Network


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

Hotspot


Dive into the research topics where Deveroux Ferguson is active.

Publication


Featured researches published by Deveroux Ferguson.


Nature | 2012

Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons

Dipesh Chaudhury; Jessica J. Walsh; Allyson K. Friedman; Barbara Juarez; Stacy M. Ku; Ja Wook Koo; Deveroux Ferguson; Hsing-Chen Tsai; Lisa E. Pomeranz; Daniel J. Christoffel; Alexander R. Nectow; Mats I. Ekstrand; Ana I. Domingos; Michelle S. Mazei-Robison; Ezekiell Mouzon; Mary Kay Lobo; Rachael L. Neve; Jeffrey M. Friedman; Scott J. Russo; Karl Deisseroth; Eric J. Nestler; Ming-Hu Han

Ventral tegmental area (VTA) dopamine neurons in the brain’s reward circuit have a crucial role in mediating stress responses, including determining susceptibility versus resilience to social-stress-induced behavioural abnormalities. VTA dopamine neurons show two in vivo patterns of firing: low frequency tonic firing and high frequency phasic firing. Phasic firing of the neurons, which is well known to encode reward signals, is upregulated by repeated social-defeat stress, a highly validated mouse model of depression. Surprisingly, this pathophysiological effect is seen in susceptible mice only, with no apparent change in firing rate in resilient individuals. However, direct evidence—in real time—linking dopamine neuron phasic firing in promoting the susceptible (depression-like) phenotype is lacking. Here we took advantage of the temporal precision and cell-type and projection-pathway specificity of optogenetics to show that enhanced phasic firing of these neurons mediates susceptibility to social-defeat stress in freely behaving mice. We show that optogenetic induction of phasic, but not tonic, firing in VTA dopamine neurons of mice undergoing a subthreshold social-defeat paradigm rapidly induced a susceptible phenotype as measured by social avoidance and decreased sucrose preference. Optogenetic phasic stimulation of these neurons also quickly induced a susceptible phenotype in previously resilient mice that had been subjected to repeated social-defeat stress. Furthermore, we show differences in projection-pathway specificity in promoting stress susceptibility: phasic activation of VTA neurons projecting to the nucleus accumbens (NAc), but not to the medial prefrontal cortex (mPFC), induced susceptibility to social-defeat stress. Conversely, optogenetic inhibition of the VTA–NAc projection induced resilience, whereas inhibition of the VTA–mPFC projection promoted susceptibility. Overall, these studies reveal novel firing-pattern- and neural-circuit-specific mechanisms of depression.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Nuclear factor-κB is a critical mediator of stress- impaired neurogenesis and depressive behavior

Ja Wook Koo; Scott J. Russo; Deveroux Ferguson; Eric J. Nestler; Ronald S. Duman

Proinflammatory cytokines, such as IL-1β, have been implicated in the cellular and behavioral effects of stress and in mood disorders, although the downstream signaling pathways underlying these effects have not been determined. In the present study, we demonstrate a critical role for NF-κB signaling in the actions of IL-1β and stress. Stress inhibition of neurogenesis in the adult hippocampus, which has been implicated in the prodepressive effects of stress, is blocked by administration of an inhibitor of NF-κB. Further analysis reveals that stress activates NF-κB signaling and decreases proliferation of neural stem-like cells but not early neural progenitor cells in the adult hippocampus. We also find that depressive-like behaviors caused by exposure to chronic stress are mediated by NF-κB signaling. Together, these data identify NF-κB signaling as a critical mediator of the antineurogenic and behavioral actions of stress and suggest previously undescribed therapeutical targets for depression.


The Journal of Neuroscience | 2011

IκB Kinase Regulates Social Defeat Stress-Induced Synaptic and Behavioral Plasticity

Daniel J. Christoffel; Sam A. Golden; Dani Dumitriu; Alfred J. Robison; William G.M. Janssen; H. Francisca Ahn; Vaishnav Krishnan; Cindy M. Reyes; Ming-Hu Han; Jessica L. Ables; Amelia J. Eisch; David M. Dietz; Deveroux Ferguson; Rachael L. Neve; Paul Greengard; Yong Kim; John H. Morrison; Scott J. Russo

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 IκB 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.


The Journal of Neuroscience | 2011

A Silent Synapse-Based Mechanism for Cocaine-Induced Locomotor Sensitization

Travis E. Brown; Brian R. Lee; Ping Mu; Deveroux Ferguson; David M. Dietz; Yoshinori N. Ohnishi; Ying Lin; Anna Suska; Masago Ishikawa; Yanhua H. Huang; Haowei Shen; Peter W. Kalivas; Barbara A. Sorg; Zukin Rs; Eric J. Nestler; Yan Dong; Oliver M. Schlüter

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.


The Journal of Neuroscience | 2014

Prefrontal cortical circuit for depression- and anxiety-related behaviors mediated by cholecystokinin: Role of ΔFosB

Vincent Vialou; Rosemary C. Bagot; Michael E. Cahill; Deveroux Ferguson; Alfred J. Robison; David M. Dietz; Barbara Fallon; Michelle S. Mazei-Robison; Stacy M. Ku; Eileen Harrigan; Catherine A. Winstanley; Tej Joshi; Jian Feng; Olivier Berton; Eric J. Nestler

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 ΔFosB in mPFC, specifically in the prelimbic (PrL) area, mediates susceptibility to stress. ΔFosB induction in PrL occurred selectively in susceptible mice after chronic social defeat stress, and overexpression of ΔFosB in this region, but not in the nearby infralimbic (IL) area, enhanced stress susceptibility. ΔFosB 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 ΔFosB and CCK through cortical projections to distinct subcortical targets.


Nature | 2014

β-catenin mediates stress resilience through Dicer1/microRNA regulation

Caroline Dias; Jian Feng; HaoSheng Sun; Ning Yi Shao; Michelle S. Mazei-Robison; Diane Damez-Werno; Kimberly N. Scobie; Rosemary C. Bagot; Benoit Labonté; Efrain Ribeiro; Xiaochuan Liu; Pamela J. Kennedy; Vincent Vialou; Deveroux Ferguson; Catherine J. Peña; Erin S. Calipari; Ja Wook Koo; Ezekiell Mouzon; Subroto Ghose; Carol A. Tamminga; Rachael L. Neve; Li Shen; Eric J. Nestler

β-catenin is a multi-functional protein that has an important role in the mature central nervous system; its dysfunction has been implicated in several neuropsychiatric disorders, including depression. Here we show that in mice β-catenin mediates pro-resilient and anxiolytic effects in the nucleus accumbens, a key brain reward region, an effect mediated by D2-type medium spiny neurons. Using genome-wide β-catenin enrichment mapping, we identify Dicer1—important in small RNA (for example, microRNA) biogenesis—as a β-catenin target gene that mediates resilience. Small RNA profiling after excising β-catenin from nucleus accumbens in the context of chronic stress reveals β-catenin-dependent microRNA regulation associated with resilience. Together, these findings establish β-catenin as a critical regulator in the development of behavioural resilience, activating a network that includes Dicer1 and downstream microRNAs. We thus present a foundation for the development of novel therapeutic targets to promote stress resilience.


Nature Neuroscience | 2012

Rac1 is essential in cocaine-induced structural plasticity of nucleus accumbens neurons.

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 Dietz; Kimberly N. Scobie; Deveroux Ferguson; Daniel J. Christoffel; Yoko H. Ohnishi; Georgia E. Hodes; Yi Zheng; Rachael L. Neve; Klaus M. Hahn; Scott J. Russo; Eric J. Nestler

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.


Nature Neuroscience | 2014

Locus-specific epigenetic remodeling controls addiction- and depression-related behaviors

Elizabeth A. Heller; Hannah M. Cates; Catherine J. Peña; HaoSheng Sun; Ningyi Shao; Jian Feng; Sam A. Golden; James P. Herman; Jessica J. Walsh; Michelle S. Mazei-Robison; Deveroux Ferguson; Scott W. Knight; Mark A. Gerber; Christian Nievera; Ming-Hu Han; Scott J. Russo; Carol S. Tamminga; Rachael L. Neve; Li Shen; H. Steve Zhang; Feng Zhang; Eric J. Nestler

Chronic exposure to drugs of abuse or stress regulates transcription factors, chromatin-modifying enzymes and histone post-translational modifications in discrete brain regions. Given the promiscuity of the enzymes involved, it has not yet been possible to obtain direct causal evidence to implicate the regulation of transcription and consequent behavioral plasticity by chromatin remodeling that occurs at a single gene. We investigated the mechanism linking chromatin dynamics to neurobiological phenomena by applying engineered transcription factors to selectively modify chromatin at a specific mouse gene in vivo. We found that histone methylation or acetylation at the Fosb locus in nucleus accumbens, a brain reward region, was sufficient to control drug- and stress-evoked transcriptional and behavioral responses via interactions with the endogenous transcriptional machinery. This approach allowed us to relate the epigenetic landscape at a given gene directly to regulation of its expression and to its subsequent effects on reward behavior.


Science | 2012

BDNF Is a Negative Modulator of Morphine Action

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

Regulating Opioid Responses Different drugs of abuse are thought to highjack similar reward systems in the brain using common mechanisms. However, Koo et al. (p. 124) now observe that some of the neural mechanisms that regulate opiate reward can be both different and even opposite to those that regulate reward by stimulant drugs. While knockdown of brain-derived neurotrophic factor (BDNF) in the ventral tegmental area in mice antagonized the response to cocaine, the same manipulation strengthened the potential of opiates to increase dopamine neuron excitability. Optogenetic stimulation of dopaminergic terminals in the nucleus accumbens could counteract the effects of BDNF on morphine reward blockade. Morphine reward is modulated by ventral tegmental area brain-derived neurotrophic factor in a way that is opposite to its modulation of cocaine reward. 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.


Nature Neuroscience | 2014

Stress and CRF gate neural activation of BDNF in the mesolimbic reward pathway

Jessica J. Walsh; Allyson K. Friedman; HaoSheng Sun; Elizabeth A. Heller; Stacy M. Ku; Barbara Juarez; Veronica L. Burnham; Michelle S. Mazei-Robison; Deveroux Ferguson; Sam A. Golden; Ja Wook Koo; Dipesh Chaudhury; Daniel J. Christoffel; Lisa E. Pomeranz; Jeffrey M. Friedman; Scott J. Russo; Eric J. Nestler; Ming-Hu Han

Mechanisms controlling release of brain-derived neurotrophic factor (BDNF) in the mesolimbic dopamine reward pathway remain unknown. We report that phasic optogenetic activation of this pathway increases BDNF amounts in the nucleus accumbens (NAc) of socially stressed mice but not of stress-naive mice. This stress gating of BDNF signaling is mediated by corticotrophin-releasing factor (CRF) acting in the NAc. These results unravel a stress context–detecting function of the brains mesolimbic circuit.

Collaboration


Dive into the Deveroux Ferguson'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

Jian Feng

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

HaoSheng Sun

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Li Shen

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Vincent Vialou

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Caroline Dias

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge