Network


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

Hotspot


Dive into the research topics where Marguerite Camp is active.

Publication


Featured researches published by Marguerite Camp.


The Journal of Neuroscience | 2010

Strain Differences in Stress Responsivity Are Associated with Divergent Amygdala Gene Expression and Glutamate-Mediated Neuronal Excitability

Khyobeni Mozhui; Rose-Marie Karlsson; Thomas L. Kash; Jessica Ihne; Maxine Norcross; Sachin Patel; Mollee R. Farrell; Elizabeth E. Hill; Carolyn Graybeal; Kathryn P. Martin; Marguerite Camp; Paul J. Fitzgerald; Daniel C. Ciobanu; Rolf Sprengel; Masayoshi Mishina; Cara L. Wellman; Danny G. Winder; Robert W. Williams; Andrew Holmes

Stress is a major risk factor for numerous neuropsychiatric diseases. However, susceptibility to stress and the qualitative nature of stress effects on behavior differ markedly among individuals. This is partly because of the moderating influence of genetic factors. Inbred mouse strains provide a relatively stable and restricted range of genetic and environmental variability that is valuable for disentangling gene–stress interactions. Here, we screened a panel of inbred strains for anxiety- and depression-related phenotypes at baseline (trait) and after exposure to repeated restraint. Two strains, DBA/2J and C57BL/6J, differed in trait and restraint-induced anxiety-related behavior (dark/light exploration, elevated plus maze). Gene expression analysis of amygdala, medial prefrontal cortex, and hippocampus revealed divergent expression in DBA/2J and C57BL/6J both at baseline and after repeated restraint. Restraint produced strain-dependent expression alterations in various genes including glutamate receptors (e.g., Grin1, Grik1). To elucidate neuronal correlates of these strain differences, we performed ex vivo analysis of glutamate excitatory neurotransmission in amygdala principal neurons. Repeated restraint augmented amygdala excitatory postsynaptic signaling and altered metaplasticity (temporal summation of NMDA receptor currents) in DBA/2J but not C57BL/6J. Furthermore, we found that the C57BL/6J-like changes in anxiety-related behavior after restraint were absent in null mutants lacking the modulatory NMDA receptor subunit Grin2a, but not the AMPA receptor subunit Gria1. Grin2a null mutants exhibited significant (∼30%) loss of dendritic spines on amygdala principal neurons under nonrestraint conditions. Collectively, our data support a model in which genetic variation in glutamatergic neuroplasticity in corticolimbic circuitry underlies phenotypic variation in responsivity to stress.


Nature Neuroscience | 2012

Chronic alcohol remodels prefrontal neurons and disrupts NMDAR-mediated fear extinction encoding

Andrew Holmes; Paul J. Fitzgerald; Kathryn P. MacPherson; Lauren DeBrouse; Giovanni Colacicco; Shaun M. Flynn; Sophie Masneuf; Kristen E. Pleil; Chia Li; Catherine A. Marcinkiewcz; Thomas L. Kash; Ozge Gunduz-Cinar; Marguerite Camp

Alcoholism is frequently co-morbid with post-traumatic stress disorder, but it is unclear how alcohol affects the neural circuits mediating recovery from trauma. We found that chronic intermittent ethanol (CIE) impaired fear extinction and remodeled the dendritic arbor of medial prefrontal cortical (mPFC) neurons in mice. CIE impaired extinction encoding by infralimbic mPFC neurons in vivo and functionally downregulated burst-mediating NMDA GluN1 receptors. These findings suggest that alcohol may increase risk for trauma-related anxiety disorders by disrupting mPFC-mediated extinction of fear.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Chronic alcohol produces neuroadaptations to prime dorsal striatal learning

Lauren DePoy; Rachel A. Daut; Jonathan L. Brigman; Kathryn P. MacPherson; Nicole A. Crowley; Ozge Gunduz-Cinar; Charles L. Pickens; Resat Cinar; Lisa M. Saksida; George Kunos; David M. Lovinger; Timothy J. Bussey; Marguerite Camp; Andrew Holmes

Significance Alcoholism is characterized by a progressive degradation of executive control and an increase in compulsive alcohol seeking that is hypothesized to involve a shift from prefrontal cortex to dorsal striatal (DLS) control over behavior. Here, we show that mice exposed to chronic intermittent alcohol exhibited expansion of dendritic material in DLS neurons, coupled with loss of endocannabinoid CB1 receptor signaling and CB1-mediated long-term depression in the DLS. Behaviorally, chronic alcohol exposure facilitated various forms of DLS-dependent learning and augmented in vivo DLS neuronal activity as correct learned choices were made. These findings support a model in which chronic ethanol causes DLS neuroadaptations that prime for greater striatal control over behavior, potentially contributing to the progression of alcoholism. Drug addictions including alcoholism are characterized by degradation of executive control over behavior and increased compulsive drug seeking. These profound behavioral changes are hypothesized to involve a shift in the regulation of behavior from prefrontal cortex to dorsal striatum (DLS). Studies in rodents have shown that ethanol disrupts cognitive processes mediated by the prefrontal cortex, but the potential effects of chronic ethanol on DLS-mediated cognition and learning are much less well understood. Here, we first examined the effects of chronic EtOH on DLS neuronal morphology, synaptic plasticity, and endocannabinoid-CB1R signaling. We next tested for ethanol-induced changes in striatal-related learning and DLS in vivo single-unit activity during learning. Mice exposed to chronic intermittent ethanol (CIE) vapor exhibited expansion of dendritic material in DLS neurons. Following CIE, DLS endocannabinoid CB1 receptor signaling was down-regulated, and CB1 receptor-dependent long-term depression at DLS synapses was absent. CIE mice showed facilitation of DLS-dependent pairwise visual discrimination and reversal learning, relative to air-exposed controls. CIE mice were also quicker to extinguish a stimulus–reward instrumental response and faster to reduce Pavlovian approach behavior under an omission schedule. In vivo single-unit recording during learning revealed that CIE mice had augmented DLS neuronal activity during correct responses. Collectively, these findings support a model in which chronic ethanol causes neuroadaptations in the DLS that prime for greater DLS control over learning. The shift to striatal dominance over behavior may be a critical step in the progression of alcoholism.


Neuropsychopharmacology | 2012

Genetic Strain Differences in Learned Fear Inhibition Associated with Variation in Neuroendocrine, Autonomic, and Amygdala Dendritic Phenotypes

Marguerite Camp; Kathryn P. MacPherson; Lauren Lederle; Carolyn Graybeal; Stefano Gaburro; Lauren DeBrouse; Jessica Ihne; Javier A. Bravo; Richard M. O'Connor; Stephane Ciocchi; Cara L. Wellman; Andreas Lüthi; John F. Cryan; Nicolas Singewald; Andrew B. Holmes

Mood and anxiety disorders develop in some but not all individuals following exposure to stress and psychological trauma. However, the factors underlying individual differences in risk and resilience for these disorders, including genetic variation, remain to be determined. Isogenic inbred mouse strains provide a valuable approach to elucidating these factors. Here, we performed a comprehensive examination of the extinction-impaired 129S1/SvImJ (S1) inbred mouse strain for multiple behavioral, autonomic, neuroendocrine, and corticolimbic neuronal morphology phenotypes. We found that S1 exhibited fear overgeneralization to ambiguous contexts and cues, impaired context extinction and impaired safety learning, relative to the (good-extinguishing) C57BL/6J (B6) strain. Fear overgeneralization and impaired extinction was rescued by treatment with the front-line anxiety medication fluoxetine. Telemetric measurement of electrocardiogram signals demonstrated autonomic disturbances in S1 including poor recovery of fear-induced suppression of heart rate variability. S1 with a history of chronic restraint stress displayed an attenuated corticosterone (CORT) response to a novel, swim stressor. Conversely, previously stress-naive S1 showed exaggerated CORT responses to acute restraint stress or extinction training, insensitivity to dexamethasone challenge, and reduced hippocampal CA3 glucocorticoid receptor mRNA, suggesting downregulation of negative feedback control of the hypothalamic–pituitary–adrenal axis. Analysis of neuronal morphology in key neural nodes within the fear and extinction circuit revealed enlarged dendritic arbors in basolateral amygdala neurons in S1, but normal infralimbic cortex and prelimbic cortex dendritic arborization. Collectively, these data provide convergent support for the utility of the S1 strain as a tractable model for elucidating the neural, molecular and genetic basis of persistent, excessive fear.


Genes, Brain and Behavior | 2009

Impaired Pavlovian fear extinction is a common phenotype across genetic lineages of the 129 inbred mouse strain.

Marguerite Camp; Maxine Norcross; Nigel Whittle; Michael Feyder; Wolfgang D’Hanis; Deniz Yilmazer-Hanke; Nicolas Singewald; Andrew B. Holmes

Fear extinction is impaired in psychiatric disorders such as post‐traumatic stress disorder and schizophrenia, which have a major genetic component. However, the genetic factors underlying individual variability in fear extinction remain to be determined. By comparing a panel of inbred mouse strains, we recently identified a strain, 129S1/SvImJ (129S1), that exhibits a profound and selective deficit in Pavlovian fear extinction, and associated abnormalities in functional activation of a key prefrontal‐amygdala circuit, as compared with C57BL/6J. The first aim of the present study was to assess fear extinction across multiple 129 substrains representing the strains four different genetic lineages (parental, steel, teratoma and contaminated). Results showed that 129P1/ReJ, 129P3/J, 129T2/SvEmsJ and 129X1/SvJ exhibited poor fear extinction, relative to C57BL/6J, while 129S1 showed evidence of fear incubation. On the basis of these results, the second aim was to further characterize the nature and specificity of the extinction phenotype in 129S1, as an exemplar of the 129 substrains. Results showed that the extinction deficit in 129S1 was neither the result of a failure to habituate to a sensitized fear response nor an artifact of a fear response to (unconditioned) tone per se. A stronger conditioning protocol (i.e. five × higher intensity shocks) produced an increase in fear expression in 129S1, relative to C57BL/6J, due to rapid rise in freezing during tone presentation. Taken together, these data show that impaired fear extinction is a phenotypic feature common across 129 substrains, and provide preliminary evidence that impaired fear extinction in 129S1 may reflect a pro‐fear incubation‐like process.


Molecular Psychiatry | 2015

Durable fear memories require PSD-95.

Paul J. Fitzgerald; Courtney R. Pinard; Marguerite Camp; Michael Feyder; Anupam Sah; Hadley C. Bergstrom; Carolyn Graybeal; Yan Liu; Oliver M. Schlüter; Seth G. N. Grant; Nicolas Singewald; Weifeng Xu; Andrew B. Holmes

Traumatic fear memories are highly durable but also dynamic, undergoing repeated reactivation and rehearsal over time. Although overly persistent fear memories underlie anxiety disorders, such as posttraumatic stress disorder, the key neural and molecular mechanisms underlying fear memory durability remain unclear. Postsynaptic density 95 (PSD-95) is a synaptic protein regulating glutamate receptor anchoring, synaptic stability and certain types of memory. Using a loss-of-function mutant mouse lacking the guanylate kinase domain of PSD-95 (PSD-95GK), we analyzed the contribution of PSD-95 to fear memory formation and retrieval, and sought to identify the neural basis of PSD-95-mediated memory maintenance using ex vivo immediate-early gene mapping, in vivo neuronal recordings and viral-mediated knockdown (KD) approaches. We show that PSD-95 is dispensable for the formation and expression of recent fear memories, but essential for the formation of precise and flexible fear memories and for the maintenance of memories at remote time points. The failure of PSD-95GK mice to retrieve remote cued fear memory was associated with hypoactivation of the infralimbic (IL) cortex (but not the anterior cingulate cortex (ACC) or prelimbic cortex), reduced IL single-unit firing and bursting, and attenuated IL gamma and theta oscillations. Adeno-associated virus-mediated PSD-95 KD in the IL, but not the ACC, was sufficient to impair recent fear extinction and remote fear memory, and remodel IL dendritic spines. Collectively, these data identify PSD-95 in the IL as a critical mechanism supporting the durability of fear memories over time. These preclinical findings have implications for developing novel approaches to treating trauma-based anxiety disorders that target the weakening of overly persistent fear memories.


Addiction Biology | 2011

A novel role for PSD-95 in mediating ethanol intoxication, drinking and place preference.

Marguerite Camp; Michael Feyder; Jessica Ihne; Benjamin Palachick; Benita Hurd; Rose-Marie Karlsson; Bianca Noronha; Yi-Chyan Chen; Marcelo P. Coba; Seth G. N. Grant; Andrew Holmes

The synaptic signaling mechanisms mediating the behavioral effects of ethanol (EtOH) remain poorly understood. Post‐synaptic density 95 (PSD‐95, SAP‐90, Dlg4) is a key orchestrator of N‐methyl‐D‐aspartate receptors (NMDAR) and glutamatergic synapses, which are known to be major sites of EtOHs behavioral actions. However, the potential contribution of PSD‐95 to EtOH‐related behaviors has not been established. Here, we evaluated knockout (KO) mice lacking PSD‐95 for multiple measures of sensitivity to the acute intoxicating effects of EtOH (ataxia, hypothermia, sedation/hypnosis), EtOH drinking under conditions of free access and following deprivation, acquisition and long‐term retention of EtOH conditioned place preference (CPP) (and lithium chloride‐induced conditioned taste aversion), and intoxication‐potentiating responses to NMDAR antagonism. PSD‐95 KO exhibited increased sensitivity to the sedative/hypnotic, but not ataxic or hypothermic, effects of acute EtOH relative to wild‐type controls (WT). PSD‐95 KO consumed less EtOH than WT, particularly at higher EtOH concentrations, although increases in KO drinking could be induced by concentration‐fading and deprivation. PSD‐95 KO showed normal EtOH CPP 1 day after conditioning, but showed significant aversion 2 weeks later. Lithium chloride‐induced taste aversion was impaired in PSD‐95 KO at both time points. Finally, the EtOH‐potentiating effects of the NMDAR antagonist MK‐801 were intact in PSD‐95 KO at the dose tested. These data reveal a major, novel role for PSD‐95 in mediating EtOH behaviors, and add to growing evidence that PSD‐95 is a key mediator of the effects of multiple abused drugs.


Addiction Biology | 2015

Chronic alcohol alters rewarded behaviors and striatal plasticity.

Lauren DePoy; Rachel A. Daut; Tara Wright; Marguerite Camp; Nicole A. Crowley; Bianca Noronha; David M. Lovinger; Andrew B. Holmes

Chronic intermittent ethanol (CIE) alters neural functions and behaviors mediated by the dorsolateral striatum (DLS) and prefrontal cortex. Here, we examined the effects of prolonged (16‐bout) CIE on DLS plasticity and DLS‐mediated behaviors. Ex vivo electrophysiological recordings revealed loss in efficacy of DLS synaptically induced activation and absent long‐term depression after CIE. CIE increased two‐bottle choice drinking and impaired Pavlovian‐to‐instrumental transfer but not discriminated approach. These data suggest prolonged CIE impaired DLS plasticity, to produce associated changes in drinking and cue‐controlled reward‐seeking. Given recent evidence that less‐prolonged CIE can promote certain dorsal striatal‐mediated behaviors, CIE may drive chronicity‐dependent adaptations in corticostriatal systems regulating behavior.


Biology of Mood & Anxiety Disorders | 2013

Temporal factors in the extinction of fear in inbred mouse strains differing in extinction efficacy

Kathryn P. MacPherson; Nigel Whittle; Marguerite Camp; Ozge Gunduz-Cinar; Nicolas Singewald; Andrew Holmes

BackgroundVarious neuropsychiatric conditions, including posttraumatic stress disorder (PTSD), are characterized by deficient fear extinction, but individuals differ greatly in risk for these. While there is growing evidence that fear extinction is influenced by certain procedural variables, it is unclear how these influences might vary across individuals and subpopulations. To model individual differences in fear extinction, prior studies identified a strain of inbred mouse, 129S1/SvImJ (S1), which exhibits a profound deficit in fear extinction, as compared to other inbred strains, such as C57BL/6J (B6).MethodsHere, we assessed the effects of procedural variables on the impaired extinction phenotype of the S1 strain and, by comparison, the extinction-intact B6 strain. The variables studied were 1) the interval between conditioning and extinction, 2) the interval between cues during extinction training, 3) single-cue exposure before extinction training, and 4) extinction of a second-order conditioned cue.ResultsConducting extinction training soon after (‘immediately’) conditioning attenuated fear retrieval in S1 mice and impaired extinction in B6 mice. Spacing cue presentations with long inter-trial intervals during extinction training augmented fear in S1 and B6 mice. The effect of spacing was lost with one-trial fear conditioning in B6, but not S1 mice. A single exposure to a conditioned cue before extinction training did not alter extinction retrieval, either in B6 or S1 mice. Both the S1 and B6 strains exhibited robust second-order fear conditioning, in which a cue associated with footshock was sufficient to serve as a conditioned exciter to condition a fear association to a second cue. B6 mice extinguished the fear response to the second-order conditioned cue, but S1 mice failed to do so.ConclusionsThese data provide further evidence that fear extinction is strongly influenced by multiple procedural variables and is so in a highly strain-dependent manner. This suggests that the efficacy of extinction-based behavioral interventions, such as exposure therapy, for trauma-related anxiety disorders will be determined by the procedural parameters employed and the degree to which the patient can extinguish.


Experimental Neurology | 2013

Convergent effects of mouse Pet-1 deletion and human PET-1 variation on amygdala fear and threat processing

Cara L. Wellman; Marguerite Camp; V. Morgan Jones; Kathryn P. MacPherson; Jessica Ihne; Paul J. Fitzgerald; Mouna Maroun; Emily M. Drabant; Ryan Bogdan; Ahmad R. Hariri; Andrew B. Holmes

Serotonin is critical for shaping the development of neural circuits regulating emotion. Pet-1 (FEV-1) is an ETS-domain transcription factor essential for differentiation and forebrain targeting of serotonin neurons. Constitutive Pet-1 knockout (KO) causes major loss of serotonin neurons and forebrain serotonin availability, and behavioral abnormalities. We phenotyped Pet-1 KO mice for fear conditioning and extinction, and on a battery of assays for anxiety- and depression-related behaviors. Morphology of Golgi-stained neurons in basolateral amygdala (BLA) and prelimbic cortex was examined. Using human imaging genetics, a common variant (rs860573) in the PET-1 (FEV) gene was tested for effects on threat-related amygdala reactivity and psychopathology in 88 Asian-ancestry subjects. Pet-1 KO mice exhibited increased acquisition and expression of fear, and elevated fear recovery following extinction, relative to wild-type (WT). BLA dendrites of Pet-1 KO mice were significantly longer than in WT. Human PET-1 variation associated with differences in amygdala threat processing and psychopathology. This novel evidence for the role of Pet-1 in fear processing and dendritic organization of amygdala neurons and in human amygdala threat processing extends a growing literature demonstrating the influence of genetic variation in the serotonin system on emotional regulation via effects on structure and function of underlying corticolimbic circuitry.

Collaboration


Dive into the Marguerite Camp's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jessica Ihne

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael Feyder

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Paul J. Fitzgerald

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Holmes

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Carolyn Graybeal

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kathryn P. MacPherson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Cara L. Wellman

Indiana University Bloomington

View shared research outputs
Researchain Logo
Decentralizing Knowledge