Network


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

Hotspot


Dive into the research topics where William Renthal is active.

Publication


Featured researches published by William Renthal.


Nature Neuroscience | 2006

Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action

Nadia M. Tsankova; Olivier Berton; William Renthal; Arvind Kumar; Rachel L. Neve; Eric J. Nestler

To better understand the molecular mechanisms of depression and antidepressant action, we administered chronic social defeat stress followed by chronic imipramine (a tricyclic antidepressant) to mice and studied adaptations at the levels of gene expression and chromatin remodeling of five brain-derived neurotrophic factor (Bdnf) splice variant mRNAs (I–V) and their unique promoters in the hippocampus. Defeat stress induced lasting downregulation of Bdnf transcripts III and IV and robustly increased repressive histone methylation at their corresponding promoters. Chronic imipramine reversed this downregulation and increased histone acetylation at these promoters. This hyperacetylation by chronic imipramine was associated with a selective downregulation of histone deacetylase (Hdac) 5. Furthermore, viral-mediated HDAC5 overexpression in the hippocampus blocked imipramines ability to reverse depression-like behavior. These experiments underscore an important role for histone remodeling in the pathophysiology and treatment of depression and highlight the therapeutic potential for histone methylation and deacetylation inhibitors in depression.


Cell | 2007

Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions.

Vaishnav Krishnan; Ming-Hu Han; Danielle L. Graham; Olivier Berton; William Renthal; Scott J. Russo; Quincey LaPlant; Ami Graham; Michael Lutter; Diane C. Lagace; Subroto Ghose; Robin Reister; Paul Tannous; Thomas A. Green; Rachael L. Neve; Sumana Chakravarty; Arvind Kumar; Amelia J. Eisch; David W. Self; Francis S. Lee; Carol A. Tamminga; Donald C. Cooper; Howard K. Gershenfeld; Eric J. Nestler

While stressful life events are an important cause of psychopathology, most individuals exposed to adversity maintain normal psychological functioning. The molecular mechanisms underlying such resilience are poorly understood. Here, we demonstrate that an inbred population of mice subjected to social defeat can be separated into susceptible and unsusceptible subpopulations that differ along several behavioral and physiological domains. By a combination of molecular and electrophysiological techniques, we identify signature adaptations within the mesolimbic dopamine circuit that are uniquely associated with vulnerability or insusceptibility. We show that molecular recapitulations of three prototypical adaptations associated with the unsusceptible phenotype are each sufficient to promote resistant behavior. Our results validate a multidisciplinary approach to examine the neurobiological mechanisms of variations in stress resistance, and illustrate the importance of plasticity within the brains reward circuits in actively maintaining an emotional homeostasis.


Neuron | 2005

Chromatin Remodeling Is a Key Mechanism Underlying Cocaine-Induced Plasticity in Striatum

Arvind Kumar; Kwang Ho Choi; William Renthal; Nadia M. Tsankova; David E.H. Theobald; Hoang Trang Truong; Scott J. Russo; Quincey LaPlant; Teresa Sasaki; Kimberly N. Whistler; Rachael L. Neve; David W. Self; Eric J. Nestler

Given that cocaine induces neuroadaptations through regulation of gene expression, we investigated whether chromatin remodeling at specific gene promoters may be a key mechanism. We show that cocaine induces specific histone modifications at different gene promoters in striatum, a major neural substrate for cocaines behavioral effects. At the cFos promoter, H4 hyperacetylation is seen within 30 min of a single cocaine injection, whereas no histone modifications were seen with chronic cocaine, consistent with cocaines ability to induce cFos acutely, but not chronically. In contrast, at the BDNF and Cdk5 promoters, genes that are induced by chronic, but not acute, cocaine, H3 hyperacetylation was observed with chronic cocaine only. DeltaFosB, a cocaine-induced transcription factor, appears to mediate this regulation of the Cdk5 gene. Furthermore, modulating histone deacetylase activity alters locomotor and rewarding responses to cocaine. Thus, chromatin remodeling is an important regulatory mechanism underlying cocaine-induced neural and behavioral plasticity.


Neuron | 2007

Histone Deacetylase 5 Epigenetically Controls Behavioral Adaptations to Chronic Emotional Stimuli

William Renthal; Ian Maze; Vaishnav Krishnan; Herbert E. Covington; Guanghua Xiao; Arvind Kumar; Scott J. Russo; Ami Graham; Nadia M. Tsankova; Tod E. Kippin; Kerry A. Kerstetter; Rachael L. Neve; Stephen J. Haggarty; Timothy A. McKinsey; Rhonda Bassel-Duby; Eric N. Olson; Eric J. Nestler

Previous work has identified alterations in histone acetylation in animal models of drug addiction and depression. However, the mechanisms which integrate drugs and stress with changes in chromatin structure remain unclear. Here, we identify the activity-dependent class II histone deacetylase, HDAC5, as a central integrator of these stimuli with changes in chromatin structure and gene expression. Chronic, but not acute, exposure to cocaine or stress decreases HDAC5 function in the nucleus accumbens (NAc), a major brain reward region, which allows for increased histone acetylation and transcription of HDAC5 target genes. This regulation is behaviorally important, as loss of HDAC5 causes hypersensitive responses to chronic, not acute, cocaine or stress. These findings suggest that proper balance of histone acetylation is a crucial factor in the saliency of a given stimulus and that disruption of this balance is involved in the transition from an acute adaptive response to a chronic psychiatric illness.


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.


The Journal of Neuroscience | 2009

Antidepressant Actions of Histone Deacetylase Inhibitors

Herbert E. Covington; Ian Maze; Quincey LaPlant; Vincent Vialou; Yoshinori N. Ohnishi; Olivier Berton; Dan M. Fass; William Renthal; A.J. Rush; Emma Y. Wu; Subroto Ghose; Vaishnav Krishnan; Scott J. Russo; Carol A. Tamminga; Stephen J. Haggarty; Eric J. Nestler

Persistent symptoms of depression suggest the involvement of stable molecular adaptations in brain, which may be reflected at the level of chromatin remodeling. We find that chronic social defeat stress in mice causes a transient decrease, followed by a persistent increase, in levels of acetylated histone H3 in the nucleus accumbens, an important limbic brain region. This persistent increase in H3 acetylation is associated with decreased levels of histone deacetylase 2 (HDAC2) in the nucleus accumbens. Similar effects were observed in the nucleus accumbens of depressed humans studied postmortem. These changes in H3 acetylation and HDAC2 expression mediate long-lasting positive neuronal adaptations, since infusion of HDAC inhibitors into the nucleus accumbens, which increases histone acetylation, exerts robust antidepressant-like effects in the social defeat paradigm and other behavioral assays. HDAC inhibitor [N-(2-aminophenyl)-4-[N-(pyridine-3-ylmethoxy-carbonyl)aminomethyl]benzamide (MS-275)] infusion also reverses the effects of chronic defeat stress on global patterns of gene expression in the nucleus accumbens, as determined by microarray analysis, with striking similarities to the effects of the standard antidepressant fluoxetine. Stress-regulated genes whose expression is normalized selectively by MS-275 may provide promising targets for the future development of novel antidepressant treatments. Together, these findings provide new insight into the underlying molecular mechanisms of depression and antidepressant action, and support the antidepressant potential of HDAC inhibitors and perhaps other agents that act at the level of chromatin structure.


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.


Trends in Molecular Medicine | 2008

Epigenetic Mechanisms in Drug Addiction

William Renthal; Eric J. Nestler

Changes in gene expression in brain reward regions are thought to contribute to the pathogenesis and persistence of drug addiction. Recent studies have begun to focus on the molecular mechanisms by which drugs of abuse and related environmental stimuli, such as drug-associated cues or stress, converge on the genome to alter specific gene programs. Increasing evidence suggests that these stable gene expression changes in neurons are mediated in part by epigenetic mechanisms that alter chromatin structure on specific gene promoters. This review discusses recent findings from behavioral, molecular and bioinformatic approaches being used to understand the complex epigenetic regulation of gene expression by drugs of abuse. This novel mechanistic insight might open new avenues for improved treatments of drug addiction.


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.


Neuron | 2008

Cocaine Regulates MEF2 to Control Synaptic and Behavioral Plasticity

Suprabha Pulipparacharuvil; William Renthal; Carly F. Hale; Makoto Taniguchi; Guanghua Xiao; Arvind Kumar; Scott J. Russo; Devanjan Sikder; Colleen M. Dewey; Maya M. Davis; Paul Greengard; Angus C. Nairn; Eric J. Nestler; Christopher W. Cowan

Repeated exposure to cocaine causes sensitized behavioral responses and increased dendritic spines on medium spiny neurons of the nucleus accumbens (NAc). We find that cocaine regulates myocyte enhancer factor 2 (MEF2) transcription factors to control these two processes in vivo. Cocaine suppresses striatal MEF2 activity in part through a mechanism involving cAMP, the regulator of calmodulin signaling (RCS), and calcineurin. We show that reducing MEF2 activity in the NAc in vivo is required for the cocaine-induced increases in dendritic spine density. Surprisingly, we find that increasing MEF2 activity in the NAc, which blocks the cocaine-induced increase in dendritic spine density, enhances sensitized behavioral responses to cocaine. Together, our findings implicate MEF2 as a key regulator of structural synapse plasticity and sensitized responses to cocaine and suggest that reducing MEF2 activity (and increasing spine density) in NAc may be a compensatory mechanism to limit long-lasting maladaptive behavioral responses to cocaine.

Collaboration


Dive into the William Renthal's collaboration.

Top Co-Authors

Avatar

Eric J. Nestler

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Arvind Kumar

Centre for Cellular and Molecular Biology

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

Quincey LaPlant

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
Top Co-Authors

Avatar

Rachael L. Neve

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Guanghua Xiao

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nadia M. Tsankova

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

David W. Self

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge