Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell ; 149(5): 1152-63, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632977

ABSTRACT

Our understanding of current treatments for depression, and the development of more specific therapies, is limited by the complexity of the circuits controlling mood and the distributed actions of antidepressants. Although the therapeutic efficacy of serotonin-specific reuptake inhibitors (SSRIs) is correlated with increases in cortical activity, the cell types crucial for their action remain unknown. Here we employ bacTRAP translational profiling to show that layer 5 corticostriatal pyramidal cells expressing p11 (S100a10) are strongly and specifically responsive to chronic antidepressant treatment. This response requires p11 and includes the specific induction of Htr4 expression. Cortex-specific deletion of p11 abolishes behavioral responses to SSRIs, but does not lead to increased depression-like behaviors. Our data identify corticostriatal projection neurons as critical for the response to antidepressants, and suggest that the regulation of serotonergic tone in this single cell type plays a pivotal role in antidepressant therapy.


Subject(s)
Antidepressive Agents/metabolism , Depression/drug therapy , Neurons/cytology , Prefrontal Cortex/cytology , Selective Serotonin Reuptake Inhibitors/metabolism , Animals , Antidepressive Agents/pharmacology , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , S100 Proteins/genetics , S100 Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
2.
Mol Pharmacol ; 105(4): 272-285, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38351270

ABSTRACT

The signal transduction protein, regulator of G protein signaling 4 (RGS4), plays a prominent role in physiologic and pharmacological responses by controlling multiple intracellular pathways. Our earlier work identified the dynamic but distinct roles of RGS4 in the efficacy of monoamine-targeting versus fast-acting antidepressants. Using a modified chronic variable stress (CVS) paradigm in mice, we demonstrate that stress-induced behavioral abnormalities are associated with the downregulation of RGS4 in the medial prefrontal cortex (mPFC). Knockout of RGS4 (RGS4KO) increases susceptibility to CVS, as mutant mice develop behavioral abnormalities as early as 2 weeks after CVS resting-state functional magnetic resonance imaging I (rs-fMRI) experiments indicate that stress susceptibility in RGS4KO mice is associated with changes in connectivity between the mediodorsal thalamus (MD-THL) and the mPFC. Notably, RGS4KO also paradoxically enhances the antidepressant efficacy of ketamine in the CVS paradigm. RNA-sequencing analysis of naive and CVS samples obtained from mPFC reveals that RGS4KO triggers unique gene expression signatures and affects several intracellular pathways associated with human major depressive disorder. Our analysis suggests that ketamine treatment in the RGS4KO group triggers changes in pathways implicated in synaptic activity and responses to stress, including pathways associated with axonal guidance and myelination. Overall, we show that reducing RGS4 activity triggers unique gene expression adaptations that contribute to chronic stress disorders and that RGS4 is a negative modulator of ketamine actions. SIGNIFICANCE STATEMENT: Chronic stress promotes robust maladaptation in the brain, but the exact intracellular pathways contributing to stress vulnerability and mood disorders have not been thoroughly investigated. In this study, the authors used murine models of chronic stress and multiple methodologies to demonstrate the critical role of the signal transduction modulator regulator of G protein signaling 4 in the medial prefrontal cortex in vulnerability to chronic stress and the efficacy of the fast-acting antidepressant ketamine.


Subject(s)
Depressive Disorder, Major , Ketamine , RGS Proteins , Mice , Humans , Animals , Ketamine/pharmacology , Transcriptome , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Mice, Knockout , RGS Proteins/genetics , RGS Proteins/metabolism , Antidepressive Agents/pharmacology , Antidepressive Agents/metabolism , Prefrontal Cortex/metabolism , Gene Expression Profiling , GTP-Binding Proteins/metabolism
3.
Cell ; 135(4): 749-62, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-19013282

ABSTRACT

Comparative analysis can provide important insights into complex biological systems. As demonstrated in the accompanying paper, translating ribosome affinity purification (TRAP) permits comprehensive studies of translated mRNAs in genetically defined cell populations after physiological perturbations. To establish the generality of this approach, we present translational profiles for 24 CNS cell populations and identify known cell-specific and enriched transcripts for each population. We report thousands of cell-specific mRNAs that were not detected in whole-tissue microarray studies and provide examples that demonstrate the benefits deriving from comparative analysis. To provide a foundation for further biological and in silico studies, we provide a resource of 16 transgenic mouse lines, their corresponding anatomic characterization, and translational profiles for cell types from a variety of central nervous system structures. This resource will enable a wide spectrum of molecular and mechanistic studies of both well-known and previously uncharacterized neural cell populations.


Subject(s)
Brain/metabolism , Genetic Techniques , Protein Biosynthesis , Animals , Central Nervous System/metabolism , Chromosomes, Artificial, Bacterial/metabolism , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Mice , Mice, Transgenic , Models, Biological , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Ribosomes/metabolism
4.
Mol Psychiatry ; 26(6): 2334-2349, 2021 06.
Article in English | MEDLINE | ID: mdl-33441982

ABSTRACT

Serotonin receptor 4 (5-HT4R) plays an important role in regulating mood, anxiety, and cognition, and drugs that activate this receptor have fast-acting antidepressant (AD)-like effects in preclinical models. However, 5-HT4R is widely expressed throughout the central nervous system (CNS) and periphery, making it difficult to pinpoint the cell types and circuits underlying its effects. Therefore, we generated a Cre-dependent 5-HT4R knockout mouse line to dissect the function of 5-HT4R in specific brain regions and cell types. We show that the loss of functional 5-HT4R specifically from excitatory neurons of hippocampus led to robust AD-like behavioral responses and an elevation in baseline anxiety. 5-HT4R was necessary to maintain the proper excitability of dentate gyrus (DG) granule cells and cell type-specific molecular profiling revealed a dysregulation of genes necessary for normal neural function and plasticity in cells lacking 5-HT4R. These adaptations were accompanied by an increase in the number of immature neurons in ventral, but not dorsal, dentate gyrus, indicating a broad impact of 5-HT4R loss on the local cellular environment. This study is the first to use conditional genetic targeting to demonstrate a direct role for hippocampal 5-HT4R signaling in modulating mood and anxiety. Our findings also underscore the need for cell type-based approaches to elucidate the complex action of neuromodulatory systems on distinct neural circuits.


Subject(s)
Anxiety , Hippocampus , Animals , Dentate Gyrus/metabolism , Hippocampus/metabolism , Mice , Neurons/metabolism , Receptors, Serotonin , Receptors, Serotonin, 5-HT4/genetics , Receptors, Serotonin, 5-HT4/metabolism
5.
PLoS Pathog ; 15(11): e1008014, 2019 11.
Article in English | MEDLINE | ID: mdl-31703116

ABSTRACT

Clostridium perfringens epsilon toxin (ETX) is responsible for causing the economically devastating disease, enterotoxaemia, in livestock. It is well accepted that ETX causes blood brain barrier (BBB) permeability, however the mechanisms involved in this process are not well understood. Using in vivo and in vitro methods, we determined that ETX causes BBB permeability in mice by increasing caveolae-dependent transcytosis in brain endothelial cells. When mice are intravenously injected with ETX, robust ETX binding is observed in the microvasculature of the central nervous system (CNS) with limited to no binding observed in the vasculature of peripheral organs, indicating that ETX specifically targets CNS endothelial cells. ETX binding to CNS microvasculature is dependent on MAL expression, as ETX binding to CNS microvasculature of MAL-deficient mice was not detected. ETX treatment also induces extravasation of molecular tracers including 376Da fluorescein salt, 60kDA serum albumin, 70kDa dextran, and 155kDA IgG. Importantly, ETX-induced BBB permeability requires expression of both MAL and caveolin-1, as mice deficient in MAL or caveolin-1 did not exhibit ETX-induced BBB permeability. Examination of primary murine brain endothelial cells revealed an increase in caveolae in ETX-treated cells, resulting in dynamin and lipid raft-dependent vacuolation without cell death. ETX-treatment also results in a rapid loss of EEA1 positive early endosomes and accumulation of large, RAB7-positive late endosomes and multivesicular bodies. Based on these results, we hypothesize that ETX binds to MAL on the apical surface of brain endothelial cells, causing recruitment of caveolin-1, triggering caveolae formation and internalization. Internalized caveolae fuse with early endosomes which traffic to late endosomes and multivesicular bodies. We believe that these multivesicular bodies fuse basally, releasing their contents into the brain parenchyma.


Subject(s)
Bacterial Toxins/pharmacology , Blood-Brain Barrier/physiopathology , Brain/physiopathology , Caveolin 1/physiology , Cell Membrane Permeability/physiology , Myelin and Lymphocyte-Associated Proteolipid Proteins/physiology , Transcytosis/drug effects , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Caveolae/drug effects , Caveolae/metabolism , Cell Membrane Permeability/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Mol Psychiatry ; 25(5): 1112-1129, 2020 05.
Article in English | MEDLINE | ID: mdl-31431686

ABSTRACT

In mood disorders, psychomotor and sensory abnormalities are prevalent, disabling, and intertwined with emotional and cognitive symptoms. Corticostriatal neurons in motor and somatosensory cortex are implicated in these symptoms, yet mechanisms of their vulnerability are unknown. Here, we demonstrate that S100a10 corticostriatal neurons exhibit distinct serotonin responses and have increased excitability, compared with S100a10-negative neurons. We reveal that prolonged social isolation disrupts the specific serotonin response which gets restored by chronic antidepressant treatment. We identify cell-type-specific transcriptional signatures in S100a10 neurons that contribute to serotonin responses and strongly associate with psychomotor and somatosensory function. Our studies provide a strong framework to understand the pathogenesis and create new avenues for the treatment of mood disorders.


Subject(s)
Annexin A2/metabolism , Antidepressive Agents/pharmacology , Neurons/drug effects , Neurons/metabolism , S100 Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/metabolism , Animals , Biomarkers/metabolism , Male , Mice , Motor Cortex/pathology , Serotonin/metabolism , Somatosensory Cortex/pathology , Stress, Psychological/physiopathology
7.
Proc Natl Acad Sci U S A ; 109(28): 11360-5, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733786

ABSTRACT

A large number of studies have demonstrated that the nucleus accumbens (NAC) is a critical site in the neuronal circuits controlling reward responses, motivation, and mood, but the neuronal cell type(s) underlying these processes are not yet known. Identification of the neuronal cell types that regulate depression-like states will guide us in understanding the biological basis of mood and its regulation by diseases like major depressive disorder. Taking advantage of recent findings demonstrating that the serotonin receptor chaperone, p11, is an important molecular regulator of depression-like states, here we identify cholinergic interneurons (CINs) as a primary site of action for p11 in the NAC. Depression-like behavior is observed in mice after decrease of p11 levels in NAC CINs. This phenotype is recapitulated by silencing neuronal transmission in these cells, demonstrating that accumbal cholinergic neuronal activity regulates depression-like behaviors and suggesting that accumbal CIN activity is crucial for the regulation of mood and motivation.


Subject(s)
Annexin A2/metabolism , Depression/physiopathology , Interneurons/metabolism , Nucleus Accumbens/metabolism , S100 Proteins/metabolism , Acetylcholine/metabolism , Animals , Antidepressive Agents/pharmacology , Behavior, Animal , Depression/metabolism , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Chaperones/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Phenotype , Receptors, Cholinergic/metabolism
8.
Front Neurosci ; 18: 1353131, 2024.
Article in English | MEDLINE | ID: mdl-38389788

ABSTRACT

Background: Post-traumatic stress disorder (PTSD) is a highly prevalent psychiatric disorder that can become chronic and debilitating when left untreated. Available pharmacotherapies are limited, take weeks to show modest benefit and remain ineffective for up to 40% of patients. Methylone is currently in clinical development for the treatment of PTSD. Preclinical studies show rapid, robust and long-lasting antidepressant-like and anxiolytic effects. The mechanism of action underlying these effects is not yet fully understood. This study investigated the downstream gene expression changes and signaling pathways affected by methylone in key brain areas linked to PTSD and MDD. It also sought to determine whether neuroplasticity-related genes were involved. We compared effects of methylone with MDMA to explore similarities and differences in their brain effects because MDMA-assisted psychotherapy has recently shown benefit in clinical trials for PTSD and methylone is a structural analog of MDMA. Methods: Monoamine binding, uptake and release studies were performed and a high-throughput-screen evaluated agonist/antagonist activities at 168 GPCRs in vitro. We used RNA sequencing (RNA-seq) to probe drug-induced gene expression changes in the amygdala and frontal cortex, two brain areas responsible for emotional learning that are affected by PTSD and MDD. Rats were treated with methylone or MDMA (both 10 mg/kg, IP), and their responses were compared with controls. We performed functional enrichment analysis to identify which pathways were regulated by methylone and/or MDMA. We confirmed changes in gene expression using immunohistochemistry. Results: Methylone, a monoamine uptake inhibitor and releaser, demonstrated no off-target effects at 168 GPCRs, unlike MDMA, which showed activity at 5HT2A and 5HT2C receptors. RNA-seq results revealed significant regulation of myelin-related genes in the amygdala, confirmed by immunohistochemistry. In the frontal cortex, methylone significantly upregulated genes implicated in neuroplasticity. Conclusion: Results suggest that (1) methylone is a rapid-acting neuroplastogen that affects key brain substrates for PTSD and MDD and that (2) methylone appears to exhibit higher specificity and fewer off-target effects than MDMA. Together, these results are consistent with the reported clinical experiences of methylone and MDMA and bolster the potential use of methylone in the treatment of PTSD and, potentially, other neuropsychiatric disorders.

9.
Nucleic Acids Res ; 38(13): 4218-30, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20308160

ABSTRACT

We have recently developed a novel method for the affinity purification of the complete suite of translating mRNA from genetically labeled cell populations. This method permits comprehensive quantitative comparisons of the genes employed by each specific cell type. We provide a detailed description of tools for analysis of data generated with this and related methodologies. An essential question that arises from these data is how to identify those genes that are enriched in each cell type relative to all others. Genes relatively specifically employed by a cell type may contribute to the unique functions of that cell, and thus may become useful targets for development of pharmacological tools for cell-specific manipulations. We describe here a novel statistic, the specificity index, which can be used for comparative quantitative analysis to identify genes enriched in specific cell populations across a large number of profiles. This measure correctly predicts in situ hybridization patterns for many cell types. We apply this measure to a large survey of CNS cell-specific microarray data to identify those genes that are significantly enriched in each population Data and algorithms are available online (www.bactrap.org).


Subject(s)
Gene Expression Profiling/methods , RNA, Messenger/analysis , Algorithms , Animals , Brain/cytology , Brain/metabolism , Data Interpretation, Statistical , Mice , Oligonucleotide Array Sequence Analysis , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism
10.
Cell Rep ; 38(12): 110556, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35320722

ABSTRACT

Many neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), lead to the selective degeneration of discrete cell types in the CNS despite the ubiquitous expression of many genes linked to disease. Therapeutic advancement depends on understanding the unique cellular adaptations that underlie pathology of vulnerable cells in the context of disease-causing mutations. Here, we employ bacTRAP molecular profiling to elucidate cell type-specific molecular responses of cortical upper motor neurons in a preclinical ALS model. Using two bacTRAP mouse lines that label distinct vulnerable or resilient projection neuron populations in motor cortex, we show that the regulation of oxidative phosphorylation (Oxphos) pathways is a common response in both cell types. However, differences in the baseline expression of genes involved in Stem and the handling of reactive oxygen species likely lead to the selective degeneration of the vulnerable cells. These results provide a framework to identify cell-type-specific processes in neurodegenerative disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Motor Cortex , Neurodegenerative Diseases , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Motor Cortex/metabolism , Motor Neurons/metabolism , Neurodegenerative Diseases/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
11.
Cereb Cortex ; 20(4): 826-36, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19643810

ABSTRACT

To understand sensory representation in cortex, it is crucial to identify its constituent cellular components based on cell-type-specific criteria. With the identification of cell types, an important question can be addressed: to what degree does the cellular properties of neurons depend on cortical location? We tested this question using pyramidal neurons in layer 5 (L5) because of their role in providing major cortical output to subcortical targets. Recently developed transgenic mice with cell-type-specific enhanced green fluorescent protein labeling of neuronal subtypes allow reliable identification of 2 cortical cell types in L5 throughout the entire neocortex. A comprehensive investigation of anatomical and functional properties of these 2 cell types in visual and somatosensory cortex demonstrates that, with important exceptions, most properties appear to be cell-type-specific rather than dependent on cortical area. This result suggests that although cortical output neurons share a basic layout throughout the sensory cortex, fine differences in properties are tuned to the cortical area in which neurons reside.


Subject(s)
Biophysical Phenomena/physiology , Neocortex/cytology , Pyramidal Cells/physiology , Somatosensory Cortex/physiology , Analysis of Variance , Animals , Cell Count/methods , Cholera Toxin/metabolism , Dendrites/physiology , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/genetics , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , In Vitro Techniques , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/physiology , Patch-Clamp Techniques/methods , Phosphopyruvate Hydratase/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Pyramidal Cells/cytology , Somatosensory Cortex/cytology , Thalamus/cytology , Thalamus/physiology
12.
Neuron ; 107(5): 821-835.e12, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32603655

ABSTRACT

A major obstacle to treating Alzheimer's disease (AD) is our lack of understanding of the molecular mechanisms underlying selective neuronal vulnerability, a key characteristic of the disease. Here, we present a framework integrating high-quality neuron-type-specific molecular profiles across the lifetime of the healthy mouse, which we generated using bacTRAP, with postmortem human functional genomics and quantitative genetics data. We demonstrate human-mouse conservation of cellular taxonomy at the molecular level for neurons vulnerable and resistant in AD, identify specific genes and pathways associated with AD neuropathology, and pinpoint a specific functional gene module underlying selective vulnerability, enriched in processes associated with axonal remodeling, and affected by amyloid accumulation and aging. We have made all cell-type-specific profiles and functional networks available at http://alz.princeton.edu. Overall, our study provides a molecular framework for understanding the complex interplay between Aß, aging, and neurodegeneration within the most vulnerable neurons in AD.


Subject(s)
Alzheimer Disease/pathology , Gene Expression Profiling/methods , Machine Learning , Neurons/pathology , Transcriptome , Aging/genetics , Aging/pathology , Alzheimer Disease/genetics , Animals , Gene Regulatory Networks/physiology , Humans , Mice
13.
J Neurosci ; 28(9): 2287-97, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18305261

ABSTRACT

Semaphorin activation of Plexin (Plex) receptors provides axonal guidance during neuronal development. Two families of cytoplasmic proteins, collapsin response mediator proteins (CRMPs) and molecules interacting with CasL (MICALs), have been implicated in Plexin function. The relationship between CRMP and MICAL signaling has not been defined nor is the mechanism by which Plexin activates MICAL clear. Here, we show that CRMP and MICAL physically associate and that Sema signaling promotes this association. MICAL enzymatic activity is inhibited by the C-terminal domain of MICAL. CRMP and Plexin associate with nonenzymatic and enzymatic domains of MICAL and together release MICAL enzymatic autoinhibition. In addition to acting as an upstream MICAL activator, CRMP functions downstream of MICAL, inhibiting the catalytic domain. A constitutively active CRMP mutant inhibits MICAL activity more potently than does wild-type CRMP, suggesting that CRMP or a CRMP-associated factor is a MICAL substrate. Thus, complex Plex/CRMP/MICAL interactions transduce Semaphorin signaling into axon guidance.


Subject(s)
Cell Adhesion Molecules/metabolism , Cytoskeletal Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Semaphorin-3A/metabolism , Semaphorins/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Line, Transformed , Chick Embryo , Cytoskeletal Proteins/genetics , Flavin-Adenine Dinucleotide/metabolism , Ganglia, Spinal/cytology , Genetic Vectors/physiology , HIV/physiology , Humans , Immunoprecipitation/methods , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins , Microfilament Proteins , Mixed Function Oxygenases , Mutation/physiology , Neurites/physiology , Neurons/cytology , Neurons/drug effects , Peptide Fragments/metabolism , Protein Binding , Transfection/methods
14.
Front Behav Neurosci ; 13: 157, 2019.
Article in English | MEDLINE | ID: mdl-31354448

ABSTRACT

Early life experiences program brain structure and function and contribute to behavioral endophenotypes in adulthood. Epigenetic control of gene expression by those experiences affect discrete brain regions involved in mood, cognitive function and regulation of hypothalamic-pituitary-adrenal (HPA) axis. In rodents, acute restraint stress increases the expression of the repressive histone H3 lysine 9 tri-methylation (H3K9me3) in hippocampal fields, including the CA3 pyramidal neurons. These CA3 neurons are crucially involved in cognitive function and mood regulation as well as activation of glucocorticoid (CORT) secretion. CA3 neurons also exhibit structural and functional changes after early-life stress (ELS) as well as after chronic stress in adulthood. Using a protocol of chronic ELS induced by limited bedding and nesting material followed by acute-swim stress (AS) in adulthood, we show that mice with a history of ELS display a blunted CORT response to AS, despite exhibiting activation of immediate early genes after stress similar to that found in control mice. We find that ELS induced persistently increased expression of the repressive H3K9me3 histone mark in the CA3 subfield at baseline that was subsequently decreased following AS. In contrast, AS induced a transient increase of this mark in control mice. Using translating ribosome affinity purification (TRAP) method to isolate CA3 translating mRNAs, we found that expression of genes of the epigenetic gene family, GABA/glutamate family, and glucocorticoid receptors binding genes were decreased transiently in control mice by AS and showed a persistent reduction in ELS mice. In most cases, AS in ELS mice did not induce gene expression changes. A stringent filtering of genes affected by AS in control and ELS mice revealed a noteworthy decrease in gene expression change in ELS mice compared to control. Only 18 genes were selectively regulated by AS in ELS mice and encompassed pathways such as circadian rhythm, inflammatory response, opioid receptors, and more genes included in the glucocorticoid receptor binding family. Thus, ELS programs a restricted translational response to stress in stress-sensitive CA3 neurons leading to persistent changes in gene expression, some of which mimic the transient effects of AS in control mice, while leaving in operation the immediate early gene response to AS.

15.
J Neurosci ; 26(18): 4961-9, 2006 May 03.
Article in English | MEDLINE | ID: mdl-16672672

ABSTRACT

Secreted Semaphorin3A (Sema3A) proteins are known to act as diffusible and repellant axonal guidance cues during nervous system development. A receptor complex consisting of a Neuropilin and a Plexin-A mediates their effects. Plexin-A signal transduction has remained poorly defined despite the documented involvement of collapsin response mediator protein and molecule interacting with CasL proteins (MICALs) as mediators of Plexin-A activation. Here, we defined a domain of Plexin-A1 required for Sema3A signaling in a reconstituted environment and then searched for proteins interacting with this domain. RanBPM is shown to physically interact with Plexin-A1, and the RanBPM/Plexin complex is regulated by MICAL expression. Overexpression of RanBPM cooperates with PlexinA1 to reduce non-neuronal cell spreading and strongly inhibit axonal outgrowth in vitro and in vivo. A truncated RanBPM protein blocks Sema3A responsiveness in non-neuronal and neuronal cells. Suppression of RanBPM expression reduces Sema3A responsiveness. Thus, RanBPM is a mediator of Sema3A signaling through Plexin-A. RanBPM has the potential to link Plexin-A receptors to retrograde transport and microtubule function in axonal guidance.


Subject(s)
Cell Adhesion Molecules/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Nuclear Proteins/physiology , Semaphorin-3A/metabolism , Signal Transduction/physiology , ran GTP-Binding Protein/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Size , Cells, Cultured , Chick Embryo , Cloning, Molecular/methods , Cricetinae , Cricetulus , Cytoskeletal Proteins , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Gene Expression/drug effects , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation/methods , In Situ Nick-End Labeling/methods , Neurites/physiology , Neurons/cytology , Neurons/drug effects , Neuropilin-1/metabolism , Semaphorin-3A/physiology , Signal Transduction/drug effects , Transcription Factor AP-1/pharmacology , Transfection/methods , Two-Hybrid System Techniques
16.
Adv Exp Med Biol ; 600: 1-11, 2007.
Article in English | MEDLINE | ID: mdl-17607942

ABSTRACT

The CRMP proteins were originally identified as mediators of Sema3A signaling and neuronal differentiation. Much has been learned about the mechanism by which CRMPs regulate cellular responses to Sema3A. In this review, the evidence for CRMP as a component of the Sema3A signaling cascade and the modulation of CRMP by plexin and phosphorylation are considered. In addition, current knowledge of the function of CRMP in a variety of cellular processes, including regulation of the cytoskeleton and endocytosis, is discussed in relationship to the mechanisms of axonal growth cone Sema3A response. The secreted protein Sema3A (collapsin-1) was the first identified vertebrate semaphorin. Sema3A acts primarily as a repulsive axon guidance cue, and can cause a dramatic collapse of the growth cone lamellipodium. This process results from the redistribution of the F-actin cytoskeleton and endocytosis of the growth cone cell membrane. Neuropilin-1 (NP1) and members of the class A plexins (PlexA) form a Sema3A receptor complex, with NP1 serving as a high-affinity ligand binding partner, and PlexA transducing the signal into the cell via its large intracellular domain. Although the effect of Sema3A on growth cones was first described nearly 15 years ago, the intracellular signaling pathways that lead to the cellular effects have only recently begun to be understood. Monomeric G-proteins, various kinases, the redox protein, MICAL, and protein turnover have all been implicated in PlexA transduction. In addition, the collapsin-response-mediator protein (CRMP) family of cytosolic phosphoproteins plays a crucial role in Sema3A/NP1/PlexA signal transduction. Current knowledge regarding CRMP functions are reviewed here.


Subject(s)
Nerve Tissue Proteins/physiology , Semaphorin-3A/physiology , Signal Transduction/physiology , Animals , Axons/chemistry , Axons/physiology , Growth Cones/chemistry , Growth Cones/physiology , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , Semaphorin-3A/chemistry , Semaphorin-3A/genetics , Signal Transduction/genetics
17.
Nat Commun ; 8(1): 808, 2017 10 09.
Article in English | MEDLINE | ID: mdl-28993643

ABSTRACT

Males and females use distinct brain circuits to cope with similar challenges. Using RNA sequencing of ribosome-bound mRNA from hippocampal CA3 neurons, we found remarkable sex differences and discovered that female mice displayed greater gene expression activation after acute stress than males. Stress-sensitive BDNF Val66Met mice of both sexes show a pre-stressed translational phenotype in which the same genes that are activated without applied stress are also induced in wild-type mice by an acute stressor. Behaviourally, only heterozygous BDNF Val66Met females exhibit spatial memory impairment, regardless of acute stress. Interestingly, this effect is not observed in ovariectomized heterozygous BDNF Val66Met females, suggesting that circulating ovarian hormones induce cognitive impairment in Met carriers. Cognitive deficits are not observed in males of either genotype. Thus, in a brain region not normally associated with sex differences, this work sheds light on ways that genes, environment and sex interact to affect the transcriptome's response to a stressor.Animals' response to acute stress is known to be influenced by sex and genetics. Here the authors performed RNA-seq on actively translated mRNAs in hippocampal CA3 neurons in mice, and document the effects of sex and genotype (i.e., BDNF Val66Met) on acute stress-induced gene expression.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Protein Biosynthesis , Pyramidal Cells/physiology , Stress, Physiological/genetics , Animals , Brain-Derived Neurotrophic Factor/metabolism , Female , Gene Expression Regulation , Glutamic Acid/genetics , Glutamic Acid/metabolism , Male , Mice, Transgenic , Ovariectomy , RNA, Messenger , Sequence Analysis, RNA , Sex Factors , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
18.
Cell Rep ; 19(3): 655-667, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423326

ABSTRACT

Translational profiling methodologies enable the systematic characterization of cell types in complex tissues, such as the mammalian brain, where neuronal isolation is exceptionally difficult. Here, we report a versatile strategy for profiling CNS cell types in a spatiotemporally restricted fashion by engineering a Cre-dependent adeno-associated virus expressing an EGFP-tagged ribosomal protein (AAV-FLEX-EGFPL10a) to access translating mRNAs by translating ribosome affinity purification (TRAP). We demonstrate the utility of this AAV to target a variety of genetically and anatomically defined neural populations expressing Cre recombinase and illustrate the ability of this viral TRAP (vTRAP) approach to recapitulate the molecular profiles obtained by bacTRAP in corticothalamic neurons across multiple serotypes. Furthermore, spatially restricting adeno-associated virus (AAV) injections enabled the elucidation of regional differences in gene expression within this cell type. Altogether, these results establish the broad applicability of the vTRAP strategy for the molecular dissection of any CNS or peripheral cell type that can be engineered to express Cre.


Subject(s)
Chromatography, Affinity/methods , Protein Biosynthesis , Ribosomes/metabolism , Viruses/metabolism , Animals , Biomarkers/metabolism , Dependovirus/metabolism , Female , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Male , Melanins/metabolism , Mice , Neurons/metabolism , Pituitary Hormones/metabolism , Reproducibility of Results , Serotyping
19.
eNeuro ; 3(5)2016.
Article in English | MEDLINE | ID: mdl-27844060

ABSTRACT

Serotonin (5-HT) regulates attention by neurobiological mechanisms that are not well understood. Layer 6 (L6) pyramidal neurons of prefrontal cortex play an important role in attention and express 5-HT receptors, but the serotonergic modulation of this layer and its excitatory output is not known. Here, we performed whole-cell recordings and pharmacological manipulations in acute brain slices from wild-type and transgenic mice expressing either eGFP or eGFP-channelrhodopsin in prefrontal L6 pyramidal neurons. Excitatory circuits between L6 pyramidal neurons and L5 GABAergic interneurons, including a population of interneurons essential for task attention, were investigated using optogenetic techniques. Our experiments show that prefrontal L6 pyramidal neurons are subject to strong serotonergic inhibition and demonstrate direct 5-HT-sensitive connections between prefrontal L6 pyramidal neurons and two classes of L5 interneurons. This work helps to build a neurobiological framework to appreciate serotonergic disruption of task attention and yields insight into the disruptions of attention observed in psychiatric disorders with altered 5-HT receptors and signaling.


Subject(s)
Attention/physiology , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Serotonin/physiology , Animals , Attention/drug effects , Female , Interneurons/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Pathways/drug effects , Neural Pathways/metabolism , Optogenetics , Patch-Clamp Techniques , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin Antagonists/pharmacology , Tissue Culture Techniques
20.
Cold Spring Harb Protoc ; 2013(3)2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23457350

ABSTRACT

The brain is a complex tissue comprising hundreds of distinct cell types, each of which has unique circuitry and plays a discrete role in nervous system function. Large-scale studies mapping gene-expression patterns throughout the nervous system have revealed that many genes are exclusively expressed in specific cell populations. The GENSAT (Gene Expression Nervous System Atlas) Project created a library of engineered mice utilizing bacterial artificial chromosomes (BACs) to drive the expression of enhanced green fluorescent protein (eGFP) in genetically defined cell populations. BACs contain large segments of genomic DNA and retain most of the transcriptional regulatory elements directing the expression of a given gene, resulting in more faithful reproduction of endogenous expression patterns. BAC transgenic mice offer a robust solution to the challenging task of stably and reproducibly accessing specific cell types from a heterogeneous tissue such as the brain. A significant advantage of utilizing eGFP as a reporter is the fact that it can fill entire cells, including neuronal dendrites and axons as well as glial processes, making GENSAT reporter mice a powerful tool for neuroimaging studies. This article provides a primer on the generation of BAC transgenic mice and advantages for their use in labeling genetically defined cell types. It also provides an overview of searching the GENSAT database and ordering engineered mouse lines.


Subject(s)
Databases, Genetic , Mice, Transgenic , Nervous System Physiological Phenomena , Animals , Chromosomes, Artificial, Bacterial , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Mice
SELECTION OF CITATIONS
SEARCH DETAIL