Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Neurosci ; 22(9): 1413-1423, 2019 09.
Article in English | MEDLINE | ID: mdl-31427770

ABSTRACT

Understanding the transcriptional changes that are engaged in stress resilience may reveal novel antidepressant targets. Here we use gene co-expression analysis of RNA-sequencing data from brains of resilient mice to identify a gene network that is unique to resilience. Zfp189, which encodes a previously unstudied zinc finger protein, is the highest-ranked key driver gene in the network, and overexpression of Zfp189 in prefrontal cortical neurons preferentially activates this network and promotes behavioral resilience. The transcription factor CREB is a predicted upstream regulator of this network and binds to the Zfp189 promoter. To probe CREB-Zfp189 interactions, we employ CRISPR-mediated locus-specific transcriptional reprogramming to direct CREB or G9a (a repressive histone methyltransferase) to the Zfp189 promoter in prefrontal cortex neurons. Induction of Zfp189 with site-specific CREB is pro-resilient, whereas suppressing Zfp189 expression with G9a increases susceptibility. These findings reveal an essential role for Zfp189 and CREB-Zfp189 interactions in mediating a central transcriptional network of resilience.


Subject(s)
Adaptation, Psychological/physiology , Stress, Psychological/genetics , Zinc Fingers/genetics , Animals , Gene Regulatory Networks/genetics , Mice , Mice, Inbred C57BL , Prefrontal Cortex/metabolism , Transcription, Genetic
2.
Nat Commun ; 9(1): 3149, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30089879

ABSTRACT

The role of somatostatin interneurons in nucleus accumbens (NAc), a key brain reward region, remains poorly understood due to the fact that these cells account for < 1% of NAc neurons. Here, we use optogenetics, electrophysiology, and RNA-sequencing to characterize the transcriptome and functioning of NAc somatostatin interneurons after repeated exposure to cocaine. We find that the activity of somatostatin interneurons regulates behavioral responses to cocaine, with repeated cocaine reducing the excitability of these neurons. Repeated cocaine also induces transcriptome-wide changes in gene expression within NAc somatostatin interneurons. We identify the JUND transcription factor as a key regulator of cocaine action and confirmed, by use of viral-mediated gene transfer, that JUND activity in somatostatin interneurons influences behavioral responses to cocaine. Our results identify alterations in NAc induced by cocaine in a sparse population of somatostatin interneurons, and illustrate the value of studying brain diseases using cell type-specific whole transcriptome RNA-sequencing.


Subject(s)
Adaptation, Physiological/drug effects , Cocaine/pharmacology , Interneurons/drug effects , Interneurons/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Somatostatin/metabolism , Transcriptome , Animals , Brain/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Transfer Techniques , Locomotion , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Optogenetics/methods , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Reward , Sequence Analysis, RNA , Somatostatin/pharmacology , Transcription Factors/drug effects
3.
Proc Natl Acad Sci U S A ; 113(10): 2726-31, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26831103

ABSTRACT

The reinforcing and rewarding properties of cocaine are attributed to its ability to increase dopaminergic transmission in nucleus accumbens (NAc). This action reinforces drug taking and seeking and leads to potent and long-lasting associations between the rewarding effects of the drug and the cues associated with its availability. The inability to extinguish these associations is a key factor contributing to relapse. Dopamine produces these effects by controlling the activity of two subpopulations of NAc medium spiny neurons (MSNs) that are defined by their predominant expression of either dopamine D1 or D2 receptors. Previous work has demonstrated that optogenetically stimulating D1 MSNs promotes reward, whereas stimulating D2 MSNs produces aversion. However, we still lack a clear understanding of how the endogenous activity of these cell types is affected by cocaine and encodes information that drives drug-associated behaviors. Using fiber photometry calcium imaging we define D1 MSNs as the specific population of cells in NAc that encodes information about drug associations and elucidate the temporal profile with which D1 activity is increased to drive drug seeking in response to contextual cues. Chronic cocaine exposure dysregulates these D1 signals to both prevent extinction and facilitate reinstatement of drug seeking to drive relapse. Directly manipulating these D1 signals using designer receptors exclusively activated by designer drugs prevents contextual associations. Together, these data elucidate the responses of D1- and D2-type MSNs in NAc to acute cocaine and during the formation of context-reward associations and define how prior cocaine exposure selectively dysregulates D1 signaling to drive relapse.


Subject(s)
Cocaine/pharmacology , Neurons/drug effects , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Reward , Analysis of Variance , Animals , Cocaine/administration & dosage , Cues , Dopamine Uptake Inhibitors/administration & dosage , Dopamine Uptake Inhibitors/pharmacology , Drug-Seeking Behavior/drug effects , Immunohistochemistry , Mice, Inbred C57BL , Mice, Transgenic , Neuroimaging/methods , Neurons/metabolism , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...