Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798398

ABSTRACT

Astrocytes form an integral component of the neurovascular unit, ensheathing brain blood vessels with projections high in aquaporin-4 (AQP4) expression. These AQP4-rich projections facilitate interaction between the vascular endothelium, astrocytes, and neurons, and help stabilize vascular morphology. Studies using preclinical models of psychological stress and post-mortem tissue from patients with major depressive disorder (MDD) have reported reductions in AQP4, loss of astrocytic structures, and vascular impairment in the prefrontal cortex (PFC). Though compelling, the role of AQP4 in mediating stress-induced alterations in blood vessel function and behavior remains unclear. Here, we address this, alongside potential sex differences in chronic unpredictable stress (CUS) effects on astrocyte phenotype, blood-brain barrier integrity, and behavior. CUS led to pronounced shifts in stress-coping behavior and working memory deficits in male -but not female- mice. Following behavioral testing, astrocytes from the frontal cortex were isolated for gene expression analyses. We found that CUS increased various transcripts associated with blood vessel maintenance in astrocytes from males, but either had no effect on- or decreased- these genes in females. Furthermore, CUS caused a reduction in vascular-localized AQP4 and elevated extravasation of a small molecule fluorescent reporter (Dextran) in the PFC in males but not females. Studies showed that knockdown of AQP4 in the PFC in males is sufficient to disrupt astrocyte phenotype and increase behavioral susceptibility to a sub-chronic stressor. Collectively, these findings provide initial evidence that sex-specific alterations in astrocyte phenotype and neurovascular integrity in the PFC contribute to behavioral and cognitive consequences following chronic stress.

2.
Brain Behav Immun ; 109: 127-138, 2023 03.
Article in English | MEDLINE | ID: mdl-36681359

ABSTRACT

In the medial prefrontal cortex (PFC), chronic stress reduces synaptic expression of glutamate receptors, leading to decreased excitatory signaling from layer V pyramidal neurons and working memory deficits. One key element driving these changes is a reduction in brain-derived neurotrophic factor (BDNF) signaling. BDNF is a potent mediator of synaptic growth and deficient BDNF signaling has been linked to stress susceptibility. Prior studies indicated that neurons are the primary source of BDNF, but more recent work suggests that microglia are also an important source of BDNF. Adding to this, our work showed that 14 days of chronic unpredictable stress (CUS) reduced Bdnf transcript in PFC microglia, evincing its relevance in the effects of stress. To explore this further, we utilized transgenic mice with microglia-specific depletion of BDNF (Cx3cr1Cre/+:Bdnffl/fl) and genotype controls (Cx3cr1Cre/+:Bdnf+/+). In the following experiments, mice were exposed to a shortened CUS paradigm (7 days) to determine if microglial Bdnf depletion promotes stress susceptibility. Analyses of PFC microglia revealed that Cx3cr1Cre/+:Bdnffl/fl mice had shifts in phenotypic markers and gene expression. In a separate cohort, synaptoneurosomes were collected from the PFC and western blotting was performed for synaptic markers. These experiments showed that Cx3cr1Cre/+:Bdnffl/fl mice had baseline deficits in GluN2B, and that 7 days of CUS additionally reduced GluN2A levels in Cx3cr1Cre/+:Bdnffl/fl mice, but not genotype controls. Behavioral and cognitive testing showed that this coincided with exacerbated stress effects on temporal object recognition in Cx3cr1Cre/+:Bdnffl/fl mice. These results indicate that microglial BDNF promotes glutamate receptor expression in the PFC. As such, mice with deficient microglial BDNF had increased susceptibility to the behavioral and cognitive consequences of stress.


Subject(s)
Brain-Derived Neurotrophic Factor , Microglia , Animals , Mice , Brain-Derived Neurotrophic Factor/metabolism , Mice, Transgenic , Microglia/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Humans
3.
J Immunol ; 208(6): 1341-1351, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35181638

ABSTRACT

Type III IFNs (IFNLs) are newly discovered cytokines, acting at epithelial and other barriers, that exert immunomodulatory functions in addition to their primary roles in antiviral defense. In this study, we define a role for IFNLs in maintaining autoreactive T cell effector function and limiting recovery in a murine model of multiple sclerosis (MS), experimental autoimmune encephalomyelitis. Genetic or Ab-based neutralization of the IFNL receptor (IFNLR) resulted in lack of disease maintenance during experimental autoimmune encephalomyelitis, with loss of CNS Th1 effector responses and limited axonal injury. Phenotypic effects of IFNLR signaling were traced to increased APC function, with associated increase in T cell production of IFN-γ and GM-CSF. Consistent with this, IFNL levels within lesions of CNS tissues derived from patients with MS were elevated compared with MS normal-appearing white matter. Furthermore, expression of IFNLR was selectively elevated in MS active lesions compared with inactive lesions or normal-appearing white matter. These findings suggest IFNL signaling as a potential therapeutic target to prevent chronic autoimmune neuroinflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Autoimmunity , Central Nervous System , Cytokines/metabolism , Humans , Mice
5.
Brain Behav Immun ; 99: 383-396, 2022 01.
Article in English | MEDLINE | ID: mdl-34695572

ABSTRACT

Innate immune responses to emerging RNA viruses are increasingly recognized as having significant contributions to neurologic sequelae, especially memory disorders. Using a recovery model of West Nile virus (WNV) encephalitis, we show that, while macrophages deliver the antiviral and anti-neurogenic cytokine IL-1ß during acute infection; viral recovery is associated with continued astrocyte inflammasome-mediated production of inflammatory levels of IL-1ß, which is maintained by hippocampal astrogenesis via IL-1R1 signaling in neural stem cells (NSC). Accordingly, aberrant astrogenesis is prevented in the absence of IL-1 signaling in NSC, indicating that only newly generated astrocytes exert neurotoxic effects, preventing synapse repair and promoting spatial learning deficits. Ex vivo evaluation of IL-1ß-treated adult hippocampal NSC revealed the upregulation of developmental differentiation pathways that derail adult neurogenesis in favor of astrogenesis, following viral infection. We conclude that NSC-specific IL-1 signaling within the hippocampus during viral encephalitis prevents synapse recovery and promotes spatial learning defects via altered fates of NSC progeny that maintain inflammation.


Subject(s)
Encephalitis, Viral , Neural Stem Cells , West Nile Fever , Humans , Inflammasomes/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , West Nile Fever/metabolism
6.
Glia ; 68(7): 1361-1374, 2020 07.
Article in English | MEDLINE | ID: mdl-31961459

ABSTRACT

During multiple sclerosis (MS), an inflammatory and neurodegenerative disease of the central nervous system (CNS), symptoms, and outcomes are determined by the location of inflammatory lesions. While we and others have shown that T cell cytokines differentially regulate leukocyte entry into perivascular spaces and regional parenchymal localization in murine models of MS, the molecular mechanisms of this latter process are poorly understood. Here, we demonstrate that astrocytes exhibit region-specific responses to T cell cytokines that promote hindbrain versus spinal cord neuroinflammation. Analysis of cytokine receptor expression in human astrocytes showed region-specific responsiveness to Th1 and Th17 inflammatory cytokines. Consistent with this, human and murine astrocytes treated with these cytokines exhibit differential expression of the T cell localizing molecules VCAM-1 and CXCR7 that is both cytokine and CNS region-specific. Using in vivo models of spinal cord versus brain stem trafficking of myelin-specific T cells and astrocyte-specific deletion strategies, we confirmed that Th1 and Th17 cytokines differentially regulate astrocyte expression of VCAM-1 and CXCR7 in these locations. Finally, stereotaxic injection of individual cytokines into the hindbrain or spinal cord revealed region- and cytokine-specific modulation of localizing cue expression by astrocytes. These findings identify a role for inflammatory cytokines in mediating local astrocyte-dependent mechanisms of immune cell trafficking within the CNS during neuroinflammation.


Subject(s)
Astrocytes/metabolism , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Neurodegenerative Diseases/pathology , Animals , Cell Movement/physiology , Central Nervous System/pathology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Mice, Transgenic , Multiple Sclerosis/pathology , Neurodegenerative Diseases/metabolism , Spinal Cord/pathology
7.
Eval Rev ; 44(4): 295-324, 2020 08.
Article in English | MEDLINE | ID: mdl-30537865

ABSTRACT

BACKGROUND: Policy makers seek to replace the "thumbs up-thumbs down" of conventional hypothesis testing with statements about the probability that program effects on key outcomes exceed policy-relevant thresholds. OBJECTIVE: We develop a Bayesian model that addresses the shortcomings of a typical frequentist approach to estimating the effects of the Comprehensive Primary Care (CPC) initiative, a Centers for Medicare and Medicaid Services demonstration. We compare findings from the two approaches to illustrate the relative merits of introducing additional assumptions through Bayesian methods. RESEARCH DESIGN: We apply Bayesian and frequentist methods to estimate the effects of CPC on total Medicare expenditures per beneficiary per month for Medicare beneficiaries attributed to participating practices. Under both paradigms, we estimated program effects using difference-in-differences regressions comparing the change in Medicare expenditures between baseline and follow-up for Medicare patients attributed to 497 primary care practices participating in CPC to Medicare patients attributed to 908 propensity score-matched comparison practices. RESULTS: Results from the Bayesian and frequentist models are comparable for the overall sample, but in regional subsamples, the Bayesian model produces more precise etimates that exhibit less variation over time. The Bayesian results also permit probabilistic inference about the magnitudes of effects, offering policy makers the ability to draw conclusions about practically meaningful thresholds. CONCLUSIONS: Carefully developed Bayesian models can enhance precision and plausibility and offer a more nuanced understanding of where and when program effects occur, without imposing undue assumptions. At the same time, these methods frame conclusions in flexible, intuitive terms that respond directly to policy makers' needs.


Subject(s)
Bayes Theorem , Program Evaluation/statistics & numerical data , Cost-Benefit Analysis/statistics & numerical data , Medicare , Primary Health Care , United States
8.
Physiol Behav ; 209: 112598, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31271833

ABSTRACT

Converging evidence supports neuroimmune factors in depression psychopathology. We previously reported reduced depression-like behavior in immunomodulatory G-protein-coupled receptor, T cell death-associated gene-8 (TDAG8) deficient mice. Here, we expand on those findings by investigating depression- and anxiety-associated behaviors, and cytokine profiles in TDAG8-deficient mice. TDAG8-deficiency reduced depression- and anxiety-associated behaviors in the forced swim test (FST), open-field test and elevated zero maze. Interestingly, cytokine expression, particularly IL-6, was attenuated within hippocampus and spleen in TDAG8-deficient mice following the FST. There were no differences in immune-cell frequencies. Collectively, these data suggest a contributory role of TDAG8 in neuroimmune regulation and depression-associated physiology.


Subject(s)
Behavior, Animal , Depression/psychology , T-Lymphocytes/physiology , Animals , Anxiety/psychology , Cell Death , Cytokines/biosynthesis , Hippocampus/metabolism , Interleukin-6/biosynthesis , Interleukin-6/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Spleen/metabolism , Swimming/psychology , T-Lymphocytes/immunology
9.
Nat Neurosci ; 22(8): 1276-1288, 2019 08.
Article in English | MEDLINE | ID: mdl-31235930

ABSTRACT

T cells clear virus from the CNS and dynamically regulate brain functions, including spatial learning, through cytokine signaling. Here we determined whether hippocampal T cells that persist after recovery from infection with West Nile virus (WNV) or Zika virus (ZIKV) impact hippocampal-dependent learning and memory. Using newly established models of viral encephalitis recovery in adult animals, we show that in mice that have recovered from WNV or ZIKV infection, T cell-derived interferon-γ (IFN-γ) signaling in microglia underlies spatial-learning defects via virus-target-specific mechanisms. Following recovery from WNV infection, mice showed presynaptic termini elimination with lack of repair, while for ZIKV, mice showed extensive neuronal apoptosis with loss of postsynaptic termini. Accordingly, animals deficient in CD8+ T cells or IFN-γ signaling in microglia demonstrated protection against synapse elimination following WNV infection and decreased neuronal apoptosis with synapse recovery following ZIKV infection. Thus, T cell signaling to microglia drives post-infectious cognitive sequelae that are associated with emerging neurotropic flaviviruses.


Subject(s)
Cognition Disorders/psychology , Flavivirus Infections/immunology , Flavivirus Infections/psychology , Microglia/immunology , Synapses/immunology , Synapses/pathology , T-Lymphocytes/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/immunology , Cognition Disorders/etiology , Female , Flavivirus Infections/pathology , Interferon-gamma , Learning Disabilities/etiology , Learning Disabilities/psychology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interferon/genetics , West Nile Fever/immunology , West Nile Fever/psychology , Zika Virus Infection/immunology , Zika Virus Infection/psychology , Interferon gamma Receptor
10.
J Clin Invest ; 128(11): 4980-4991, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30247157

ABSTRACT

The E3 ubiquitin ligase Pellino 1 (Peli1) is a microglia-specific mediator of autoimmune encephalomyelitis. Its role in neurotropic flavivirus infection is largely unknown. Here, we report that mice deficient in Peli1 (Peli1-/-) were more resistant to lethal West Nile virus (WNV) infection and exhibited reduced viral loads in tissues and attenuated brain inflammation. Peli1 mediates chemokine and proinflammatory cytokine production in microglia and promotes T cell and macrophage infiltration into the CNS. Unexpectedly, Peli1 was required for WNV entry and replication in mouse macrophages and mouse and human neurons and microglia. It was also highly expressed on WNV-infected neurons and adjacent inflammatory cells from postmortem patients who died of acute WNV encephalitis. WNV passaged in Peli1-/- macrophages or neurons induced a lower viral load and impaired activation in WT microglia and thereby reduced lethality in mice. Smaducin-6, which blocks interactions between Peli1 and IRAK1, RIP1, and IKKε, did not inhibit WNV-triggered microglia activation. Collectively, our findings suggest a nonimmune regulatory role for Peli1 in promoting microglia activation during WNV infection and identify a potentially novel host factor for flavivirus cell entry and replication.


Subject(s)
Neurons/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication/physiology , West Nile Fever/metabolism , West Nile virus/physiology , Animals , Chemokines/genetics , Chemokines/metabolism , Chlorocebus aethiops , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Macrophages/metabolism , Macrophages/pathology , Macrophages/virology , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Microglia/virology , Neurons/pathology , Neurons/virology , Nuclear Proteins/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Ubiquitin-Protein Ligases/genetics , Vero Cells , Viral Load , West Nile Fever/genetics , West Nile Fever/pathology
11.
Nat Immunol ; 19(2): 151-161, 2018 02.
Article in English | MEDLINE | ID: mdl-29292385

ABSTRACT

Memory impairment following West Nile virus neuroinvasive disease (WNND) is associated with loss of hippocampal synapses with lack of recovery. Adult neurogenesis and synaptogenesis are fundamental features of hippocampal repair, which suggests that viruses affect these processes. Here, in an established model of WNND-induced cognitive dysfunction, transcriptional profiling revealed alterations in the expression of genes encoding molecules that limit adult neurogenesis, including interleukin 1 (IL-1). Mice that had recovered from WNND exhibited fewer neuroblasts and increased astrogenesis without recovery of hippocampal neurogenesis at 30 d. Analysis of cytokine production in microglia and astrocytes isolated ex vivo revealed that the latter were the predominant source of IL-1. Mice deficient in the IL-1 receptor IL-1R1 and that had recovered from WNND exhibited normal neurogenesis, recovery of presynaptic termini and resistance to spatial learning defects, the last of which likewise occurred after treatment with an IL-1R1 antagonist. Thus, 'preferential' generation of proinflammatory astrocytes impaired the homeostasis of neuronal progenitor cells via expression of IL-1; this might underlie the long-term cognitive consequences of WNND but also provides a therapeutic target.


Subject(s)
Astrocytes/metabolism , Interleukin-1/biosynthesis , Neurogenesis/physiology , West Nile Fever/complications , Adult Stem Cells/metabolism , Animals , Astrocytes/immunology , Cell Differentiation/physiology , Cognitive Dysfunction/etiology , Memory Disorders/etiology , Mice , Neural Stem Cells/metabolism
12.
Brain Behav Immun ; 68: 34-43, 2018 02.
Article in English | MEDLINE | ID: mdl-28943292

ABSTRACT

Mounting evidence supports immune dysfunction in psychiatric conditions such as post-traumatic stress disorder (PTSD). The association of immunomodulatory mechanisms with PTSD-relevant behavior and physiology is not well understood. Communication between neurons and microglia, resident immune cells of the central nervous system, is crucial for optimal regulation of behavior and physiology. In this regard, the fractalkine CX3CL1, secreted from neurons and its target, the microglial CX3CR1 receptor represent a primary neuron-microglia inter-regulatory system important for synaptic plasticity and function. The current study investigated the impact of CX3CR1 deficiency on behaviors relevant to PTSD, such as fear acquisition and memory, acoustic startle response and anxiety-like behavior. Morphological analysis of microglia and neuronal activation within PTSD-relevant forebrain nuclei regulating stress and fear behaviors was also conducted. CX3CR1-deficient (CX3CR1-/-) mice elicited increased fear acquisition as well as reinstatement of fear as compared to wild type (CX3CR1+/+) mice. Conditioned fear and extinction were not significantly different between genotypes. No significant differences were observed in unconditioned acoustic startle response between genotypes. CX3CR1-/- mice showed reduced anxiety-like behaviors as compared with CX3CR1+/+ mice. Morphological assessment of microglia showed region-selective effects of CX3CR1 deficiency, primarily within hypothalamic and cortical areas. Lastly, CX3CR1-/- mice elicited elevated neuronal activity in the PVN and the ventral tegmental-interpeduncular area following reinstatement of fear. Collectively, our data suggest that impaired CX3CR1 function may evoke region-selective alterations in forebrain circuits regulating stress, anxiety and fear, impacting behaviors relevant to disorders such as PTSD.


Subject(s)
CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Fear/physiology , Animals , Anxiety/metabolism , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Disease Models, Animal , Limbic System/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neuronal Plasticity/physiology , Neurons/physiology , Prosencephalon/physiology , Reflex, Startle/physiology , Stress Disorders, Post-Traumatic/metabolism , Stress, Psychological/physiopathology
13.
Brain Behav Immun ; 67: 36-41, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28736033

ABSTRACT

BACKGROUND: While disruption of acid-base homeostasis has been pathoetiologically implicated in panic disorder (PD), the mechanism by which pH imbalance is translated to panic pathophysiology is poorly understood. Recently, in a translational rodent model of PD, we reported a role of microglial acid sensing G-protein coupled receptor, T cell death associated gene-8 (TDAG8) in panic-associated behavior and physiology. However, the clinical validity of the TDAG8 receptor has not been investigated. OBJECTIVE: To assess TDAG8 in PD, we evaluated TDAG8 receptor expression in adolescents and young adults with PD and healthy comparison subjects. METHODS: Relative expression of TDAG8 mRNA was determined in peripheral blood mononuclear cells from patients with PD, and compared to expression in healthy subjects. Linear models were utilized to evaluate the relationship between TDAG8 expression and panic disorder symptom severity scale (PDSS) score as well as other potential explanatory variables (e.g., CRP, body mass index, sex, age). Models were refined based on the estimated parameter significance, evidence of omitted variable bias and Bayesian/Akaike information criteria. RESULTS: Relative to healthy comparison subjects (n=17), expression of TDAG8 mRNA was significantly increased in patients with PD (n=15) (1.60±0.65 vs. 1.01±0.50, p=0.008). TDAG8 mRNA expression predicted PD symptom severity in a fixed effect model incorporating age and sex (p=0.003). CONCLUSIONS: Collectively, our results suggest greater TDAG8 expression in patients with PD compared to healthy subjects, and directly link TDAG8 expression and the severity of the PD symptoms. Further investigation of the TDAG8 receptor in panic pathophysiology is warranted.


Subject(s)
Panic Disorder/metabolism , Receptors, G-Protein-Coupled/metabolism , Adolescent , Adult , C-Reactive Protein/metabolism , Female , Humans , Male , Pilot Projects , RNA, Messenger/metabolism , Severity of Illness Index , Young Adult
14.
Biol Psychiatry ; 80(7): 541-51, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27422366

ABSTRACT

BACKGROUND: Carbon dioxide (CO2) inhalation, a biological challenge and pathologic marker in panic disorder, evokes intense fear and panic attacks in susceptible individuals. The molecular identity and anatomic location of CO2-sensing systems that translate CO2-evoked fear remain unclear. We investigated contributions of microglial acid sensor T cell death-associated gene-8 (TDAG8) and microglial proinflammatory responses in CO2-evoked behavioral and physiological responses. METHODS: CO2-evoked freezing, autonomic, and respiratory responses were assessed in TDAG8-deficient ((-/-)) and wild-type ((+/+)) mice. Involvement of TDAG8-dependent microglial activation and proinflammatory cytokine interleukin (IL)-1ß with CO2-evoked responses was investigated using microglial blocker, minocycline, and IL-1ß antagonist IL-1RA. CO2-chemosensitive firing responses using single-cell patch clamping were measured in TDAG8(-/-) and TDAG8(+/+) mice to gain functional insights. RESULTS: TDAG8 expression was localized in microglia enriched within the sensory circumventricular organs. TDAG8(-/-) mice displayed attenuated CO2-evoked freezing and sympathetic responses. TDAG8 deficiency was associated with reduced microglial activation and proinflammatory cytokine IL-1ß within the subfornical organ. Central infusion of microglial activation blocker minocycline and IL-1ß antagonist IL-1RA attenuated CO2-evoked freezing. Finally, CO2-evoked neuronal firing in patch-clamped subfornical organ neurons was dependent on acid sensor TDAG8 and IL-1ß. CONCLUSIONS: Our data identify TDAG8-dependent microglial acid sensing as a unique chemosensor for detecting and translating hypercapnia to fear-associated behavioral and physiological responses, providing a novel mechanism for homeostatic threat detection of relevance to psychiatric conditions such as panic disorder.


Subject(s)
Carbon Dioxide/pharmacology , Chemoreceptor Cells/physiology , Fear/drug effects , Microglia/drug effects , Microglia/physiology , Action Potentials/physiology , Animals , Hydrogen-Ion Concentration , Immobility Response, Tonic/drug effects , Immobility Response, Tonic/physiology , Interleukin 1 Receptor Antagonist Protein , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Male , Mice , Mice, Knockout , Microglia/metabolism , Microinjections , Minocycline/administration & dosage , Minocycline/pharmacology , Neurons/physiology , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Subfornical Organ/metabolism
15.
J Neurosci ; 36(4): 1306-15, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26818517

ABSTRACT

Neuropeptide Y (NPY), a 36 aa peptide, regulates stress and emotional behaviors. Preclinical and clinical studies support an association of NPY with trauma-evoked syndromes such as posttraumatic stress disorder (PTSD), although the exact contribution of NPY is not clear. In the current study, we examined functional attributes of NPY in the infralimbic (IL) cortex, an area that regulates fear memories and is reported to be hypoactive in PTSD. Carriers of NPY gene polymorphism rs16147 have been reported to have elevated prefrontal NPY expression. Infusion of NPY into the IL cortex in rats significantly impaired fear extinction memory without affecting conditioned fear expression or acquisition of extinction. Neuroendocrine stress response, depression-like behavior, and working memory performance were not affected by NPY infusion into the IL. The NPY Y1 receptor antagonist BIBO3304 completely abolished NPY effects on fear extinction retrieval. Y1 receptor expression was localized on CaMKII-positive pyramidal projection neurons and GAD67-positive interneurons in the IL. Patch-clamp recordings revealed increased inhibitory synaptic transmission onto IL projection neurons in the presence of NPY. Thus, NPY dampens excitability of IL projection neurons and impairs retrieval of extinction memory by inhibiting consolidation of extinction. Of relevance to PTSD, elevation of prefrontal NPY attributable to the genetic polymorphism rs16147 may contribute to IL hypoactivity, resulting in impaired extinction memory and susceptibility to the disorder. SIGNIFICANCE STATEMENT: Neuropeptide Y (NPY), a stress modulatory transmitter, is associated with posttraumatic stress disorder (PTSD). Contribution of NPY to PTSD symptomology is unclear. PTSD patients have reduced activity in the infralimbic (IL) subdivision of the medial prefrontal cortex (mPFC), associated with compromised extinction memory. No information exists on fear modulation by NPY in the IL cortex, although NPY and NPY receptors are abundant in these areas. This study shows that IL NPY inhibits consolidation of extinction, resulting in impaired retrieval of extinction memory and modulates excitability of IL projection neurons. In addition to providing a novel perspective on extinction memory modulation by NPY, our findings suggest that elevated mPFC NPY in gene polymorphism rs16147 carriers or after chronic stress could increase susceptibility to PTSD.


Subject(s)
Extinction, Psychological/drug effects , Fear/drug effects , Learning Disabilities/chemically induced , Mental Recall/drug effects , Neurons/drug effects , Neuropeptide Y/toxicity , Prefrontal Cortex/cytology , Animals , Arginine/analogs & derivatives , Arginine/therapeutic use , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Corticosterone/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Glutamate Decarboxylase/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/metabolism , Synaptic Potentials/drug effects
16.
Physiol Behav ; 150: 78-82, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-25770699

ABSTRACT

Inflammation has been suggested to contribute to the pathophysiology of depression. The T cell death associated gene-8 (TDAG8) receptor is a proton-sensing G-protein-coupled receptor (GPCR) expressed on immune cells in both the CNS and periphery. Previous work has shown modulation of inflammation by the TDAG8 receptor, with pro-inflammatory responses reported in the central nervous system (CNS). Given the link between depression and inflammation, the aim of the present study was to investigate the role of TDAG8 in depression relevant behaviors. Mice deficient in TDAG8 (TDAG8(-/-)) were tested in the forced swim test (FST) and sucrose preference paradigm. TDAG8 deficiency resulted in significant attenuation of immobility in the FST as compared to wild type TDAG8 (TDAG8(+/+)) mice. These differences were not due to alterations in motor activity evoked by TDAG8 deficiency as TDAG8(+/+) and TDAG8(-/-) mice displayed similar activity in the home cage or in a novel context. TDAG8(-/-) mice showed significantly higher consumption of sucrose compared to wild type mice although sucrose preference was not significantly different between genotypes. Collectively, our results support the involvement of the TDAG8 receptor in behavioral response relevant to depression. Further investigation is required to validate TDAG8 as a novel target linking inflammation and depression.


Subject(s)
Depression/genetics , Depression/physiopathology , Analysis of Variance , Animals , Disease Models, Animal , Drinking/genetics , Eating/genetics , Food Preferences , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Motor Activity/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Swimming/psychology
17.
Stress ; 18(2): 244-53, 2015.
Article in English | MEDLINE | ID: mdl-25721540

ABSTRACT

Chronicity of trauma exposure plays an important role in the pathophysiology of posttraumatic stress disorder (PTSD). Thus, exposure to multiple traumas on a chronic scale leads to worse outcomes than acute events. The rationale for the current study was to investigate the effects of a single adverse event versus the same event on a background of chronic stress. We hypothesized that a history of chronic stress would lead to worse behavioral outcomes than a single event alone. Male rats (n = 14/group) were exposed to either a single traumatic event in the form of electric foot shocks (acute shock, AS), or to footshocks on a background of chronic stress (chronic variable stress-shock, CVS-S). PTSD-relevant behaviors (fear memory and acoustic startle responses) were measured following 7 d recovery. In line with our hypothesis, CVS-S elicited significant increases in fear acquisition and conditioning versus the AS group. Unexpectedly, CVS-S elicited reduced startle reactivity to an acoustic stimulus in comparison with the AS group. Significant increase in FosB/ΔFosB-like immunostaining was observed in the dentate gyrus, basolateral amygdala and medial prefrontal cortex of CVS-S rats. Assessments of neuropeptide Y (NPY), a stress-regulatory transmitter associated with chronic PTSD, revealed selective reduction in the hippocampus of CVS-S rats. Collectively, our data show that cumulative stress potentiates delayed fear memory and impacts defensive responding. Altered neuronal activation in forebrain limbic regions and reduced NPY may contribute to these phenomena. Our preclinical studies support clinical findings reporting worse PTSD outcomes stemming from cumulative traumatization in contrast to acute trauma.


Subject(s)
Fear , Memory , Reflex, Startle/physiology , Stress, Psychological/physiopathology , Amygdala/metabolism , Animals , Conditioning, Psychological , Dentate Gyrus/metabolism , Male , Prefrontal Cortex/metabolism , Prosencephalon , Proto-Oncogene Proteins c-fos/metabolism , Rats , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/physiopathology , Stress Disorders, Post-Traumatic/psychology , Stress, Psychological/metabolism , Stress, Psychological/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...