Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David M. Dietz is active.

Publication


Featured researches published by David M. Dietz.


Science | 2010

Cell type-specific loss of BDNF signaling mimics optogenetic control of cocaine reward.

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

BDNF, Dopamine, and Cocaine Reward The nucleus accumbens plays a crucial role in mediating the rewarding effects of drugs of abuse. Different subpopulations of nucleus accumbens projection neurons exhibit balanced but antagonistic influences on their downstream outputs and behaviors. However, their roles in regulating reward behaviors remains unclear. Lobo et al. (p. 385) evaluated the roles of the two subtypes of nucleus accumbens projection neurons, those expressing dopamine D1 versus D2 receptors, in cocaine reward. Deleting TrkB, the receptor for brain-derived neurotrophic factor, selectively in each cell type, and selectively controlling the firing of each cell type using optogenetic techniques allowed for confirmation that D1- and D2-containing neurons produced opposite effects on cocaine reward. Selective manipulation of neuron subtypes produces opposite effects on behavioral responses to cocaine. 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.


Science | 2010

Essential Role of the Histone Methyltransferase G9a in Cocaine-induced Plasticity

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

Cocaine Addiction and Histone Methylation Long-lasting behavioral syndromes associated with chronic cocaine exposure may result from dysregulation of the global transcriptional machinery. Maze et al. (p. 213) observed that histone lysine methylation in the nucleus accumbens plays a critical role in mediating the regulation of gene expression in response to repeated cocaine self-administration. Chronic cocaine was linked to overall reductions in dimethylation of lysine 9 of histone 3 (H3K9) in this brain region. Repressing H3K9 after chronic cocaine administration facilitated reward-related changes in behavior. The authors identifed the methyltransferase G9a as an essential mediator and an important regulator of dendritic spine plasticity. Downregulation of G9a was linked to the transcription factor ΔFosB. Cocaine suppression of histone methylation in the nucleus accumbens mediates the drug’s ability to enhance reward. 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 ∆FosB. 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.


Trends in Neurosciences | 2010

The Addicted Synapse: Mechanisms of Synaptic and Structural Plasticity in Nucleus Accumbens

Scott J. Russo; David M. Dietz; Dani Dumitriu; John H. Morrison; Robert C. Malenka; Eric J. Nestler

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.


Nature Neuroscience | 2010

Dnmt3a regulates emotional behavior and spine plasticity in the nucleus accumbens

Quincey LaPlant; Vincent Vialou; Herbert E. Covington; Dani Dumitriu; Jian Feng; Brandon L. Warren; Ian Maze; David M. Dietz; Emily L. Watts; Sergio D. Iñiguez; Ja Wook Koo; Ezekiell Mouzon; William Renthal; Fiona Hollis; Hui Wang; Michele A. Noonan; Yanhua Ren; Amelia J. Eisch; Carlos A. Bolaños; Mohamed Kabbaj; Guanghua Xiao; Rachael L. Neve; Yasmin L. Hurd; Ronald S. Oosting; Gouping Fan; John H. Morrison; Eric J. Nestler

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.


Nature Neuroscience | 2010

[Delta]FosB in brain reward circuits mediates resilience to stress and antidepressant responses

Vincent Vialou; Alfred J. Robison; Quincey LaPlant; Herbert E. Covington; David M. Dietz; Yoshinori N. Ohnishi; Ezekiell Mouzon; A.J. Rush; Emily L. Watts; Deanna L. Wallace; Sergio D. Iñiguez; Yoko H. Ohnishi; Michel A. Steiner; Brandon L. Warren; Vaishnav Krishnan; Carlos A. Bolaños; Rachael L. Neve; Subroto Ghose; Olivier Berton; Carol A. Tamminga; Eric J. Nestler

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 ΔFosB mediates an essential mechanism of resilience in mice. Induction of ΔFosB in the nucleus accumbens, an important brain reward-associated region, in response to chronic social defeat stress was both necessary and sufficient for resilience. ΔFosB induction was also required for the standard antidepressant fluoxetine to reverse behavioral pathology induced by social defeat. ΔFosB 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.


Nature Neuroscience | 2012

Impaired adult myelination in the prefrontal cortex of socially isolated mice

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 L. Dupree; Patrizia Casaccia

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.


Neuron | 2009

Genome-wide Analysis of Chromatin Regulation by Cocaine Reveals a Role for Sirtuins

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 Kodadek; Ashley Stack; Mohammed Kabbaj; Eric J. Nestler

Changes in gene expression contribute to the long-lasting regulation of the brains 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.


Biological Psychiatry | 2011

Paternal Transmission of Stress-Induced Pathologies

David M. Dietz; Quincey LaPlant; Emily L. Watts; Georgia E. Hodes; Scott J. Russo; Jian Feng; Ronald S. Oosting; Vincent Vialou; Eric J. Nestler

BACKGROUND 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. METHODS 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. RESULTS 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. CONCLUSIONS 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.


The Journal of Neuroscience | 2009

Nuclear factor kB signaling regulates neuronal morphology and cocaine reward

Scott J. Russo; Matthew Wilkinson; Michelle S. Mazei-Robison; David M. Dietz; Ian Maze; Vaishnav Krishnan; William Renthal; Ami Graham; Shari G. Birnbaum; Thomas A. Green; Bruce Robison; Alan Lesselyong; Linda I. Perrotti; Carlos A. Bolaños; Arvind Kumar; Michael S. Clark; John F. Neumaier; Rachael L. Neve; Asha L. Bhakar; Philip A. Barker; Eric J. Nestler

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 κ B (NFκB), is rapidly activated by diverse stimuli and regulates expression of many genes known to maintain cell structure. Therefore, we evaluated the role of NFκB 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 NFκB-dependent transcription in the NAc of NFκB-Lac transgenic mice. This induction of NFκB activity is accompanied by increased expression of several NFκB 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 κ B kinase (IKKca or IKKdn), which normally activates NFκB signaling, in the NAc. We found that activation of NFκB by IKKca increases the number of dendritic spines on NAc neurons, whereas inhibition of NFκB by IKKdn decreases basal dendritic spine number and blocks the increase in dendritic spines after chronic cocaine. Moreover, inhibition of NFκB blocks the rewarding effects of cocaine and the ability of previous cocaine exposure to increase an animals preference for cocaine. Together, these studies establish a direct role for NFκB pathways in the NAc to regulate structural and behavioral plasticity to cocaine.


Nature Neuroscience | 2012

HDAC2 regulates atypical antipsychotic responses through the modulation of mGlu2 promoter activity.

Mitsumasa Kurita; Terrell Holloway; Aintzane García-Bea; Alexey Kozlenkov; Allyson K. Friedman; José L. Moreno; Mitra Heshmati; Sam A. Golden; Pamela J. Kennedy; Nagahide Takahashi; David M. Dietz; Giuseppe Mocci; Ane M. Gabilondo; James B. 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 González-Maeso

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-HT2A 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.

Collaboration


Dive into the David M. Dietz's collaboration.

Top Co-Authors

Avatar

Eric J. Nestler

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Rachael L. Neve

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

HaoSheng Sun

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Scott J. Russo

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Diane Damez-Werno

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Ezekiell Mouzon

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Ja Wook Koo

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Ian Maze

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge