1
|
Obray JD, Denton AR, Carroll-Deaton J, Marquardt K, Chandler LJ, Scofield MD. Enhanced Fear Extinction Through Infralimbic Perineuronal Net Digestion: The Modulatory Role of Adolescent Alcohol Exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619810. [PMID: 39484370 PMCID: PMC11526981 DOI: 10.1101/2024.10.23.619810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Perineuronal nets (PNNs) are specialized components of the extracellular matrix that play a critical role in learning and memory. In a Pavlovian fear conditioning paradigm, degradation of PNNs affects the formation and storage of fear memories. This study examined the impact of adolescent intermittent ethanol (AIE) exposure by vapor inhalation on the expression of PNNs in the adult rat prelimbic (PrL) and infralimbic (IfL) subregions of the medial prefrontal cortex. Results indicated that following AIE, the total number of PNN positive cells in the PrL cortex increased in layer II/III but did not change in layer V. Conversely, in the IfL cortex, the number of PNN positive cells decreased in layer V, with no change in layer II/III. In addition, the intensity of PNN staining was significantly altered by AIE exposure, which narrowed the distribution of signal intensity, reducing the number of high and low intensity PNNs. Given these changes in PNNs, the next experiment assessed the effects of AIE and PNN digestion on extinction of a conditioned fear memory. In Air control rats, digestion of PNNs by bilateral infusion of Chondroitinase ABC (ChABC) into the IfL cortex enhanced fear extinction and reduced contextual fear renewal. In contrast, both fear extinction learning and contextual fear renewal remained unchanged following PNN digestion in AIE exposed rats. These results highlight the sensitivity of prefrontal PNNs to adolescent alcohol exposure and suggest that ChABC-induced plasticity is reduced in the IfL cortex following AIE exposure.
Collapse
Affiliation(s)
- J. Daniel Obray
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Adam R. Denton
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425
- Department of Psychology, Tusculum University, Tusculum, TN 37745
| | - Jayda Carroll-Deaton
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Kristin Marquardt
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - L. Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Michael D. Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
2
|
Kovlyagina I, Wierczeiko A, Todorov H, Jacobi E, Tevosian M, von Engelhardt J, Gerber S, Lutz B. Leveraging interindividual variability in threat conditioning of inbred mice to model trait anxiety. PLoS Biol 2024; 22:e3002642. [PMID: 38805548 PMCID: PMC11161093 DOI: 10.1371/journal.pbio.3002642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/07/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
Trait anxiety is a major risk factor for stress-induced and anxiety disorders in humans. However, animal models accounting for the interindividual variability in stress vulnerability are largely lacking. Moreover, the pervasive bias of using mostly male animals in preclinical studies poorly reflects the increased prevalence of psychiatric disorders in women. Using the threat imminence continuum theory, we designed and validated an auditory aversive conditioning-based pipeline in both female and male mice. We operationalised trait anxiety by harnessing the naturally occurring variability of defensive freezing responses combined with a model-based clustering strategy. While sustained freezing during prolonged retrieval sessions was identified as an anxiety-endophenotype behavioral marker in both sexes, females were consistently associated with an increased freezing response. RNA-sequencing of CeA, BLA, ACC, and BNST revealed massive differences in phasic and sustained responders' transcriptomes, correlating with transcriptomic signatures of psychiatric disorders, particularly post-traumatic stress disorder (PTSD). Moreover, we detected significant alterations in the excitation/inhibition balance of principal neurons in the lateral amygdala. These findings provide compelling evidence that trait anxiety in inbred mice can be leveraged to develop translationally relevant preclinical models to investigate mechanisms of stress susceptibility in a sex-specific manner.
Collapse
Affiliation(s)
- Irina Kovlyagina
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Anna Wierczeiko
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Hristo Todorov
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Eric Jacobi
- Institute of Pathophysiology, and Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Margarita Tevosian
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
| | - Jakob von Engelhardt
- Institute of Pathophysiology, and Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
| |
Collapse
|
3
|
Park K, Park H, Chung C. Fear conditioning and extinction distinctively alter bidirectional synaptic plasticity within the amygdala of an animal model of post-traumatic stress disorder. Neurobiol Stress 2024; 29:100606. [PMID: 38292517 PMCID: PMC10825524 DOI: 10.1016/j.ynstr.2024.100606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
Synaptic plasticity in the amygdala plays an essential role in the formation and inhibition of fear memory; however, this plasticity has mainly been studied in the lateral amygdala, making it largely uninvestigated in other subnuclei. Here, we investigated long-term potentiation (LTP) and long-term depression (LTD) in the basolateral amygdala (BLA) to the medial division of the central amygdala (CEm) synapses of juvenile C57BL/6N (B6) and 129S1/SvImJ (S1) mice. We found that in naïve B6 and S1 mice, LTP was not induced at the BLA to CEm synapses, whereas fear conditioning lowered the threshold for LTP induction in these synapses of both B6 and S1 mice. Interestingly, fear extinction disrupted the induction of LTP at the BLA to CEm synapses of B6 mice, whereas LTP was left intact in S1 mice. Both low-frequency stimulation (LFS) and modest LFS (mLFS) induced LTD in naïve B6 and S1 mice, suggesting that the BLA to CEm synapses express bidirectional plasticity. Fear conditioning disrupted both types of LTD induction selectively in S1 mice and LFS-LTD, presumably NMDAR-dependent LTD was partially recovered by fear extinction. However, mLFS-LTD which has been known to be endocannabinoid receptor 1 (CB1R)-dependent was not induced after fear extinction in both mouse strains. Our observations suggest that fear conditioning enhances LTP while fear extinction diminishes LTP at the BLA to the CEm synapses of B6 mice with successful extinction. Considering that S1 mice showed strong fear conditioning and impaired extinction, strong fear conditioning in the S1 strain may be related to disrupted LTD, and impaired extinction may be due to constant LTP and weak LFS-LTD at the BLA to CEm synapses. Our study contributes to the further understanding of the dynamics of synaptic potentiation and depression between the subnuclei of the amygdala in juvenile mice after fear conditioning and extinction.
Collapse
Affiliation(s)
- Kwanghoon Park
- Department of Biological Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Hoyong Park
- Department of Biological Sciences, Konkuk University, Seoul, 05029, South Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul, 05029, South Korea
| |
Collapse
|
4
|
Gunduz-Cinar O, Castillo LI, Xia M, Van Leer E, Brockway ET, Pollack GA, Yasmin F, Bukalo O, Limoges A, Oreizi-Esfahani S, Kondev V, Báldi R, Dong A, Harvey-White J, Cinar R, Kunos G, Li Y, Zweifel LS, Patel S, Holmes A. A cortico-amygdala neural substrate for endocannabinoid modulation of fear extinction. Neuron 2023; 111:3053-3067.e10. [PMID: 37480845 PMCID: PMC10592324 DOI: 10.1016/j.neuron.2023.06.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 07/24/2023]
Abstract
Preclinical and clinical studies implicate endocannabinoids (eCBs) in fear extinction, but the underlying neural circuit basis of these actions is unclear. Here, we employed in vivo optogenetics, eCB biosensor imaging, ex vivo electrophysiology, and CRISPR-Cas9 gene editing in mice to examine whether basolateral amygdala (BLA)-projecting medial prefrontal cortex (mPFC) neurons represent a neural substrate for the effects of eCBs on extinction. We found that photoexcitation of mPFC axons in BLA during extinction mobilizes BLA eCBs. eCB biosensor imaging showed that eCBs exhibit a dynamic stimulus-specific pattern of activity at mPFC→BLA neurons that tracks extinction learning. Furthermore, using CRISPR-Cas9-mediated gene editing, we demonstrated that extinction memory formation involves eCB activity at cannabinoid CB1 receptors expressed at vmPFC→BLA synapses. Our findings reveal the temporal characteristics and a neural circuit basis of eCBs' effects on fear extinction and inform efforts to target the eCB system as a therapeutic approach in extinction-deficient neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ozge Gunduz-Cinar
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA.
| | - Laura I Castillo
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Maya Xia
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Elise Van Leer
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Emma T Brockway
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Gabrielle A Pollack
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Farhana Yasmin
- Northwestern Center for Psychiatric Neuroscience, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Olena Bukalo
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Aaron Limoges
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Sarvar Oreizi-Esfahani
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Veronika Kondev
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Rita Báldi
- Northwestern Center for Psychiatric Neuroscience, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ao Dong
- Peking University School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Judy Harvey-White
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA; Section on Fibrotic Disorders, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Yulong Li
- Peking University School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Sachin Patel
- Northwestern Center for Psychiatric Neuroscience, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
5
|
Ramos A, Granzotto N, Kremer R, Boeder AM, de Araújo JFP, Pereira AG, Izídio GS. Hunting for Genes Underlying Emotionality in the Laboratory Rat: Maps, Tools and Traps. Curr Neuropharmacol 2023; 21:1840-1863. [PMID: 36056863 PMCID: PMC10514530 DOI: 10.2174/1570159x20666220901154034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/13/2022] [Accepted: 07/28/2022] [Indexed: 11/22/2022] Open
Abstract
Scientists have systematically investigated the hereditary bases of behaviors since the 19th century, moved by either evolutionary questions or clinically-motivated purposes. The pioneer studies on the genetic selection of laboratory animals had already indicated, one hundred years ago, the immense complexity of analyzing behaviors that were influenced by a large number of small-effect genes and an incalculable amount of environmental factors. Merging Mendelian, quantitative and molecular approaches in the 1990s made it possible to map specific rodent behaviors to known chromosome regions. From that point on, Quantitative Trait Locus (QTL) analyses coupled with behavioral and molecular techniques, which involved in vivo isolation of relevant blocks of genes, opened new avenues for gene mapping and characterization. This review examines the QTL strategy applied to the behavioral study of emotionality, with a focus on the laboratory rat. We discuss the challenges, advances and limitations of the search for Quantitative Trait Genes (QTG) playing a role in regulating emotionality. For the past 25 years, we have marched the long journey from emotionality-related behaviors to genes. In this context, our experiences are used to illustrate why and how one should move forward in the molecular understanding of complex psychiatric illnesses. The promise of exploring genetic links between immunological and emotional responses are also discussed. New strategies based on humans, rodents and other animals (such as zebrafish) are also acknowledged, as they are likely to allow substantial progress to be made in the near future.
Collapse
Affiliation(s)
- André Ramos
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Natalli Granzotto
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Rafael Kremer
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Developmental and Cellular Biology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Ariela Maína Boeder
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Julia Fernandez Puñal de Araújo
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Developmental and Cellular Biology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Aline Guimarães Pereira
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Developmental and Cellular Biology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Geison Souza Izídio
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Developmental and Cellular Biology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| |
Collapse
|
6
|
Fawcett JW, Fyhn M, Jendelova P, Kwok JCF, Ruzicka J, Sorg BA. The extracellular matrix and perineuronal nets in memory. Mol Psychiatry 2022; 27:3192-3203. [PMID: 35760878 PMCID: PMC9708575 DOI: 10.1038/s41380-022-01634-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 02/06/2023]
Abstract
All components of the CNS are surrounded by a diffuse extracellular matrix (ECM) containing chondroitin sulphate proteoglycans (CSPGs), heparan sulphate proteoglycans (HSPGs), hyaluronan, various glycoproteins including tenascins and thrombospondin, and many other molecules that are secreted into the ECM and bind to ECM components. In addition, some neurons, particularly inhibitory GABAergic parvalbumin-positive (PV) interneurons, are surrounded by a more condensed cartilage-like ECM called perineuronal nets (PNNs). PNNs surround the soma and proximal dendrites as net-like structures that surround the synapses. Attention has focused on the role of PNNs in the control of plasticity, but it is now clear that PNNs also play an important part in the modulation of memory. In this review we summarize the role of the ECM, particularly the PNNs, in the control of various types of memory and their participation in memory pathology. PNNs are now being considered as a target for the treatment of impaired memory. There are many potential treatment targets in PNNs, mainly through modulation of the sulphation, binding, and production of the various CSPGs that they contain or through digestion of their sulphated glycosaminoglycans.
Collapse
Affiliation(s)
- James W Fawcett
- John van Geest Centre for Brain Repair, Department Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK.
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic.
| | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Pavla Jendelova
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Jessica C F Kwok
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Jiri Ruzicka
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Barbara A Sorg
- Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| |
Collapse
|
7
|
|
8
|
Zhang WH, Zhang JY, Holmes A, Pan BX. Amygdala Circuit Substrates for Stress Adaptation and Adversity. Biol Psychiatry 2021; 89:847-856. [PMID: 33691931 DOI: 10.1016/j.biopsych.2020.12.026] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/24/2020] [Accepted: 12/18/2020] [Indexed: 12/19/2022]
Abstract
Brain systems that promote maintenance of homeostasis in the face of stress have significant adaptive value. A growing body of work across species demonstrates a critical role for the amygdala in promoting homeostasis by regulating physiological and behavioral responses to stress. This review focuses on an emerging body of evidence that has begun to delineate the contribution of specific long-range amygdala circuits in mediating the effects of stress. After summarizing the major anatomical features of the amygdala and its connectivity to other limbic structures, we discuss recent findings from rodents showing how stress causes structural and functional remodeling of amygdala neuronal outputs to defined cortical and subcortical target regions. We also consider some of the environmental and genetic factors that have been found to moderate how the amygdala responds to stress and relate the emerging preclinical literature to the current understanding of the pathophysiology and treatment of stress-related neuropsychiatric disorders. Future effort to translate these findings to clinics may help to develop valuable tools for prevention, diagnosis, and treatment of these diseases.
Collapse
Affiliation(s)
- Wen-Hua Zhang
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Jun-Yu Zhang
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institues of Health, Bethesda, Maryland
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China.
| |
Collapse
|
9
|
Nonaka M, Taylor WW, Bukalo O, Tucker LB, Fu AH, Kim Y, McCabe JT, Holmes A. Behavioral and Myelin-Related Abnormalities after Blast-Induced Mild Traumatic Brain Injury in Mice. J Neurotrauma 2021; 38:1551-1571. [PMID: 33605175 DOI: 10.1089/neu.2020.7254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In civilian and military settings, mild traumatic brain injury (mTBI) is a common consequence of impacts to the head, sudden blows to the body, and exposure to high-energy atmospheric shockwaves from blast. In some cases, mTBI from blast exposure results in long-term emotional and cognitive deficits and an elevated risk for certain neuropsychiatric diseases. Here, we tested the effects of mTBI on various forms of auditory-cued fear learning and other measures of cognition in male C57BL/6J mice after single or repeated blast exposure (blast TBI; bTBI). bTBI produced an abnormality in the temporal organization of cue-induced freezing behavior in a conditioned trace fear test. Spatial working memory, evaluated by the Y-maze task performance, was also deleteriously affected by bTBI. Reverse-transcription quantitative real-time polymerase chain reaction (RT-qPCR) analysis for glial markers indicated an alteration in the expression of myelin-related genes in the hippocampus and corpus callosum 1-8 weeks after bTBI. Immunohistochemical and ultrastructural analyses detected bTBI-related myelin and axonal damage in the hippocampus and corpus callosum. Together, these data suggest a possible link between blast-induced mTBI, myelin/axonal injury, and cognitive dysfunction.
Collapse
Affiliation(s)
- Mio Nonaka
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIH), Rockville, Maryland, USA
| | - William W Taylor
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIH), Rockville, Maryland, USA
| | - Olena Bukalo
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIH), Rockville, Maryland, USA
| | - Laura B Tucker
- Department of Anatomy, Physiology and Genetics, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Preclinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Amanda H Fu
- Department of Anatomy, Physiology and Genetics, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Preclinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Yeonho Kim
- Department of Anatomy, Physiology and Genetics, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Preclinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Joseph T McCabe
- Department of Anatomy, Physiology and Genetics, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Preclinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health (NIH), Rockville, Maryland, USA
| |
Collapse
|
10
|
Taylor WW, Imhoff BR, Sathi ZS, Liu WY, Garza KM, Dias BG. Contributions of glucocorticoid receptors in cortical astrocytes to memory recall. Learn Mem 2021; 28:126-133. [PMID: 33723032 PMCID: PMC7970741 DOI: 10.1101/lm.053041.120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/14/2021] [Indexed: 01/15/2023]
Abstract
Dysfunctions in memory recall lead to pathological fear; a hallmark of trauma-related disorders, like posttraumatic stress disorder (PTSD). Both, heightened recall of an association between a cue and trauma, as well as impoverished recall that a previously trauma-related cue is no longer a threat, result in a debilitating fear toward the cue. Glucocorticoid-mediated action via the glucocorticoid receptor (GR) influences memory recall. This literature has primarily focused on GRs expressed in neurons or ignored cell-type specific contributions. To ask how GR action in nonneuronal cells influences memory recall, we combined auditory fear conditioning in mice and the knockout of GRs in astrocytes in the prefrontal cortex (PFC), a brain region implicated in memory recall. We found that knocking out GRs in astrocytes of the PFC disrupted memory recall. Specifically, we found that knocking out GRs in astrocytes in the PFC (AstroGRKO) after fear conditioning resulted in higher levels of freezing to the CS+ tone when compared with controls (AstroGRintact). While we did not find any differences in extinction of fear toward the CS+ between these groups, AstroGRKO female but not male mice showed impaired recall of extinction training. These results suggest that GRs in cortical astrocytes contribute to memory recall. These data demonstrate the need to examine GR action in cortical astrocytes to elucidate the basic neurobiology underlying memory recall and potential mechanisms that underlie female-specific biases in the incidence of PTSD.
Collapse
Affiliation(s)
- William W Taylor
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia 30322, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California 90007, USA
- Developmental Neuroscience and Neurogenetics Program, Division of Research on Children, Youth, and Families, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | - Barry R Imhoff
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia 30322, USA
| | - Zakia Sultana Sathi
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia 30322, USA
| | - Wei Y Liu
- Developmental Neuroscience and Neurogenetics Program, Division of Research on Children, Youth, and Families, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | - Kristie M Garza
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, USA
| | - Brian G Dias
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia 30322, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California 90007, USA
- Developmental Neuroscience and Neurogenetics Program, Division of Research on Children, Youth, and Families, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia 30322, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California 90027, USA
| |
Collapse
|
11
|
Rodriguez G, Moore SJ, Neff RC, Glass ED, Stevenson TK, Stinnett GS, Seasholtz AF, Murphy GG, Cazares VA. Deficits across multiple behavioral domains align with susceptibility to stress in 129S1/SvImJ mice. Neurobiol Stress 2020; 13:100262. [PMID: 33344715 PMCID: PMC7739066 DOI: 10.1016/j.ynstr.2020.100262] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/07/2020] [Accepted: 10/16/2020] [Indexed: 01/08/2023] Open
Abstract
Acute physical or psychological stress can elicit adaptive behaviors that allow an organism maintain homeostasis. However, intense and/or prolonged stressors often have the opposite effect, resulting in maladaptive behaviors and curbing goal-directed action; in the extreme, this may contribute to the development of psychiatric conditions like generalized anxiety disorder, major depressive disorder, or post-traumatic stress disorder. While treatment of these disorders generally focuses on reducing reactivity to potentially threatening stimuli, there are in fact impairments across multiple domains including valence, arousal, and cognition. Here, we use the genetically stress-susceptible 129S1 mouse strain to explore the effects of stress across multiple domains. We find that 129S1 mice exhibit a potentiated neuroendocrine response across many environments and paradigms, and that this is associated with reduced exploration, neophobia, decreased novelty- and reward-seeking, and spatial learning and memory impairments. Taken together, our results suggest that the 129S1 strain may provide a useful model for elucidating mechanisms underlying myriad aspects of stress-linked psychiatric disorders as well as potential treatments that may ameliorate symptoms.
Collapse
Affiliation(s)
- G Rodriguez
- Michigan Neuroscience Institute, USA.,Neuroscience Graduate Program, USA
| | - S J Moore
- Department of Molecular and Integrative Physiology, USA.,Michigan Neuroscience Institute, USA
| | - R C Neff
- Department of Molecular and Integrative Physiology, USA
| | - E D Glass
- Department of Molecular and Integrative Physiology, USA.,Michigan Neuroscience Institute, USA
| | | | | | - A F Seasholtz
- Michigan Neuroscience Institute, USA.,Neuroscience Graduate Program, USA.,Department of Biological Chemistry University of Michigan, Ann Arbor, MI, USA
| | - G G Murphy
- Department of Molecular and Integrative Physiology, USA.,Michigan Neuroscience Institute, USA.,Neuroscience Graduate Program, USA
| | - V A Cazares
- Department of Molecular and Integrative Physiology, USA.,Michigan Neuroscience Institute, USA.,Department of Psychology, Williams College, MA, USA
| |
Collapse
|
12
|
Tubbs JD, Ding J, Baum L, Sham PC. Systemic neuro-dysregulation in depression: Evidence from genome-wide association. Eur Neuropsychopharmacol 2020; 39:1-18. [PMID: 32896454 DOI: 10.1016/j.euroneuro.2020.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/10/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022]
Abstract
Depression is the world's leading cause of disability. Greater understanding of the neurobiological basis of depression is necessary for developing novel treatments with improved efficacy and acceptance. Recently, major advances have been made in the search for genetic variants associated with depression which may help to elucidate etiological mechanisms. The present review has two major objectives. First, we offer a brief review of two major biological systems with strong evidence for involvement in depression pathology: neurotransmitter systems and the stress response. Secondly, we provide a synthesis of the functions of the 269 genes implicated by the most recent genome-wide meta-analysis, supporting the importance of these systems in depression and providing insights into other possible mechanisms involving neurodevelopment, neurogenesis, and neurodegeneration. Our goal is to undertake a broad, preliminary stock-taking of the most recent hypothesis-free findings and examine the weight of the evidence supporting these existing theories and highlighting novel directions. This qualitative review and accompanying gene function table provides a valuable resource and guide for basic and translational researchers, with suggestions for future mechanistic research, leveraging genetics to prioritize studies on the neurobiological processes involved in depression etiology and treatment.
Collapse
Affiliation(s)
- Justin D Tubbs
- Department of Psychiatry, The University of Hong Kong, Hong Kong
| | - Jiahong Ding
- Department of Psychiatry, The University of Hong Kong, Hong Kong
| | - Larry Baum
- Department of Psychiatry, The University of Hong Kong, Hong Kong; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
| | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong; Centre of PanorOmic Sciences, The University of Hong Kong, Hong Kong.
| |
Collapse
|
13
|
Effects of optogenetic photoexcitation of infralimbic cortex inputs to the basolateral amygdala on conditioned fear and extinction. Behav Brain Res 2020; 396:112913. [PMID: 32950607 DOI: 10.1016/j.bbr.2020.112913] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/28/2020] [Accepted: 09/10/2020] [Indexed: 11/21/2022]
Abstract
Deficiencies in the ability to extinguish fear is a hallmark of Trauma- and stressor-related disorders, Anxiety disorders, and certain other neuropsychiatric conditions. Hence, a greater understanding of the brain mechanisms involved in the inhibition of fear is of significant translational relevance. Previous studies in rodents have shown that glutamatergic projections from the infralimbic prefrontal cortex (IL) to basolateral amygdala (BLA) play a crucial instructional role in the formation of extinction memories, and also indicate that variation in the strength of this input correlates with extinction efficacy. To further examine the relationship between the IL→BLA pathway and extinction we expressed three different titers of the excitatory opsin, channelrhodopsin (ChR2), in IL neurons and photostimulated their projections in the BLA during partial extinction training. The behavioral effects of photoexcitation differed across the titer groups: the low titer had no effect, the medium titer selectively facilitated extinction memory formation, and the high titer produced both an acute suppression of fear and a decrease in fear during (light-free) extinction retrieval. We discuss various possible explanations for these titer-specific effects, including the possibility of IL-mediated inhibition of BLA fear-encoding neurons under conditions of sufficiently strong photoexcitation. These findings further support the role of IL→BLA pathway in regulating fear and highlight the importance of methodological factors in optogenetic studies of neural circuits underling behavior.
Collapse
|
14
|
Bergstrom HC, Lieberman AG, Graybeal C, Lipkin AM, Holmes A. Dorsolateral striatum engagement during reversal learning. ACTA ACUST UNITED AC 2020; 27:418-422. [PMID: 32934094 PMCID: PMC7497112 DOI: 10.1101/lm.051714.120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/19/2020] [Indexed: 01/05/2023]
Abstract
Most experimental preparations demonstrate a role for dorsolateral striatum (DLS) in stimulus-response, but not outcome-based, learning. Here, we assessed DLS involvement in a touchscreen-based reversal task requiring mice to update choice following a change in stimulus-reward contingencies. In vivo single-unit recordings in the DLS showed reversal produced a population-level shift from excited to inhibited neuronal activity prior to choices being made. The larger the shift, the faster mice reversed. Furthermore, optogenetic photosilencing DLS neurons during choice increased early reversal errors. These findings suggest dynamic DLS engagement may facilitate reversal, possibly by signaling a change in contingencies to other striatal and cortical regions.
Collapse
Affiliation(s)
- Hadley C Bergstrom
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland 20852, USA
| | - Abby G Lieberman
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland 20852, USA
| | - Carolyn Graybeal
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland 20852, USA
| | - Anna M Lipkin
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland 20852, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcoholism and Alcohol Abuse (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland 20852, USA
| |
Collapse
|
15
|
Improved visual discrimination learning in mice with partial 5-HT2B gene deletion. Neurosci Lett 2020; 738:135378. [PMID: 32920046 DOI: 10.1016/j.neulet.2020.135378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/31/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
The neurotransmitter serotonin (5-hydroxytryptamine, 5-HT) has been linked to multiple aspects of cognition. For example, in rodents, discrimination and reversal learning are altered by experimentally induced changes in brain serotonin levels, and reduced expression of the 5-HT2B receptor subtype in mice and humans is associated with decreased serotonergic tone and increased behavioral impulsivity. Serotonin modulates cognitive flexibility as well as fear and anxiety, but the specific contributions of 5-HT2B receptors to these behaviors is unknown. The current study assessed mice with partial Htr2b deletion for performance on a touchscreen-based pairwise visual discrimination and reversal learning task followed by a test of cued fear learning. Male Htr2b heterozygous mice (+/-) and littermate controls (+/+) were trained to discriminate between two visual stimuli presented on a touch-sensitive screen, one which predicted delivery of a 14-mg food pellet and the other which was not rewarded. Once discrimination performance criterion was attained, the stimulus-reward contingencies were reversed. Htr2b +/- mice were faster to reach discrimination criterion than +/+ controls, and made fewer errors. Htr2b +/- mice were also slower to make responses and collect rewards. Conversely, measures of reversal learning were not different between genotypes. Pavlovian cued fear conditioning was also normal in Htr2b +/-mice. These data demonstrate a selective improvement in touchscreen-based discrimination learning in mice with partial deletion of the 5-HT2B receptor, and provide further insight into the role of the 5-HT2B receptor in cognition.
Collapse
|
16
|
Kainate Receptor Auxiliary Subunit NETO2-Related Cued Fear Conditioning Impairments Associate with Defects in Amygdala Development and Excitability. eNeuro 2020; 7:ENEURO.0541-19.2020. [PMID: 32788298 PMCID: PMC7470932 DOI: 10.1523/eneuro.0541-19.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 07/17/2020] [Accepted: 07/24/2020] [Indexed: 11/21/2022] Open
Abstract
NETO2 is an auxiliary subunit for kainate-type glutamate receptors that mediate normal cued fear expression and extinction. Since the amygdala is critical for these functions, we asked whether Neto2 -/- mice have compromised amygdala function. We measured the abundance of molecular markers of neuronal maturation and plasticity, parvalbumin-positive (PV+), perineuronal net-positive (PNN+), and double positive (PV+PNN+) cells in the Neto2 -/- amygdala. We found that Neto2 -/- adult, but not postnatal day (P)23, mice had 7.5% reduction in the fraction of PV+PNN+ cells within the total PNN+ population, and 23.1% reduction in PV staining intensity compared with Neto2 +/+ mice, suggesting that PV interneurons in the adult Neto2 -/- amygdala remain in an immature state. An immature PV inhibitory network would be predicted to lead to stronger amygdalar excitation. In the amygdala of adult Neto2 -/- mice, we identified increased glutamatergic and reduced GABAergic transmission using whole-cell patch-clamp recordings. This was accompanied by increased spine density of thin dendrites in the basal amygdala (BA) compared with Neto2 +/+ mice, indicating stronger glutamatergic synapses. Moreover, after fear acquisition Neto2 -/- mice had a higher number of c-Fos-positive cells than Neto2 +/+ mice in the lateral amygdala (LA), BA, and central amygdala (CE). Altogether, our findings indicate that Neto2 is involved in the maturation of the amygdala PV interneuron network. Our data suggest that this defect, together with other processes influencing amygdala principal neurons, contribute to increased amygdalar excitability, higher fear expression, and delayed extinction in cued fear conditioning, phenotypes that are common in fear-related disorders, including the posttraumatic stress disorder (PTSD).
Collapse
|
17
|
Selective sub-nucleus effects of intra-amygdala oxytocin on fear extinction. Behav Brain Res 2020; 393:112798. [PMID: 32653556 DOI: 10.1016/j.bbr.2020.112798] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/16/2020] [Accepted: 07/03/2020] [Indexed: 01/20/2023]
Abstract
There is growing evidence that the neuropeptide oxytocin (OT) modulates fear and extinction in humans and rodents through actions in corticolimbic circuits including the central amygdala (CeA). Prior studies have, however, been limited to subjects that exhibit intact basal extinction, rather than extinction-impaired populations that could potentially therapeutically benefit from viable OT-targeting treatments. Here, we assessed the effects of pre-extinction training infusion of OT into the CeA, or basolateral amygdala (BLA), on extinction in an inbred mouse strain (S1) model of impaired extinction. We found that intra-CeA OT, at a dose of 0.01 μg, enabled extinction memory formation, as evidenced by lesser freezing as compared to vehicle-infused controls on a drug-free retrieval test. Conversely, infusion of a higher, 1.0 μg OT dose, markedly reduced freezing and increased grooming during extinction training and produced elevated freezing on drug-free retrieval. Infusion of the 0.01 μg dose into the BLA was without behavioral effects. Together, our data show that OT acts in a dose-dependent manner within the CeA to promote extinction in otherwise extinction-deficient mice. These findings provide further support for the potential utility of OT as an adjunctive treatment to extinction-based therapies for trauma and anxiety disorders.
Collapse
|
18
|
Halladay LR, Kocharian A, Piantadosi PT, Authement ME, Lieberman AG, Spitz NA, Coden K, Glover LR, Costa VD, Alvarez VA, Holmes A. Prefrontal Regulation of Punished Ethanol Self-administration. Biol Psychiatry 2020; 87:967-978. [PMID: 31937415 PMCID: PMC7217757 DOI: 10.1016/j.biopsych.2019.10.030] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND A clinical hallmark of alcohol use disorder is persistent drinking despite potential adverse consequences. The ventromedial prefrontal cortex (vmPFC) and dorsomedial prefrontal cortex (dmPFC) are positioned to exert top-down control over subcortical regions, such as the nucleus accumbens shell (NAcS) and basolateral amygdala, which encode positive and negative valence of ethanol (EtOH)-related stimuli. Prior rodent studies have implicated these regions in regulation of punished EtOH self-administration (EtOH-SA). METHODS We conducted in vivo electrophysiological recordings in mouse vmPFC and dmPFC to obtain neuronal correlates of footshock-punished EtOH-SA. Ex vivo recordings were performed in NAcS D1 receptor-expressing medium spiny neurons receiving vmPFC input to examine punishment-related plasticity in this pathway. Optogenetic photosilencing was employed to assess the functional contribution of the vmPFC, dmPFC, vmPFC projections to NAcS, or vmPFC projections to basolateral amygdala, to punished EtOH-SA. RESULTS Punishment reduced EtOH lever pressing and elicited aborted presses (lever approach followed by rapid retraction). Neurons in the vmPFC and dmPFC exhibited phasic firing to EtOH lever presses and aborts, but only in the vmPFC was there a population-level shift in coding from lever presses to aborts with punishment. Closed-loop vmPFC, but not dmPFC, photosilencing on a postpunishment probe test negated the reduction in EtOH lever presses but not in aborts. Punishment was associated with altered plasticity at vmPFC inputs to D1 receptor-expressing medium spiny neurons in the NAcS. Photosilencing vmPFC projections to the NAcS, but not to the basolateral amygdala, partially reversed suppression of EtOH lever presses on probe testing. CONCLUSIONS These findings demonstrate a key role for the vmPFC in regulating EtOH-SA after punishment, with implications for understanding the neural basis of compulsive drinking in alcohol use disorder.
Collapse
Affiliation(s)
- Lindsay R Halladay
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Department of Psychology, Santa Clara University, Santa Clara, California.
| | - Adrina Kocharian
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Patrick T Piantadosi
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Michael E Authement
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Abby G Lieberman
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Nathen A Spitz
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Kendall Coden
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Lucas R Glover
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Vincent D Costa
- Department of Behavioral Neuroscience, Oregon Health Sciences University, Portland, Oregon
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, Maryland
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
19
|
Pan HQ, Zhang WH, Liao CZ, He Y, Xiao ZM, Qin X, Liu WZ, Wang N, Zou JX, Liu XX, Pan BX. Chronic Stress Oppositely Regulates Tonic Inhibition in Thy1-Expressing and Non-expressing Neurons in Amygdala. Front Neurosci 2020; 14:299. [PMID: 32362809 PMCID: PMC7180173 DOI: 10.3389/fnins.2020.00299] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/16/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic or prolonged exposure to stress ranks among the most important socioenvironmental factors contributing to the development of neuropsychiatric diseases, a process generally associated with loss of inhibitory tone in amygdala. Recent studies have identified distinct neuronal circuits within the basolateral amygdala (BLA) engaged in different emotional processes. However, the potential circuit involved in stress-induced dysregulation of inhibitory tones in BLA remains elusive. Here, a transgenic mouse model expressing yellow fluorescent protein under control of the Thy1 promoter was used to differentiate subpopulations of projection neurons (PNs) within the BLA. We observed that the tonic inhibition in amygdala neurons expressing and not expressing Thy1 (Thy1+/-) was oppositely regulated by chronic social defeat stress (CSDS). In unstressed control mice, the tonic inhibitory currents were significantly stronger in Thy1- PNs than their Thy1+ counterparts. CSDS markedly reduced the currents in Thy1- projection neurons (PNs), but increased that in Thy1+ ones. By contrast, CSDS failed to affect both the phasic A-type γ-aminobutyric acid receptor (GABAAR) currents and GABABR currents in these two PN populations. Moreover, chronic corticosterone administration was sufficient to mimic the effect of CSDS on the tonic inhibition of Thy1+ and Thy1- PNs. As a consequence, the suppression of tonic GABAAR currents on the excitability of Thy1- PNs was weakened by CSDS, but enhanced in Thy1+ PNs. The differential regulation of chronic stress on the tonic inhibition in Thy1+ and Thy1- neurons may orchestrate cell-specific adaptation of amygdala neurons to chronic stress.
Collapse
Affiliation(s)
- Han-Qing Pan
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Wen-Hua Zhang
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Cai-Zhi Liao
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Ye He
- Center for Medical Experiments, Nanchang University, Nanchang, China
| | - Zhi-Ming Xiao
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Xia Qin
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Wei-Zhu Liu
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Na Wang
- Department of Physiology, Mudanjiang Medical University, Mudanjiang, China
| | - Jia-Xin Zou
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Xiao-Xuan Liu
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Bing-Xing Pan
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| |
Collapse
|
20
|
Bjorni M, Rovero NG, Yang ER, Holmes A, Halladay LR. Phasic signaling in the bed nucleus of the stria terminalis during fear learning predicts within- and across-session cued fear expression. ACTA ACUST UNITED AC 2020; 27:83-90. [PMID: 32071254 PMCID: PMC7029722 DOI: 10.1101/lm.050807.119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/22/2019] [Indexed: 01/02/2023]
Abstract
While results from many past studies have implicated the bed nucleus of the stria terminalis (BNST) in mediating the expression of sustained negative affect, recent studies have highlighted a more complex role for BNST that includes aspects of fear learning in addition to defensive responding. As BNST is thought to encode ambiguous or unpredictable threat, it seems plausible that it may be involved in encoding early cued fear learning, especially immediately following a first tone-shock pairing when the conditioned stimulus–unconditioned stimulus (CS–US) contingency is not fully apparent. To investigate this, we conducted in vivo electrophysiological recording studies to examine neural dynamics of BNST units during cued fear acquisition and recall. We identified two functionally distinct subpopulations of BNST neurons that encode the intertrial interval (ITI) and may contribute to within- and across-session fear learning. “Ramping” cell activity during cued fear acquisition parallels the increase in freezing expression as mice learn the CS–US contingency, while “Phasic” cells encode postshock (USpost) periods (30 sec following encounter with footshock) only during early trials. Importantly, the magnitude of Phasic unit responsivity to the first USpost period predicted not only freezing expression in response to the subsequent CS during acquisition, but also CS freezing evoked 24 h later during CS retrieval. These findings suggest for the first time that BNST activity may serve as an instructive signal during cued fear learning.
Collapse
Affiliation(s)
- Max Bjorni
- Department of Psychology, Santa Clara University, Santa Clara, California 95053, USA
| | - Natalie G Rovero
- Department of Psychology, Santa Clara University, Santa Clara, California 95053, USA
| | - Elissa R Yang
- Department of Psychology, Santa Clara University, Santa Clara, California 95053, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852, USA
| | - Lindsay R Halladay
- Department of Psychology, Santa Clara University, Santa Clara, California 95053, USA.,Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852, USA
| |
Collapse
|
21
|
Excitation of Diverse Classes of Cholecystokinin Interneurons in the Basal Amygdala Facilitates Fear Extinction. eNeuro 2019; 6:ENEURO.0220-19.2019. [PMID: 31636080 PMCID: PMC6838687 DOI: 10.1523/eneuro.0220-19.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 01/25/2023] Open
Abstract
There is growing evidence that interneurons (INs) orchestrate neural activity and plasticity in corticoamygdala circuits to regulate fear behaviors. However, defining the precise role of cholecystokinin-expressing INs (CCK INs) remains elusive due to the technical challenge of parsing this population from CCK-expressing principal neurons (CCK PNs). Here, we used an intersectional genetic strategy in CCK-Cre;Dlx5/6-Flpe double-transgenic mice to study the anatomical, molecular and electrophysiological properties of CCK INs in the basal amygdala (BA) and optogenetically manipulate these cells during fear extinction. Electrophysiological recordings confirmed that this strategy targeted GABAergic cells and that a significant proportion expressed functional cannabinoid CB1 receptors; a defining characteristic of CCK-expressing basket cells. However, immunostaining showed that subsets of the genetically-targeted cells expressed either neuropeptide Y (NPY; 29%) or parvalbumin (PV; 17%), but not somatostatin (SOM) or Ca2+/calmodulin-dependent protein kinase II (CaMKII)-α. Further morphological and electrophysiological analyses showed that four IN types could be identified among the EYFP-expressing cells: CCK/cannabinoid receptor type 1 (CB1R)-expressing basket cells, neurogliaform cells, PV+ basket cells, and PV+ axo-axonic cells. At the behavioral level, in vivo optogenetic photostimulation of the targeted population during extinction acquisition led to reduced freezing on a light-free extinction retrieval test, indicating extinction memory facilitation; whereas photosilencing was without effect. Conversely, non-selective (i.e., inclusive of INs and PNs) photostimulation or photosilencing of CCK-targeted cells, using CCK-Cre single-transgenic mice, impaired extinction. These data reveal an unexpectedly high degree of phenotypic complexity in a unique population of extinction-modulating BA INs.
Collapse
|
22
|
Toward an animal model of borderline personality disorder. Psychopharmacology (Berl) 2019; 236:2485-2500. [PMID: 31201478 PMCID: PMC6697600 DOI: 10.1007/s00213-019-05289-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 05/30/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Borderline personality disorder (BPD) is a pervasive psychiatric disorder characterized by emotion dysregulation, impulsivity, impaired self-perceptions, and interpersonal relationships and currently affects 1-3% of the US population as reported by Torgersen et al. (Arch Gen Psychiatry 58:590-596, Torgersen et al. 2001), Lenzenweger et al. (Biol Psychiatry 62:553-564, Lenzenweger et al. 2007), and Tomko et al. (J Personal Disord 28:734-750, Tomko et al. 2014). One major obstacle to our understanding of the neural underpinnings of BPD is a lack of valid animal models that translate the key known features of the disorder to a system that is amenable to study. OBJECTIVE To summarize the etiology, major symptoms, and symptom triggers of BPD and then propose a blueprint for building an animal model of BPD by choosing key components of the disorder that can be implemented in rodents. RESULTS We identify the role of early life stress and subsequent mild stress in adulthood as contributing etiological factors and the potential use of altered communication between frontal cortices and the amygdala in extinction and habituation, increased impulsivity, dysregulation of the hypothalamic pituitary axis (HPA), and increased neuroinflammation as biological markers of BPD. Building upon these features of BPD, we propose a two-hit animal model that uses maternal abandonment to alter maturation of the HPA axis and mild secondary adult stress to evoke behavioral symptoms such as increased impulsivity and impaired extinction, habituation, and social interactions. CONCLUSION Through exploration of the etiology, symptom presentation, and altered neurological function, we propose an animal model of BPD. We believe that a number of existing animal paradigms that model other mental health disorders should be combined in a unique way to reflect the etiology, symptom presentation, and altered neurological function that is evident in BPD. These model, when compared with available human data, will inform research and treatment in humans for better understanding of systems from the micro-molecular level to more global physiology underlying BPD.
Collapse
|
23
|
Sengupta A, Holmes A. A Discrete Dorsal Raphe to Basal Amygdala 5-HT Circuit Calibrates Aversive Memory. Neuron 2019; 103:489-505.e7. [PMID: 31204082 DOI: 10.1016/j.neuron.2019.05.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/14/2019] [Accepted: 05/15/2019] [Indexed: 11/26/2022]
Abstract
Despite a wealth of clinical and preclinical data implicating the serotonin (5-HT) system in fear-related affective disorders, a precise definition of this neuromodulator's role in fear remains elusive. Using convergent anatomical and functional approaches, we interrogate the contribution to fear of basal amygdala (BA) 5-HT inputs from the dorsal raphe nucleus (DRN). We show the DRN→BA 5-HT pathway is engaged during fear memory formation and retrieval, and activity of these projections facilitates fear and impairs extinction. The DRN→BA 5-HT pathway amplifies fear-associated BA neuronal firing and theta power and phase-locking. Although fear recruits 5-HT and VGluT3 co-expressing DRN neurons, the fear-potentiating influence of the DRN→BA 5-HT pathway requires signaling at BA 5-HT1A/2A receptors. Input-output mapping illustrates how the DRN→BA 5-HT pathway is anatomically distinct and connected with other brain regions that mediate fear. These findings reveal how a discrete 5-HT circuit orchestrates a broader neural network to calibrate aversive memory.
Collapse
Affiliation(s)
- Ayesha Sengupta
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA.
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA.
| |
Collapse
|
24
|
Cazares VA, Rodriguez G, Parent R, Ouillette L, Glanowska KM, Moore SJ, Murphy GG. Environmental variables that ameliorate extinction learning deficits in the 129S1/SvlmJ mouse strain. GENES BRAIN AND BEHAVIOR 2019; 18:e12575. [PMID: 30973205 DOI: 10.1111/gbb.12575] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 03/28/2019] [Accepted: 04/09/2019] [Indexed: 10/27/2022]
Abstract
Fear conditioning is an associative learning process by which organisms learn to avoid environmental stimuli that are predictive of aversive outcomes. Fear extinction learning is a process by which avoidance of fear-conditioned stimuli is attenuated when the environmental stimuli is no longer predictive of the aversive outcome. Aberrant fear conditioning and extinction learning are key elements in the development of several anxiety disorders. The 129S1 inbred strain of mice is used as an animal model for maladaptive fear learning because this strain has been shown to generalize fear to other nonaversive stimuli and is less capable of extinguishing fear responses relative to other mouse strains, such as the C57BL/6. Here we report new environmental manipulations that enhance fear and extinction learning, including the ability to discriminate between an aversively paired tone and a neutral tone, in both the 129S1 and C57BL/6 strains of mice. Specifically, we show that discontinuous ("pipped") tone stimuli significantly enhance within-session extinction learning and the discrimination between neutral and aversively paired stimuli in both strains. Furthermore, we find that extinction training in novel contexts significantly enhances the consolidation and recall of extinction learning for both strains. Cumulatively, these results underscore how environmental changes can be leveraged to ameliorate maladaptive learning in animal models and may advance cognitive and behavioral therapeutic strategies.
Collapse
Affiliation(s)
- Victor A Cazares
- Department of Molecular and Integrative Physiology and Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| | - Genesis Rodriguez
- Department of Molecular and Integrative Physiology and Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| | - Rachel Parent
- Department of Molecular and Integrative Physiology and Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| | - Lara Ouillette
- Department of Molecular and Integrative Physiology and Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| | | | - Shannon J Moore
- Department of Molecular and Integrative Physiology and Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| | - Geoffrey G Murphy
- Department of Molecular and Integrative Physiology and Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
25
|
O’Connor RM, McCafferty CP, Bravo JA, Singewald N, Holmes A, Cryan JF. Increased amygdalar metabotropic glutamate receptor 7 mRNA in a genetic mouse model of impaired fear extinction. Psychopharmacology (Berl) 2019; 236:265-272. [PMID: 30215216 PMCID: PMC6739849 DOI: 10.1007/s00213-018-5031-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/05/2018] [Indexed: 12/18/2022]
Abstract
RATIONALE Post-traumatic stress disorder (PTSD) is a devastating anxiety-related disorder which develops subsequent to a severe psychologically traumatic event. Only ~ 9% of people who experience such a trauma develop PTSD. It is clear that a number of factors, including genetics, influence whether an individual will develop PTSD subsequent to a trauma. The 129S1/SvImJ (S1) inbred mouse strain displays poor fear extinction and may be useful to model this specific aspect of PTSD. The metabotropic glutamate receptor 7 (mGlu7 receptor) has previously been shown to be involved in cognitive processes and anxiety-like behaviour placing it in a key position to regulate fear extinction processes. We sought to compare mGlu7 receptor mRNA levels in the S1 strain with those in the robustly extinguishing C57BL/6J (B6) inbred strain using in situ hybridisation (ISH) in three brain regions associated with fear extinction: the amygdala, hippocampus and prefrontal cortex (PFC). RESULTS Compared to the B6 strain, S1 mice had increased mGlu7 receptor mRNA levels in the lateral amygdala (LA) and basolateral amygdala (BLA) subdivisions. An increase was also seen in the hippocampal CA1 and CA3 subregions of S1 mice. No difference in mGlu7 receptor levels were seen in the central nucleus (CeA) of the amygdala, dentate gyrus (DG) of the hippocampus or prefrontal cortex. CONCLUSIONS These data show altered mGlu7 receptor expression in key brain regions associated with fear extinction in two different inbred mouse strains which differ markedly in their fear extinction behaviour. Altered mGlu7 receptor levels may contribute to the deficit fear extinction processes seen in fear extinction in the S1 strain.
Collapse
Affiliation(s)
- Richard M. O’Connor
- Department of Anatomy and Neuroscience and APC Microbiome Institute, University College Cork, Cork, Ireland,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, S10-20 Hess CSM, 1470 Madison Avenue, New York, NY 10029, USA,Present address: Department of Neuroscience, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Cian P. McCafferty
- Department of Anatomy and Neuroscience and APC Microbiome Institute, University College Cork, Cork, Ireland,Present address: Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Javier A. Bravo
- Grupo de NeuroGastroBioquímica, Laboratorio e Química Biológica & Bioquímica de Sistemas, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA
| | - John F. Cryan
- Department of Anatomy and Neuroscience and APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
26
|
Abstract
The measurement of Pavlovian forms of fear extinction offers a relatively simple behavioral preparation that is nonetheless tractable, from a translational perspective, as an approach to study mechanisms of exposure therapy and biological underpinnings of anxiety and trauma-related disorders such as post-traumatic stress disorder (PTSD). Deficient fear extinction is considered a robust clinical endophenotype for these disorders and, as such, has particular significance in the current "age of RDoC (research domain criteria)." Various rodent models of impaired extinction have thus been generated with the objective of approximating this clinical, relapse prone aberrant extinction learning. These models have helped to reveal neurobiological correlates of extinction circuitry failure, gene variants, and other mechanisms underlying deficient fear extinction. In addition, they are increasingly serving as tools to investigate ways to therapeutically overcome poor extinction to support long-term retention of extinction memory and thus protection against various forms of fear relapse; modeled in the laboratory by measuring spontaneous recovery, reinstatement and renewal of fear. In the current article, we review models of impaired extinction built around (1) experimentally induced brain region and neural circuit disruptions (2) spontaneously-arising and laboratory-induced genetic modifications, or (3) exposure to environmental insults, including stress, drugs of abuse, and unhealthy diet. Collectively, these models have been instrumental in advancing in our understanding of extinction failure and underlying susceptibilities at the neural, genetic, molecular, and neurochemical levels; generating renewed interest in developing novel, targeted and effective therapeutic treatments for anxiety and trauma-related disorders.
Collapse
Affiliation(s)
- Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD USA
| |
Collapse
|
27
|
Baker JD, Ozsan I, Rodriguez Ospina S, Gulick D, Blair LJ. Hsp90 Heterocomplexes Regulate Steroid Hormone Receptors: From Stress Response to Psychiatric Disease. Int J Mol Sci 2018; 20:ijms20010079. [PMID: 30585227 PMCID: PMC6337637 DOI: 10.3390/ijms20010079] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 01/30/2023] Open
Abstract
The hypothalamus-pituitary-adrenal (HPA) axis directly controls the stress response. Dysregulation of this neuroendocrine system is a common feature among psychiatric disorders. Steroid hormone receptors, like glucocorticoid receptor (GR), function as transcription factors of a diverse set of genes upon activation. This activity is regulated by molecular chaperone heterocomplexes. Much is known about the structure and function of these GR/heterocomplexes. There is strong evidence suggesting altered regulation of steroid receptor hormones by chaperones, particularly the 51 kDa FK506-binding protein (FKBP51), may work with environmental factors to increase susceptibility to various psychiatric illnesses including post-traumatic stress disorder (PTSD), major depressive disorder (MDD), and anxiety. This review highlights the regulation of steroid receptor dynamics by the 90kDa heat shock protein (Hsp90)/cochaperone heterocomplexes with an in depth look at how the structural regulation and imbalances in cochaperones can cause functional effects on GR activity. Links between the stress response and circadian systems and the development of novel chaperone-targeting therapeutics are also discussed.
Collapse
Affiliation(s)
- Jeremy D Baker
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Ilayda Ozsan
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Santiago Rodriguez Ospina
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Danielle Gulick
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Laura J Blair
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| |
Collapse
|
28
|
Lowery-Gionta EG, Crowley NA, Bukalo O, Silverstein S, Holmes A, Kash TL. Chronic stress dysregulates amygdalar output to the prefrontal cortex. Neuropharmacology 2018; 139:68-75. [PMID: 29959957 PMCID: PMC6067970 DOI: 10.1016/j.neuropharm.2018.06.032] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/20/2018] [Accepted: 06/24/2018] [Indexed: 11/19/2022]
Abstract
Chronic stress contributes to the neuropathology of mental health disorders, including those associated with anxiety. The basolateral amygdala (BLA) coordinates emotional behavioral responses through glutamatergic outputs to downstream regions such as the prefrontal cortex (PFC), nucleus accumbens core (NAcc) and bed nucleus of the stria terminalis (BNST). We explored the effects of chronic stress on BLA outputs to the PFC, NAcc and BNST using slice electrophysiology combined with optogenetics in two inbred mouse strains with distinct stress-induced anxiety responses. We found that ten consecutive days of chronic restraint stress enhanced pre-synaptic glutamate release at BLA-to-PFC synapses in C57BL/6J mice, but reduced pre-synaptic glutamate release at these synapses in DBA/2J mice. To assess the behavioral relevance of enhanced glutamate output at BLA-to-PFC synapses, we approximated the effects of chronic stress on the BLA-PFC circuit using optogenetics. We found that photostimulation of the BLA-PFC circuit in unstressed C57BL/6J mice produced persistent (i.e., post-stimulation) increased anxiety-like behavior and hyperactivity in the elevated plus-maze - a profile consistent with prototypical behavioral responses of stressed C57BL/6J mice. These data demonstrate that chronic stress dysregulates the BLA-PFC circuit by altering pre-synaptic glutamate release from BLA outputs, and provide a mechanism by which chronic stress can lead to increased anxiety.
Collapse
Affiliation(s)
- Emily G Lowery-Gionta
- Department of Pharmacology, University of North Carolina at Chapel Hill, Thurston Bowles Building 104 Manning Drive, Chapel Hill, NC, 27599, USA
| | - Nicole A Crowley
- Department of Pharmacology, University of North Carolina at Chapel Hill, Thurston Bowles Building 104 Manning Drive, Chapel Hill, NC, 27599, USA
| | - Olena Bukalo
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, 5625 Fishers Lane Rockville, MD, 20852-9411, USA
| | - Shana Silverstein
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, 5625 Fishers Lane Rockville, MD, 20852-9411, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, 5625 Fishers Lane Rockville, MD, 20852-9411, USA
| | - Thomas Louis Kash
- Department of Pharmacology, University of North Carolina at Chapel Hill, Thurston Bowles Building 104 Manning Drive, Chapel Hill, NC, 27599, USA.
| |
Collapse
|