nucleus accumbens (NAc) and caudate-putamen (CPu) by repeated exposure to drugs of abuse such as cocaine. This induction contributes to aberrant patterns of gene expression and behavioral abnormalities seen with repeated drug exposure. Here, we assessed whether a remote history of cocaine exposure in rats might alter inducibility of the Fosb gene elicited by subsequent drug exposure. We show that prior chronic cocaine administration, followed by extended withdrawal, increases inducibility of Fosb in NAc, as evidenced by greater acute induction of FosB mRNA and faster accumulation of FosB protein after repeated cocaine reexposure. No such primed Fosb induction was observed in CPu; in fact, subsequent acute induction of FosB mRNA was suppressed in CPu. These abnormal patterns of Fosb expression are associated with chromatin modifications at the Fosb gene promoter. Prior chronic cocaine administration induces a long-lasting increase in RNA polymerase II (Pol II) binding at the Fosb promoter in NAc only, suggesting that Pol II "stalling" primes Fosb for induction in this region upon reexposure to cocaine. A cocaine challenge then triggers the release of Pol II from the gene promoter, allowing for more rapid Fosb transcription. A cocaine challenge also decreases repressive histone modifications at the Fosb promoter in NAc, but increases such repressive marks and decreases activating marks in CPu. These results provide new insight into the chromatin dynamics at the Fosb promoter and reveal a novel mechanism for primed Fosb induction in NAc upon reexposure to cocaine.
PubMedId: 22836260
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 32, 2012
Authors: Mitsumasa Kurita, Terrell Holloway, Aintzane Garc??a-Bea, Alexey Kozlenkov, Allyson K Friedman, Jos?? L Moreno, Mitra Heshmati, Sam A Golden, Pamela J Kennedy, Nagahide Takahashi, David M Dietz, Giuseppe Mocci, Ane M Gabilondo, James Hanks, Adrienne Umali, Luis F Callado, Amelia L Gallitano, Rachael L Neve, Li Shen, Joseph D Buxbaum, Ming-Hu Han, Eric J Nestler, J Javier Meana, Scott J Russo, Javier Gonz??lez-Maeso
Abstract: Histone deacetylases (HDACs) compact chromatin structure and repress gene transcription. In schizophrenia, clinical studies demonstrate that HDAC inhibitors are efficacious when given in combination with atypical antipsychotics. However, the molecular mechanism that integrates a better response to antipsychotics with changes in chromatin structure remains unknown. Here we found that chronic atypical antipsychotics downregulated the transcription of metabotropic glutamate 2 receptor (mGlu2, also known as Grm2), an effect that was associated with decreased histone acetylation at its promoter in mouse and human frontal cortex. This epigenetic change occurred in concert with a serotonin 5-HT(2A) receptor-dependent upregulation and increased binding of HDAC2 to the mGlu2 promoter. Virally mediated overexpression of HDAC2 in frontal cortex decreased mGlu2 transcription and its electrophysiological properties, thereby increasing psychosis-like behavior. Conversely, HDAC inhibitors prevented the repressive histone modifications induced at the mGlu2 promoter by atypical antipsychotics, and augmented their therapeutic-like effects. These observations support the view of HDAC2 as a promising new target for schizophrenia treatment.
PubMedId: 22864611
Journal-article: Nature Neuroscience, vol. 15, pp. 1245-1254, 2012
Authors: Ja Wook Koo, Michelle S Mazei-Robison, Dipesh Chaudhury, Barbara Juarez, Quincey LaPlant, Deveroux Ferguson, Jian Feng, Haosheng Sun, Kimberly N Scobie, Diane Damez-Werno, Marshall Crumiller, Yoshinori N Ohnishi, Yoko H Ohnishi, Ezekiell Mouzon, David M Dietz, Mary Kay Lobo, Rachael L Neve, Scott J Russo, Ming-Hu Han, Eric J Nestler
Abstract: Brain-derived neurotrophic factor (BDNF) is a key positive regulator of neural plasticity, promoting, for example, the actions of stimulant drugs of abuse such as cocaine. We discovered a surprising opposite role for BDNF in countering responses to chronic morphine exposure. The suppression of BDNF in the ventral tegmental area (VTA) enhanced the ability of morphine to increase dopamine (DA) neuron excitability and promote reward. In contrast, optical stimulation of VTA DA terminals in nucleus accumbens (NAc) completely reversed the suppressive effect of BDNF on morphine reward. Furthermore, we identified numerous genes in the NAc, a major target region of VTA DA neurons, whose regulation by BDNF in the context of chronic morphine exposure mediated this counteractive function. These findings provide insight into the molecular basis of morphine-induced neuroadaptations in the brain's reward circuitry.
PubMedId: 23042896
Journal-article: Science (New York, N.Y.), vol. 338, 2012
Authors: Jia Liu, Karen Dietz, Jacqueline M DeLoyht, Xiomara Pedre, Dipti Kelkar, Jasbir Kaur, Vincent Vialou, Mary Kay Lobo, David M Dietz, Eric J Nestler, Jeffrey Dupree, Patrizia Casaccia
Abstract: Protracted social isolation of adult mice induced behavioral, transcriptional and ultrastructural changes in oligodendrocytes of the prefrontal cortex (PFC) and impaired adult myelination. Social re-integration was sufficient to normalize behavioral and transcriptional changes. Short periods of isolation affected chromatin and myelin, but did not induce behavioral changes. Thus, myelinating oligodendrocytes in the adult PFC respond to social interaction with chromatin changes, suggesting that myelination acts as a form of adult plasticity.
PubMedId: 23143512
Journal-article: Nature neuroscience, vol. 15, 2012
Authors: Haosheng Sun, Ian Maze, David M Dietz, Kimberly N Scobie, Pamela J Kennedy, Diane Damez-Werno, Rachael L Neve, Venetia Zachariou, Li Shen, Eric J Nestler
Abstract: Dysregulation of histone modifying enzymes has been associated with numerous psychiatric disorders. Alterations in G9a (Ehmt2), a histone methyltransferase that catalyzes the euchromatic dimethylation of histone H3 at lysine 9 (H3K9me2), has been implicated recently in mediating neural and behavioral plasticity in response to chronic cocaine administration. Here, we show that chronic morphine, like cocaine, decreases G9a expression, and global levels of H3K9me2, in mouse nucleus accumbens (NAc), a key brain reward region. In contrast, levels of other histone methyltransferases or demethylases, or of other methylated histone marks, were not affected in NAc by chronic morphine. Through viral-mediated gene transfer and conditional mutagenesis, we found that overexpression of G9a in NAc opposes morphine reward and locomotor sensitization and concomitantly promotes analgesic tolerance and naloxone-precipitated withdrawal, whereas downregulation of G9a in NAc enhances locomotor sensitization and delays the development of analgesic tolerance. We identified downstream targets of G9a by providing a comprehensive chromatin immunoprecipitation followed by massively parallel sequencing analysis of H3K9me2 distribution in NAc in the absence and presence of chronic morphine. These data provide novel insight into the epigenomic regulation of H3K9me2 by chronic morphine and suggest novel chromatin-based mechanisms through which morphine-induced addictive-like behaviors arise.
PubMedId: 23197736
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 32, 2012
Authors: M. B. Wilkinson, C. Dias, J. Magida, M. Mazei-Robison, M. Lobo, P. Kennedy, D. Dietz, H. Covington, S. Russo, R. Neve, S. Ghose, C. Tamminga, E. J. Nestler
Abstract: Based on earlier gene expression and chromatin array data, we identified the protein, dishevelled (DVL)-2, as being regulated in the nucleus accumbens (NAc), a key brain reward region, in the mouse social defeat model of depression. Here, we validate these findings by showing that DVL2 mRNA and protein levels are downregulated in NAc of mice susceptible to social defeat stress, effects not seen in resilient mice. Other DVL isoforms, DVL1 and DVL3, show similar patterns of regulation. Downregulation of DVL was also demonstrated in the NAc of depressed humans examined postmortem. Interestingly, several members of the WNT (Wingless)-DVL signaling cascade, including phospho-GSK3 (glycogen synthase kinase-3), also show significant downregulation in the NAc of susceptible, but not resilient, mice, demonstrating concerted regulation of this pathway in the NAc due to social defeat stress. By using viral-mediated gene transfer to overexpress a dominant-negative mutant of DVL in NAc, or by using a pharmacological inhibitor of DVL administered into this brain region, we show that blockade of DVL function renders mice more susceptible to social defeat stress and promotes depression-like behavior in other assays. Similar prodepression-like effects were induced upon overexpressing GSK3 in the NAc, while overexpressing a dominant-negative mutant of GSK3 promoted resilience to social defeat stress. These findings are consistent with the knowledge that downregulation of DVL and phospho-GSK3 reflects an increase in GSK3 activity. These studies reveal a novel role for the DVL-GSK3 signaling pathway, acting within the brain's reward circuitry, in regulating susceptibility to chronic stress.
PubMedId: 21697359
Journal-article: Journal of Neuroscience, vol. 31, pp. 9084-9092, 2011
Authors: D. J. Christoffel, S. A. Golden, D. Dumitriu, A. J. Robison, W. G. Janssen, H. F. Ahn, V. Krishnan, C. M. Reyes, M.-H. Han, J. L. Ables, A. J. Eisch, D. M. Dietz, D. Ferguson, R. L. Neve, P. Greengard, Y. Kim, J. H. Morrison, S. J. Russo
Abstract: The neurobiological underpinnings of mood and anxiety disorders have been linked to the nucleus accumbens (NAc), a region important in processing the rewarding and emotional salience of stimuli. Using chronic social defeat stress, an animal model of mood and anxiety disorders, we investigated whether alterations in synaptic plasticity are responsible for the long-lasting behavioral symptoms induced by this form of stress. We hypothesized that chronic social defeat stress alters synaptic strength or connectivity of medium spiny neurons (MSNs) in the NAc to induce social avoidance. To test this, we analyzed the synaptic profile of MSNs via confocal imaging of Lucifer-yellow-filled cells, ultrastructural analysis of the postsynaptic density, and electrophysiological recordings of miniature EPSCs (mEPSCs) in mice after social defeat. We found that NAc MSNs have more stubby spine structures with smaller postsynaptic densities and an increase in the frequency of mEPSCs after social defeat. In parallel to these structural changes, we observed significant increases in IB kinase (IKK) in the NAc after social defeat, a molecular pathway that has been shown to regulate neuronal morphology. Indeed, we find using viral-mediated gene transfer of dominant-negative and constitutively active IKK mutants that activation of IKK signaling pathways during social defeat is both necessary and sufficient to induce synaptic alterations and behavioral effects of the stress. These studies establish a causal role for IKK in regulating stress-induced adaptive plasticity and may present a novel target for drug development in the treatment of mood and anxiety disorders in humans.
PubMedId: 21209217
Journal-article: Journal of Neuroscience, vol. 31, pp. 314-321, 2011
Authors: T. E. Brown, B. R. Lee, P. Mu, D. Ferguson, D. Dietz, Y. N. Ohnishi, Y. Lin, A. Suska, M. Ishikawa, Y. H. Huang, H. Shen, P. W. Kalivas, B. A. Sorg, R. S. Zukin, E. J. Nestler, Y. Dong, O. M. Schluter
Abstract: Locomotor sensitization is a common and robust behavioral alteration in rodents whereby following exposure to abused drugs such as cocaine, the animal becomes significantly more hyperactive in response to an acute drug challenge. Here, we further analyzed the role of cocaine-induced silent synapses in the nucleus accumbens (NAc) shell and their contribution to the development of locomotor sensitization. Using a combination of viral vector-mediated genetic manipulations, biochemistry, and electrophysiology in a locomotor sensitization paradigm with repeated, daily, noncontingent cocaine (15 mg/kg) injections, we show that dominant-negative cAMP-element binding protein (CREB) prevents cocaine-induced generation of silent synapses of young (30 d old) rats, whereas constitutively active CREB is sufficient to increase the number of NR2B-containing NMDA receptors (NMDARs) at synapses and to generate silent synapses. We further show that occupancy of CREB at the NR2B promoter increases and is causally related to the increase in synaptic NR2B levels. Blockade of NR2B-containing NMDARs by administration of the NR2B-selective antagonist Ro256981 directly into the NAc, under conditions that inhibit cocaine-induced silent synapses, prevents the development of cocaine-elicited locomotor sensitization. Our data are consistent with a cellular cascade whereby cocaine-induced activation of CREB promotes CREB-dependent transcription of NR2B and synaptic incorporation of NR2B-containing NMDARs, which generates new silent synapses within the NAc. We propose that cocaine-induced activation of CREB and generation of new silent synapses may serve as key cellular events mediating cocaine-induced locomotor sensitization. These findings provide a novel cellular mechanism that may contribute to cocaine-induced behavioral alterations.
PubMedId: 21632938
Journal-article: Journal of Neuroscience, vol. 31, pp. 8163-8174, 2011
Authors: Herbert E Covington, Ian Maze, HaoSheng Sun, Howard M Bomze, Kristine D DeMaio, Emma Y Wu, David M Dietz, Mary Kay Lobo, Subroto Ghose, Ezekiel Mouzon, Rachael L Neve, Carol A Tamminga, Eric J Nestler
Abstract: Substance abuse increases an individual's vulnerability to stress-related illnesses, which is presumably mediated by drug-induced neural adaptations that alter subsequent responses to stress. Here, we identify repressive histone methylation in nucleus accumbens (NAc), an important brain reward region, as a key mechanism linking cocaine exposure to increased stress vulnerability. Repeated cocaine administration prior to subchronic social defeat stress potentiated depressive-like behaviors in mice through decreased levels of histone H3 lysine 9 dimethylation in NAc. Cre-mediated reduction of the histone methyltransferase, G9a, in NAc promoted increased susceptibility to social stress, similar to that observed with repeated cocaine. Conversely, G9a overexpression in NAc after repeated cocaine protected mice from the consequences of subsequent stress. This resilience was mediated, in part, through repression of BDNF-TrkB-CREB signaling, which was induced after repeated cocaine or stress. Identifying such common regulatory mechanisms may aid in the development of new therapies for addiction and depression.
PubMedId: 21867882
Journal-article: Neuron, vol. 71, 2011
Authors: Allison T Knoll, John W Muschamp, Stephanie E Sillivan, Deveroux Ferguson, David M Dietz, Edward G Meloni, F Ivy Carroll, Eric J Nestler, Christine Konradi, William A Carlezon
Abstract: The kappa opioid receptor (KOR) system contributes to the prodepressive and aversive consequences of stress and is implicated in the facilitation of conditioned fear and anxiety in rodents. Here, we sought to identify neural circuits that mediate KOR system effects on fear and anxiety in rats. We assessed whether fear conditioning induces plasticity in KOR or dynorphin (the endogenous KOR ligand) messenger RNA (mRNA) expression in the basolateral (BLA) and central (CeA) nuclei of the amygdala, hippocampus, or striatum. We then assessed whether microinfusions of the KOR antagonist JDTic (0-10 g/side) into the BLA or CeA affect the expression of conditioned fear or anxiety. Finally, we examined whether fear extinction induces plasticity in KOR mRNA expression that relates to the quality of fear extinction. Fear conditioning upregulated KOR mRNA in the BLA by 65% and downregulated it in the striatum by 22%, without affecting KOR levels in the CeA or hippocampus, or dynorphin levels in any region. KOR antagonism in either the BLA or CeA decreased conditioned fear in the fear-potentiated startle paradigm, whereas KOR antagonism in the BLA, but not the CeA, produced anxiolytic-like effects in the elevated plus maze. Effective fear extinction was associated with a 67% reduction in KOR mRNA in the BLA. These findings suggest that fear conditioning and extinction dynamically regulate KOR expression in the BLA and provide evidence that the BLA and CeA are important neural substrates mediating the anxiolytic-like effects of KOR antagonists in models of fear and anxiety.
PubMedId: 21531393
Journal-article: Biological psychiatry, vol. 70, 2011
Authors: David M Dietz, Quincey Laplant, Emily L Watts, Georgia E Hodes, Scott J Russo, Jian Feng, Ronald S Oosting, Vincent Vialou, Eric J Nestler
Abstract: There has been recent interest in the possibility that epigenetic mechanisms might contribute to the transgenerational transmission of stress-induced vulnerability. Here, we focused on possible paternal transmission with the social defeat stress paradigm. Adult male mice exposed to chronic social defeat stress or control nondefeated mice were bred with normal female mice, and their offspring were assessed behaviorally for depressive- and anxiety-like measures. Plasma levels of corticosterone and vascular endothelial growth factor were also assayed. To directly assess the role of epigenetic mechanisms, we used in vitro fertilization (IVF); behavioral assessments were conducted on offspring of mice from IVF-control and IVF-defeated fathers. We show that both male and female offspring from defeated fathers exhibit increased measures of several depression- and anxiety-like behaviors. The male offspring of defeated fathers also display increased baseline plasma levels of corticosterone and decreased levels of vascular endothelial growth factor. However, most of these behavioral changes were not observed when offspring were generated through IVF. These results suggest that, although behavioral adaptations that occur after chronic social defeat stress can be transmitted from the father to his male and female F1 progeny, only very subtle changes might be transmitted epigenetically under the conditions tested.
PubMedId: 21679926
Journal-article: Biological psychiatry, vol. 70, 2011
Authors: David M Dietz, Eric J Nestler
PubMedId: 22157877
Journal-article: Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, vol. 37, 2011
Authors: F Hollis, M Gaval-Cruz, N Carrier, D M Dietz, M Kabbaj
Abstract: Adolescents are more likely to experiment with and become addicted to drugs of abuse. A number of studies indicate that the developmental forebrain may be responsible for making adolescents vulnerable to the addictive properties of such drugs. The aim of this study was to first compare behavioral responses to novelty and cocaine between juvenile and adult rats and then compare levels of the immediate-early gene zif268 activation in several forebrain areas via in situ hybridization. We found that juveniles demonstrated higher locomotion scores and required a higher dose of cocaine than adults to establish a conditioned place preference. Additionally, at this higher dose, juvenile rats exhibited higher levels of zif268 mRNA in the prefrontal cortex compared with adults. A developmental effect for increased zif268 mRNA was also observed in the striatum and nucleus accumbens, but there was no interaction with the cocaine dose. These findings hold interesting implications for the study of the molecular mechanisms underlying juvenile drug addiction.
PubMedId: 22056598
Journal-article: Neuroscience, vol. 200, 2011
Authors: Ian Maze, Herbert E Covington, David M Dietz, Quincey LaPlant, William Renthal, Scott J Russo, Max Mechanic, Ezekiell Mouzon, Rachael L Neve, Stephen J Haggarty, Yanhua Ren, Srihari C Sampath, Yasmin L Hurd, Paul Greengard, Alexander Tarakhovsky, Anne Schaefer, Eric J Nestler
Abstract: Cocaine-induced alterations in gene expression cause changes in neuronal morphology and behavior that may underlie cocaine addiction. In mice, we identified an essential role for histone 3 lysine 9 (H3K9) dimethylation and the lysine dimethyltransferase G9a in cocaine-induced structural and behavioral plasticity. Repeated cocaine administration reduced global levels of H3K9 dimethylation in the nucleus accumbens. This reduction in histone methylation was mediated through the repression of G9a in this brain region, which was regulated by the cocaine-induced transcription factor DeltaFosB. Using conditional mutagenesis and viral-mediated gene transfer, we found that G9a down-regulation increased the dendritic spine plasticity of nucleus accumbens neurons and enhanced the preference for cocaine, thereby establishing a crucial role for histone methylation in the long-term actions of cocaine.
PubMedId: 20056891
Journal-article: Science (New York, N.Y.), vol. 327, 2010
Authors: Vincent Vialou, Alfred J Robison, Quincey C LaPlant, Herbert E Covington, David M Dietz, Yoshinori N Ohnishi, Ezekiell Mouzon, Augustus J Rush, Emily L Watts, Deanna L Wallace, Sergio D I??iguez, Yoko H Ohnishi, Michel A Steiner, Brandon L Warren, Vaishnav Krishnan, Carlos A Bola??os, Rachael L Neve, Subroto Ghose, Olivier Berton, Carol A Tamminga, Eric J Nestler
Abstract: In contrast with the many studies of stress effects on the brain, relatively little is known about the molecular mechanisms of resilience, the ability of some individuals to escape the deleterious effects of stress. We found that the transcription factor DeltaFosB mediates an essential mechanism of resilience in mice. Induction of DeltaFosB in the nucleus accumbens, an important brain reward-associated region, in response to chronic social defeat stress was both necessary and sufficient for resilience. DeltaFosB induction was also required for the standard antidepressant fluoxetine to reverse behavioral pathology induced by social defeat. DeltaFosB produced these effects through induction of the GluR2 AMPA glutamate receptor subunit, which decreased the responsiveness of nucleus accumbens neurons to glutamate, and through other synaptic proteins. Together, these findings establish a previously unknown molecular pathway underlying both resilience and antidepressant action.
PubMedId: 20473292
Journal-article: Nature Neuroscience, vol. 13, pp. 745-752, 2010
Authors: Kimberly A Young, Yan Liu, Kyle L Gobrogge, David M Dietz, Hui Wang, Mohamed Kabbaj, Zuoxin Wang
Abstract: We have recently established the socially monogamous prairie vole (Microtus ochrogaster) as an animal model with which to investigate the involvement of mesocorticolimbic dopamine (DA) in the amphetamine (AMPH)-induced impairment of social behavior. As the majority of our work, to date, has focused on males, and sex differences are commonly reported in the behavioral and neurobiological responses to AMPH, the current study was designed to examine the behavioral and neurobiological effects of AMPH treatment in female prairie voles. We used a conditioned place preference (CPP) paradigm to determine a dose-response curve for the behavioral effects of AMPH in female prairie voles, and found that conditioning with low to intermediate (0.2 and 1.0 mg/kg), but not very low (0.1 mg/kg), doses of AMPH induced a CPP. We also found that exposure to a behaviorally relevant dose of AMPH (1.0 mg/kg) induced an increase in DA concentration in the nucleus accumbens (NAcc) and caudate putamen but not the medial prefrontal cortex or ventral tegmental area (VTA). Finally, repeated AMPH exposure (1.0 mg/kg once per day for 3 consecutive days; an injection paradigm that has been recently shown to alter DA receptor expression and impair social bonding in male prairie voles) increased D1, but not D2, receptor mRNA in the NAcc, and decreased D2 receptor mRNA and D2-like receptor binding in the VTA. Together, these data indicate that AMPH alters mesocorticolimbic DA neurotransmission in a region- and receptor-specific manner, which, in turn, could have profound consequences on social behavior in female prairie voles.
PubMedId: 20933511
Journal-article: Brain research, vol. 1367, 2010
Authors: Mary Kay Lobo, Herbert E Covington, Dipesh Chaudhury, Allyson K Friedman, HaoSheng Sun, Diane Damez-Werno, David M Dietz, Samir Zaman, Ja Wook Koo, Pamela J Kennedy, Ezekiell Mouzon, Murtaza Mogri, Rachael L Neve, Karl Deisseroth, Ming-Hu Han, Eric J Nestler
Abstract: The nucleus accumbens is a key mediator of cocaine reward, but the distinct roles of the two subpopulations of nucleus accumbens projection neurons, those expressing dopamine D1 versus D2 receptors, are poorly understood. We show that deletion of TrkB, the brain-derived neurotrophic factor (BDNF) receptor, selectively from D1+ or D2+ neurons oppositely affects cocaine reward. Because loss of TrkB in D2+ neurons increases their neuronal excitability, we next used optogenetic tools to control selectively the firing rate of D1+ and D2+ nucleus accumbens neurons and studied consequent effects on cocaine reward. Activation of D2+ neurons, mimicking the loss of TrkB, suppresses cocaine reward, with opposite effects induced by activation of D1+ neurons. These results provide insight into the molecular control of D1+ and D2+ neuronal activity as well as the circuit-level contribution of these cell types to cocaine reward.
PubMedId: 20947769
Journal-article: Science (New York, N.Y.), vol. 330, 2010
Authors: Yan Liu, Brandon J Aragona, Kimberly A Young, David M Dietz, Mohamed Kabbaj, Michelle Mazei-Robison, Eric J Nestler, Zuoxin Wang
Abstract: The prairie vole (Microtus ochrogaster) is a socially monogamous rodent species that forms pair bonds after mating, a behavior in which central dopamine (DA) has been implicated. Here, we used male prairie voles to examine the effects of drug exposure on pair bonding and related neural circuitry. In our first experiment, amphetamine (AMPH) motivated behavior was examined using a conditioned place preference (CPP) paradigm and was shown to be mediated by activation of D1-like DA receptors. Next, we examined the effects of repeated AMPH exposure on pair bonding. Intact and saline pretreated control males displayed mating-induced partner preferences, whereas males pretreated with AMPH at the doses effective to induce CPP failed to show mating-induced partner preferences. Such AMPH treatment also enhanced D1, but not D2, DA receptor expression in the nucleus accumbens (NAcc). Furthermore, pharmacological blockade of D1-like DA receptors in the NAcc rescued mating-induced partner preferences in AMPH-treated males. Together, our data indicate that repeated AMPH exposure may narrow the behavioral repertoire of male prairie voles via a DA receptor-specific mechanism in the NAcc, resulting in the impairment of pair bond formation.
PubMedId: 20080553
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 107, 2010
Authors: Quincey LaPlant, Vincent Vialou, Herbert E Covington, Dani Dumitriu, Jian Feng, Brandon L Warren, Ian Maze, David M Dietz, Emily L Watts, Sergio D I??iguez, Ja Wook Koo, Ezekiell Mouzon, William Renthal, Fiona Hollis, Hui Wang, Michele A Noonan, Yanhua Ren, Amelia J Eisch, Carlos A Bola??os, Mohamed Kabbaj, Guanghua Xiao, Rachael L Neve, Yasmin L Hurd, Ronald S Oosting, Gouping Fan, John H Morrison, Eric J Nestler
Abstract: Despite abundant expression of DNA methyltransferases (Dnmts) in brain, the regulation and behavioral role of DNA methylation remain poorly understood. We found that Dnmt3a expression was regulated in mouse nucleus accumbens (NAc) by chronic cocaine use and chronic social defeat stress. Moreover, NAc-specific manipulations that block DNA methylation potentiated cocaine reward and exerted antidepressant-like effects, whereas NAc-specific Dnmt3a overexpression attenuated cocaine reward and was pro-depressant. On a cellular level, we found that chronic cocaine use selectively increased thin dendritic spines on NAc neurons and that DNA methylation was both necessary and sufficient to mediate these effects. These data establish the importance of Dnmt3a in the NAc in regulating cellular and behavioral plasticity to emotional stimuli.
PubMedId: 20729844
Journal-article: Nature Neuroscience, vol. 13, pp. 1137-1143, 2010
Authors: D M Dietz, K C Dietz, E J Nestler, S J Russo
Abstract: Drug addiction is characterized by persistent behavioral and cellular plasticity throughout the brain's reward regions. Among the many neuroadaptations that occur following repeated drug administration are alterations in cell morphology including changes in dendritic spines. While this phenomenon has been well documented, the underlying molecular mechanisms are poorly understood. Here, within the context of drug abuse, we review and integrate several of the established pathways known to regulate synaptic remodeling, and discuss the contributions of neurotrophic and dopamine signaling in mediating this structural plasticity. Finally, we discuss how such upstream mechanisms could regulate actin dynamics, the common endpoint involved in structural remodeling in neurons.
PubMedId: 19434558
Journal-article: Pharmacopsychiatry, vol. 42 Suppl 1, 2009
Authors: Fiona Hollis, Hui Wang, David Dietz, Akash Gunjan, Mohamed Kabbaj
Abstract: Social stress has been linked to several neuropsychiatric diseases, including depression, which is a debilitating disease that has genetic, environmental, and epigenetic underpinnings. This study examined the effects of repeated social defeat on both depressive-like behaviors and histone acetylation in the hippocampus, amygdala, and dorsal prefrontal cortex of male Sprague-Dawley rats. Subjects were exposed to four consecutive social defeats. Depressive-like behaviors were assayed in the sucrose preference, forced swim, contextual fear, and social approach and avoidance tests. Histone H3 and H4 acetylation in the hippocampus, amygdala, and prefrontal cortex were examined by Western blots under basal conditions and at several time points. We also investigated the potential involvement of N-methyl-D: -aspartic acid (NMDA) receptors and glucocorticoid receptors (GR) by injecting respective antagonists prior to each social defeat and examining their effect on histone acetylation in the hippocampus. Social defeat resulted in behavioral changes in the forced swim, social avoidance, and contextual fear tests nearly 6 weeks after defeat, with no change in sucrose preference. Additionally, histone H3 acetylation was increased in the hippocampus 30 min following the last defeat and was not blocked by antagonism of either NMDA or GR receptors. There were no changes in histone H4 acetylation. These results indicate that social defeat induces several long-lasting depressive-like behaviors in rats and induces a significant, short-lived increase in H3 acetylation in the hippocampus, although the underlying mechanism behind this change warrants further investigation.
PubMedId: 20454892
Journal-article: Psychopharmacology, vol. 211, 2009
Authors: Scott J Russo, David M Dietz, Dani Dumitriu, John H Morrison, Robert C Malenka, Eric J Nestler
Abstract: Addictive drugs cause persistent restructuring of several neuronal cell types in the limbic regions of brain thought to be responsible for long-term behavioral plasticity driving addiction. Although these structural changes are well documented in nucleus accumbens medium spiny neurons, little is known regarding the underlying molecular mechanisms. Additionally, it remains unclear whether structural plasticity and its synaptic concomitants drive addictive behaviors or whether they reflect homeostatic compensations to the drug not related to addiction per se. Here, we discuss recent paradoxical data, which either support or oppose the hypothesis that drug-induced changes in dendritic spines drive addictive behavior. We define areas where future investigation can provide a more detailed picture of drug-induced synaptic reorganization, including ultrastructural, electrophysiological and behavioral studies.
PubMedId: 20207024
Journal-article: Trends in neurosciences, vol. 33, 2009
Authors: Ashley Stack, Nicole Carrier, David Dietz, Fiona Hollis, Jamie Sorenson, Mohamed Kabbaj
Abstract: Given both the high prevalence of anxiety disorders in women and the fact that little is known about the mechanisms of gender differences in anxiety, our primary aim in this study was to investigate the neurobiological mechanisms underlying sex differences in social anxiety-like behavior in rats. Through the use of zif268 antisense oligodeoxynucleotides (zif ASO), we induced a temporary downregulation of zif268 expression in the medial prefrontal cortex of male and female rats and found that zif268 ASO male rats show more social anxiety-like behaviors when compared with control male rats in the social interaction test. In fact, zif268 ASO males displayed social anxiety-like behaviors, which were similar to control females, thus downregulation of zif268 expression in the mPFC of male rats eliminated sex differences previously found in the social anxiety-like behavior tests. Interestingly, zif268 ASO in female rats had no effect on their social interaction. Our novel findings have led us to ascertain that sexually dimorphic zif268 expression in the mPFC is a key molecular factor in mediating sex-specific anxiety-like behavior in the social interaction test.
PubMedId: 19847159
Journal-article: Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, vol. 35, 2009
Authors: S. J. Russo, M. B. Wilkinson, M. S. Mazei-Robison, D. M. Dietz, I. Maze, V. Krishnan, W. Renthal, A. Graham, S. G. Birnbaum, T. A. Green, B. Robison, A. Lesselyong, L. I. Perrotti, C. A. Bolanos, A. Kumar, M. S. Clark, J. F. Neumaier, R. L. Neve, A. L. Bhakar, P. A. Barker, E. J. Nestler
Abstract: Although chronic cocaine-induced changes in dendritic spines on nucleus accumbens (NAc) neurons have been correlated with behavioral sensitization, the molecular pathways governing these structural changes, and their resulting behavioral effects, are poorly understood. The transcription factor, nuclear factor kappa B (NFkappaB), is rapidly activated by diverse stimuli and regulates expression of many genes known to maintain cell structure. Therefore, we evaluated the role of NFkappaB in regulating cocaine-induced dendritic spine changes on medium spiny neurons of the NAc and the rewarding effects of cocaine. We show that chronic cocaine induces NFkappaB-dependent transcription in the NAc of NFkappaB-Lac transgenic mice. This induction of NFkappaB activity is accompanied by increased expression of several NFkappaB genes, the promoters of which show chromatin modifications after chronic cocaine exposure consistent with their transcriptional activation. To study the functional significance of this induction, we used viral-mediated gene transfer to express either a constitutively active or dominant-negative mutant of Inhibitor of kappa B kinase (IKKca or IKKdn), which normally activates NFkappaB signaling, in the NAc. We found that activation of NFkappaB by IKKca increases the number of dendritic spines on NAc neurons, whereas inhibition of NFkappaB by IKKdn decreases basal dendritic spine number and blocks the increase in dendritic spines after chronic cocaine. Moreover, inhibition of NFkappaB blocks the rewarding effects of cocaine and the ability of previous cocaine exposure to increase an animal's preference for cocaine. Together, these studies establish a direct role for NFkappaB pathways in the NAc to regulate structural and behavioral plasticity to cocaine.
PubMedId: 19295158
Journal-article: Journal of Neuroscience, vol. 29, pp. 3529-3537, 2009
Authors: William Renthal, Arvind Kumar, Guanghua Xiao, Matthew Wilkinson, Herbert E Covington, Ian Maze, Devanjan Sikder, Alfred J Robison, Quincey LaPlant, David M Dietz, Scott J Russo, Vincent Vialou, Sumana Chakravarty, Thomas J Kodadek, Ashley Stack, Mohamed Kabbaj, Eric J Nestler
Abstract: Changes in gene expression contribute to the long-lasting regulation of the brain's reward circuitry seen in drug addiction; however, the specific genes regulated and the transcriptional mechanisms underlying such regulation remain poorly understood. Here, we used chromatin immunoprecipitation coupled with promoter microarray analysis to characterize genome-wide chromatin changes in the mouse nucleus accumbens, a crucial brain reward region, after repeated cocaine administration. Our findings reveal several interesting principles of gene regulation by cocaine and of the role of DeltaFosB and CREB, two prominent cocaine-induced transcription factors, in this brain region. The findings also provide comprehensive insight into the molecular pathways regulated by cocaine-including a new role for sirtuins (Sirt1 and Sirt2)-which are induced in the nucleus accumbens by cocaine and, in turn, dramatically enhance the behavioral effects of the drug.
PubMedId: 19447090
Journal-article: Neuron, vol. 62, 2008
Authors: D M Dietz, K C Dietz, S Moore, C C Ouimet, M Kabbaj
Abstract: In this study, we sought to examine individual differences in stress-induced behavioral sensitization to d-amphetamine after repeated social defeat stress. In an effort to understand what mechanisms underlie stress-induced sensitization to d-amphetamine, we examined striatal gene expression of the dopamine receptor D(2). Additionally, we investigated if repeated social defeat was associated with changes in dendritic spine density in the hippocampus, prefrontal cortex, and nucleus accumbens of rats that exhibit stress-induced sensitization. Male rats were classified into high responders (HR) and low responders (LR) based on their locomotor response to a novel environment. Then, rats were either handled as a control or defeated on four occasions by aggressive rats. Two weeks after the last defeat, animals were challenged with one of three doses of d-amphetamine and their locomotor activity was recorded. Non-defeated HR rats exhibited higher locomotor activity in response to d-amphetamine when compared to LR non-defeated rats. Fourteen days from the last repeated social defeat, LR rats and HR rats were behaviorally identical in response to acute injections of amphetamine. Furthermore, HR non-defeated rats had less D(2) mRNA expression in the nucleus accumbens core and dorsal striatum than do LR non-defeated rats. However, after repeated social defeat, HR and LR rats had identical D(2) mRNA expression in both the core and dorsal striatum. Finally, there were no changes in dendritic spine density in any of the brain areas examined in LR rats. Repeated social defeat abolishes individual differences in behavioral responses to d-amphetamine which may be due to a down-regulation of striatal dopamine D(2) receptors in LR rats.
PubMedId: 18415082
Journal-article: Psychopharmacology, vol. 198, 2007
Authors: David Dietz, Hui Wang, Mohamed Kabbaj
Abstract: In some rats, the hormone corticosterone is reinforcing. High novelty-seeking rats (high responders, HR) self-administered corticosterone at a much higher rate than low novelty-seeking rats (low responders, LR) do [Piazza PV, Deroche V, Deminiere JM, Maccari S, Le Moal M, Simon H, Corticosterone in the range of stress-induced levels possesses reinforcing properties: implications for sensation-seeking behaviors, Proc Natl Acad Sci USA 1993;90:11738-42]. While previous studies demonstrated that corticosterone reinforces nose poking in a self-administration paradigm, no studies to date have examined whether corticosterone is rewarding. Using the conditioned place preference (CPP) paradigm, we examined the rewarding effects of corticosterone in HR and LR rats. Male Sprague-Dawley rats were classified into HR and LR groups based on their locomotor activity in a novel environment. Subsequently, independent groups of HR and LR rats underwent CPP for corticosterone (0, 2.5 or 10 mg/kg; i.p.) or cocaine (12 mg/kg; i.p). CPP for cocaine was used as a positive control. While cocaine produced a strong CPP in both HR and LR rats, corticosterone failed to produce either preference or aversion in both phenotypes. Corticosterone is neither rewarding nor aversive in either behavioral phenotype.
PubMedId: 17521748
Journal-article: Behavioural brain research, vol. 181, 2006
Authors: Karen Lancaster, David M Dietz, Timothy H Moran, Mikhail V Pletnikov
Abstract: Autism spectrum disorders (ASD) have been the focus of a great deal of research and clinical speculation. This intense interest relates to both the perplexing pathogenesis and devastating consequences of these disorders. One of the obstacles to understanding the pathogenesis of autism and to developing efficient treatment has been the paucity of animal models that could be used for hypotheses-driven mechanistic studies of abnormal brain and behavior development and for the pre-clinical testing novel pharmacological treatments. In this report, we briefly review our animal model of ASD based on neonatal Borna disease virus (BDV) infection and present new data about abnormal social interaction in adult BDV-infected rats. We found that neonatal BDV infection profoundly affected social behaviors in adult rats. Compared to the control rats, both 90- and 180-day-old infected rats spent less time in active social interaction and more time in following their partners. In the intruder-resident test, the BDV-infected resident rats exhibited less aggression towards the intruders and showed more the following-the-intruder behavior. The following-the-partner behavior may be an example of "stereotypic" activity due to BDV-induced abnormal social communication between rats. The previously published results and present findings indicate that neonatal BDV infection significantly altered the normal pattern of social interaction in rats. Co-localization of activated microglia and dying Purkinje cells in BDV-infected rats suggests that the BDV model could be used to study a pathogenic link of Purkinje cell dropout and neuroinflammation to abnormal social behaviors.
PubMedId: 16860408
Journal-article: Behavioural brain research, vol. 176, 2006
Authors: David M Dietz, Kathleen S Curtis, Robert J Contreras
Abstract: Previous studies have shown that repeated sodium depletions using the natriuretic-diuretic furosemide induce progressive increases in NaCl ingestion. We investigated the role of taste in this behavioral sensitization in Sprague-Dawley rats using short-term lickometer testing along with 2-h stimulated intake tests. Our results show maximal licking across a range of NaCl concentrations after each of the three depletions, regardless of whether the solutions contained sucrose or were presented alone. Similarly, the presence of sucrose did not affect stimulated NaCl intake in long-term tests, although ingestion of NaCl solutions increased progressively with successive depletions. Finally, both licking and ingestion returned to baseline levels during need-free conditions. These results suggest that sodium imbalance acutely increases the salience of sodium taste and thereby the likelihood of NaCl ingestion, which may, in turn, contribute to progressive increases in NaCl intake that occur with multiple furosemide-induced sodium depletions.
PubMedId: 16306317
Journal-article: Chemical senses, vol. 31, 2005
Authors: David M Dietz, Jenica Tapocik, Meriem Gaval-Cruz, Mohamed Kabbaj
Abstract: Repeated administration of psychostimulants, such as amphetamine and cocaine, results in a long-lasting enhancement of behavioral responses elicited by a subsequent challenge injection of these drugs. This phenomenon has been termed behavioral sensitization. A well established model of individual differences based on the locomotor response to a novel environment has been shown to reliably predict the degree of behavioral sensitization to amphetamine. Rats that have high locomotor response in a novel environment (high responders or HR) develop greater behavioral sensitization to psychostimulants when compared to rats that show low locomotor activity in the same novel environment (low Responders or LR). Therefore, this model is ideal to study genetic factors that may underlie behavioral sensitization to psychostimulants. In this study, adult Sprague-Dawley rats were daily injected with amphetamine (1 mg/kg, i.p.) or saline for 9 days. Locomotor activity was recorded every other day. Following a one week-withdrawal a subsequent challenge of a lower dose of amphetamine (0.5 mg/kg, i.p.) was given to all rats (amphetamine pretreated and saline pretreated) and their locomotor activity was recorded. Our results show that HR rats, but not LR rats, develop behavioral sensitization to the locomotor activating effects of amphetamine. Furthermore, only HR rats pretreated with amphetamine exhibited an increase in dopamine transporter mRNA in the ventral tegmental area (VTA) and substantia nigra (SN). Tyrosine hydroxylase mRNA in the VTA and SN was upregulated in both HR and LR rats pretreated with amphetamine when compared to HR and LR rats pretreated with saline. These results demonstrate the existence of individual differences in behavioral sensitization to amphetamine and suggest that dopamine transporter, but not tyrosine hydroxylase, may be a critical factor in the development and expression of behavioral sensitization to the locomotor activating effects of amphetamine.
PubMedId: 16126238
Journal-article: Physiology & behavior, vol. 86, 2005
Authors: David Dietz, Michael Vogel, Steven Rubin, Timothy Moran, Kathryn Carbone, Mikhail Pletnikov
Abstract: Neonatal Borna disease virus (BDV) infection of the rat brain serves as a valuable model for studying the pathogenesis of neurodevelopmental abnormalities following early brain injury. Previous experiments have demonstrated significant alterations in regional tissue content of serotonin (5-HT) in neonatally BDV-infected Lewis rats. The present study sought to provide more insights into postnatal virus-associated alterations in 5-HT neurotransmission by evaluating the density of 5-HT1a receptors in the hippocampus and 5-HT2a receptors in the cortex, regional 5-HT tissue concentrations, behavioral responses to a 5-HT agonist, quipazine, and numbers of neurons in specific subfields of the hippocampus on days 7, 14, and 30 after neonatal BDV infection in Lewis rats. Neonatal BDV infection was found to be associated with a gradual increase in the density of 5-HT2a and 5-HT1a postsynaptic receptors followed by an elevation of 5-HT contents at both the levels of synaptic terminals (i.e., cortex and hippocampus) and cell bodies (i.e., raphe nuclei). In addition, there was an enhanced behavioral response to quipazine. Virus-associated neurochemical and behavioral changes were accompanied by a decline in the number of neurons in the dentate gyrus and in the CA1 field of the hippocampus. No change in the number of neurons in the CA3/2 field of the hippocampus was observed. The present pattern of BDV-associated alterations in 5-HT brain system along with available data from other laboratories suggest that BDV might compromise axonal transport and/or release of 5-HT, resulting in decreased 5-HT neurotransmission.
PubMedId: 15385249
Journal-article: Journal of neurovirology, vol. 10, 2004
Authors: David M Dietz, Mikhail V Pletnikov
PubMedId: 14637236
Comparative Study: Physiology & behavior, vol. 80, 2003
Presented by CUBS © 2024