Network


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

Hotspot


Dive into the research topics where Daniel J. Christoffel is active.

Publication


Featured researches published by Daniel J. Christoffel.


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 | 2014

Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress

Georgia E. Hodes; Madeline L. Pfau; Marylene Leboeuf; Sam A. Golden; Daniel J. Christoffel; Dana Bregman; Nicole Rebusi; Mitra Heshmati; Hossein Aleyasin; Brandon L. Warren; Benoit Labonté; Sarah R. Horn; Kyle A.B. Lapidus; Viktoria Stelzhammer; Erik H. F. Wong; Sabine Bahn; Vaishnav Krishnan; Carlos A. Bolaños-Guzmán; James W. Murrough; Miriam Merad; Scott J. Russo

Significance Depression and anxiety have been linked to increased inflammation. However, we do not know if inflammatory status predates onset of disease or whether it contributes to depression symptomatology. We report preexisting individual differences in the peripheral immune system that predict and promote stress susceptibility. Replacing a stress-naive animal’s peripheral immune system with that of a stressed animal increases susceptibility to social stress including repeated social defeat stress (RSDS) and witness defeat (a purely emotional form of social stress). Depleting the cytokine IL-6 from the whole body or just from leukocytes promotes resilience, as does sequestering IL-6 outside of the brain. These studies demonstrate that the emotional response to stress can be generated or blocked in the periphery, and offer a potential new form of treatment for stress disorders. Depression and anxiety disorders are associated with increased release of peripheral cytokines; however, their functional relevance remains unknown. Using a social stress model in mice, we find preexisting individual differences in the sensitivity of the peripheral immune system that predict and promote vulnerability to social stress. Cytokine profiles were obtained 20 min after the first social stress exposure. Of the cytokines regulated by stress, IL-6 was most highly up-regulated only in mice that ultimately developed a susceptible behavioral phenotype following a subsequent chronic stress, and levels remained elevated for at least 1 mo. We confirmed a similar elevation of serum IL-6 in two separate cohorts of patients with treatment-resistant major depressive disorder. Before any physical contact in mice, we observed individual differences in IL-6 levels from ex vivo stimulated leukocytes that predict susceptibility versus resilience to a subsequent stressor. To shift the sensitivity of the peripheral immune system to a pro- or antidepressant state, bone marrow (BM) chimeras were generated by transplanting hematopoietic progenitor cells from stress-susceptible mice releasing high IL-6 or from IL-6 knockout (IL-6−/−) mice. Stress-susceptible BM chimeras exhibited increased social avoidance behavior after exposure to either subthreshold repeated social defeat stress (RSDS) or a purely emotional stressor termed witness defeat. IL-6−/− BM chimeric and IL-6−/− mice, as well as those treated with a systemic IL-6 monoclonal antibody, were resilient to social stress. These data establish that preexisting differences in stress-responsive IL-6 release from BM-derived leukocytes functionally contribute to social stress-induced behavioral abnormalities.


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.


Nature Medicine | 2013

Epigenetic regulation of RAC1 induces synaptic remodeling in stress disorders and depression

Sam A. Golden; Daniel J. Christoffel; Mitra Heshmati; Georgia E. Hodes; Jane Magida; Keithara Davis; Michael E. Cahill; Caroline Dias; Efrain Ribeiro; Jessica L. Ables; Pamela J. Kennedy; Alfred J. Robison; Javier González-Maeso; Rachael L. Neve; Gustavo Turecki; Subroto Ghose; Carol A. Tamminga; Scott J. Russo

Depression induces structural and functional synaptic plasticity in brain reward circuits, although the mechanisms promoting these changes and their relevance to behavioral outcomes are unknown. Transcriptional profiling of the nucleus accumbens (NAc) for Rho GTPase–related genes, which are known regulators of synaptic structure, revealed a sustained reduction in RAS-related C3 botulinum toxin substrate 1 (Rac1) expression after chronic social defeat stress. This was associated with a repressive chromatin state surrounding the proximal promoter of Rac1. Inhibition of class 1 histone deacetylases (HDACs) with MS-275 rescued both the decrease in Rac1 transcription after social defeat stress and depression-related behavior, such as social avoidance. We found a similar repressive chromatin state surrounding the RAC1 promoter in the NAc of subjects with depression, which corresponded with reduced RAC1 transcription. Viral-mediated reduction of Rac1 expression or inhibition of Rac1 activity in the NAc increases social defeat–induced social avoidance and anhedonia in mice. Chronic social defeat stress induces the formation of stubby excitatory spines through a Rac1-dependent mechanism involving the redistribution of synaptic cofilin, an actin-severing protein downstream of Rac1. Overexpression of constitutively active Rac1 in the NAc of mice after chronic social defeat stress reverses depression-related behaviors and prunes stubby spines. Taken together, our data identify epigenetic regulation of RAC1 in the NAc as a disease mechanism in depression and reveal a functional role for Rac1 in rodents in regulating stress-related behaviors.Depression involves plasticity of brain reward neurons, although the mechanisms and behavioral relevance are unknown. Transcriptional profiling of nucleus accumbens (NAc) for RhoGTPase related genes, known regulators of synaptic structure, following chronic social defeat stress, revealed a long-term reduction in Rac1 transcription. This was marked by a repressive chromatin state surrounding its proximal promoter. Inhibition of class 1 HDACs with MS-275 rescued both decreased Rac1 transcription and social avoidance behavior. A similar repressive chromatin state was found surrounding the Rac1 promoter in human postmortem NAc from depressed subjects, which corresponded with reduced Rac1 transcription. We show Rac1 is necessary and sufficient for social avoidance and anhedonia, and the formation of stubby excitatory spines by redistributing synaptic cofilin, an actin severing protein downstream of Rac1. Our data identifies epigenetic regulation of Rac1 in NAc as a bona fide disease mechanism in depression and reveals a functional role in regulating stress-related behaviors.


Reviews in The Neurosciences | 2011

Structural and synaptic plasticity in stress-related disorders

Daniel J. Christoffel; Sam A. Golden; Scott J. Russo

Abstract Stress can have a lasting impact on the structure and function of brain circuitry that results in long-lasting changes in the behavior of an organism. Synaptic plasticity is the mechanism by which information is stored and maintained within individual synapses, neurons, and neuronal circuits to guide the behavior of an organism. Although these mechanisms allow the organism to adapt to its constantly evolving environment, not all of these adaptations are beneficial. Under prolonged bouts of physical or psychological stress, these mechanisms become dysregulated, and the connectivity between brain regions becomes unbalanced, resulting in pathological behaviors. In this review, we highlight the effects of stress on the structure and function of neurons within the mesocorticolimbic brain systems known to regulate mood and motivation. We then discuss the implications of these spine adaptations on neuronal activity and pathological behaviors implicated in mood disorders. Finally, we end by discussing recent brain imaging studies in human depression within the context of these basic findings to provide insight into the underlying mechanisms leading to neural dysfunction in depression.


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

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.


The Journal of Neuroscience | 2015

Sex Differences in Nucleus Accumbens Transcriptome Profiles Associated with Susceptibility versus Resilience to Subchronic Variable Stress

Georgia E. Hodes; Madeline L. Pfau; Immanuel Purushothaman; H. Francisca Ahn; Sam A. Golden; Daniel J. Christoffel; Jane Magida; Anna Brancato; Aki Takahashi; Meghan E. Flanigan; Caroline Ménard; Hossein Aleyasin; Ja Wook Koo; Zachary S. Lorsch; Jian Feng; Mitra Heshmati; Minghui Wang; Gustavo Turecki; Rachel Neve; Bin Zhang; Li Shen; Eric J. Nestler; Scott J. Russo

Depression and anxiety disorders are more prevalent in females, but the majority of research in animal models, the first step in finding new treatments, has focused predominantly on males. Here we report that exposure to subchronic variable stress (SCVS) induces depression-associated behaviors in female mice, whereas males are resilient as they do not develop these behavioral abnormalities. In concert with these different behavioral responses, transcriptional analysis of nucleus accumbens (NAc), a major brain reward region, by use of RNA sequencing (RNA-seq) revealed markedly different patterns of stress regulation of gene expression between the sexes. Among the genes displaying sex differences was DNA methyltransferase 3a (Dnmt3a), which shows a greater induction in females after SCVS. Interestingly, Dnmt3a expression levels were increased in the NAc of depressed humans, an effect seen in both males and females. Local overexpression of Dnmt3a in NAc rendered male mice more susceptible to SCVS, whereas Dnmt3a knock-out in this region rendered females more resilient, directly implicating this gene in stress responses. Associated with this enhanced resilience of female mice upon NAc knock-out of Dnmt3a was a partial shift of the NAc female transcriptome toward the male pattern after SCVS. These data indicate that males and females undergo different patterns of transcriptional regulation in response to stress and that a DNA methyltransferase in NAc contributes to sex differences in stress vulnerability. SIGNIFICANCE STATEMENT Women have a higher incidence of depression than men. However, preclinical models, the first step in developing new diagnostics and therapeutics, have been performed mainly on male subjects. Using a stress-based animal model of depression that causes behavioral effects in females but not males, we demonstrate a sex-specific transcriptional profile in brain reward circuitry. This transcriptional profile can be altered by removal of an epigenetic mechanism, which normally suppresses DNA transcription, creating a hybrid male/female transcriptional pattern. Removal of this epigenetic mechanism also induces behavioral resilience to stress in females. These findings shed new light onto molecular factors controlling sex differences in stress response.


Current Opinion in Neurobiology | 2015

Illuminating circuitry relevant to psychiatric disorders with optogenetics

Elizabeth E. Steinberg; Daniel J. Christoffel; Karl Deisseroth; Robert C. Malenka

The brains remarkable capacity to generate cognition and behavior is mediated by an extraordinarily complex set of neural interactions that remain largely mysterious. This complexity poses a significant challenge in developing therapeutic interventions to ameliorate psychiatric disease. Accordingly, few new classes of drugs have been made available for patients with mental illness since the 1950s. Optogenetics offers the ability to selectively manipulate individual neural circuit elements that underlie disease-relevant behaviors and is currently accelerating the pace of preclinical research into neurobiological mechanisms of disease. In this review, we highlight recent findings from studies that employ optogenetic approaches to gain insight into normal and aberrant brain function relevant to mental illness. Emerging data from these efforts offers an exquisitely detailed picture of disease-relevant neural circuits in action, and hints at the potential of optogenetics to open up entirely new avenues in the treatment of psychiatric disorders.


Nature | 2016

Basal forebrain projections to the lateral habenula modulate aggression reward

Sam A. Golden; Mitra Heshmati; Meghan E. Flanigan; Daniel J. Christoffel; Kevin G. Guise; Madeline L. Pfau; Hossein Aleyasin; Caroline Ménard; Hongxing Zhang; Georgia E. Hodes; Dana Bregman; Lena A. Khibnik; Jonathan Tai; Nicole Rebusi; Brian Krawitz; Dipesh Chaudhury; Jessica J. Walsh; Ming-Hu Han; Matt L. Shapiro; Scott J. Russo

Maladaptive aggressive behaviour is associated with a number of neuropsychiatric disorders and is thought to result partly from the inappropriate activation of brain reward systems in response to aggressive or violent social stimuli. Nuclei within the ventromedial hypothalamus, extended amygdala and limbic circuits are known to encode initiation of aggression; however, little is known about the neural mechanisms that directly modulate the motivational component of aggressive behaviour. Here we established a mouse model to measure the valence of aggressive inter-male social interaction with a smaller subordinate intruder as reinforcement for the development of conditioned place preference (CPP). Aggressors develop a CPP, whereas non-aggressors develop a conditioned place aversion to the intruder-paired context. Furthermore, we identify a functional GABAergic projection from the basal forebrain (BF) to the lateral habenula (lHb) that bi-directionally controls the valence of aggressive interactions. Circuit-specific silencing of GABAergic BF–lHb terminals of aggressors with halorhodopsin (NpHR3.0) increases lHb neuronal firing and abolishes CPP to the intruder-paired context. Activation of GABAergic BF–lHb terminals of non-aggressors with channelrhodopsin (ChR2) decreases lHb neuronal firing and promotes CPP to the intruder-paired context. Finally, we show that altering inhibitory transmission at BF–lHb terminals does not control the initiation of aggressive behaviour. These results demonstrate that the BF–lHb circuit has a critical role in regulating the valence of inter-male aggressive behaviour and provide novel mechanistic insight into the neural circuits modulating aggression reward processing.

Collaboration


Dive into the Daniel J. Christoffel's collaboration.

Top Co-Authors

Avatar

Scott J. Russo

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Sam A. Golden

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Georgia E. Hodes

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Mitra Heshmati

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Madeline L. Pfau

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric J. Nestler

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Hossein Aleyasin

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Jessica J. Walsh

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Ming-Hu Han

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge