1001
|
Goto T, Kubota Y, Toyoda A. Plasma and Liver Metabolic Profiles in Mice Subjected to Subchronic and Mild Social Defeat Stress. J Proteome Res 2014; 14:1025-32. [DOI: 10.1021/pr501044k] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Tatsuhiko Goto
- College
of Agriculture, Ibaraki University, Ami, Ibaraki 300-0393, Japan
- Ibaraki University Cooperation between Agriculture and Medical Science (IUCAM), Ami, Ibaraki 300-0393, Japan
| | - Yoshifumi Kubota
- College
of Agriculture, Ibaraki University, Ami, Ibaraki 300-0393, Japan
| | - Atsushi Toyoda
- College
of Agriculture, Ibaraki University, Ami, Ibaraki 300-0393, Japan
- Ibaraki University Cooperation between Agriculture and Medical Science (IUCAM), Ami, Ibaraki 300-0393, Japan
- United
Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu-city, Tokyo 183-8509, Japan
| |
Collapse
|
1002
|
Ho L, Lange G, Zhao W, Wang J, Rooney R, Patel DH, Fobler MM, Helmer DA, Elder G, Shaughness MC, Ahlers ST, Russo SJ, Pasinetti GM. Select small nucleolar RNAs in blood components as novel biomarkers for improved identification of comorbid traumatic brain injury and post-traumatic stress disorder in veterans of the conflicts in Afghanistan and Iraq. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2014; 3:170-81. [PMID: 25628968 PMCID: PMC4299721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/11/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND The present study was designed to validate the ability of our recently identified set of small noncoding RNA candidate mild traumatic brain injury (mTBI) biomarkers to diagnose mTBI in the presence or absence of post-traumatic stress disorder (PTSD) comorbidity. Using qPCR, we explored the regulation of the candidate biomarkers in peripheral blood mononuclear cells (PBMC) from 58 veterans. RESULTS We confirmed that 4 small nucleolar RNAs (snoRNAs), ACA48, U35, U55, and U83A, are significantly down-regulated in PBMC from veterans with mTBI and PTSD compared to non-TBI, control subjects with PTSD only. We found that the snoRNA biomarkers are able to dissect subjects with comorbid mTBI and PTSD from PTSD subjects without mTBI with 100% sensitivity, 81% accuracy, and 72% specificity. No significant differential expression of snoRNA biomarkers was found in mTBI subjects without comorbid PTSD. However, we found significantly lower U55 contents in subjects with PTSD. We explored the regulation of ACA48 in rodent models of PTSD or blast-induced mTBI to gather proof-of-concept evidence that would connect the regulation of the biomarkers and the development of mTBI or PTSD. We found no change in the regulation of ACA48 in the mTBI rat model. We did, however, find significant down-regulation of ACA48 in the PTSD mouse model 24 hours following psychological trauma exposure. This may reflect a short-term response to trauma exposure, since we found no change in the regulation of ACA48 in veteran PTSD subjects 3.6 years post-deployment. CONCLUSIONS Additional application of the 4 snoRNA biomarker to current diagnostic criteria may provide an objective biomarker pattern to help identify veterans with comorbid mTBI and PTSD. Our observations suggest that biological interactions between TBI and PTSD may contribute to the clinical features of veterans with comorbid mTBI and PTSD. Future investigations on mTBI mechanisms or TBI biomarkers should consider their interactions with PTSD.
Collapse
Affiliation(s)
- Lap Ho
- Department of Neurology, Icahn School of Medicine at Mount SinaiNew York, USA
| | - Gudrun Lange
- War-Related Illness and Injury Study Center, VA-New Jersey Healthcare SystemEast Orange, NJ, USA
| | - Wei Zhao
- Department of Neurology, Icahn School of Medicine at Mount SinaiNew York, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical CenterBronx, New York, USA
| | - Jun Wang
- Department of Neurology, Icahn School of Medicine at Mount SinaiNew York, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical CenterBronx, New York, USA
| | | | | | - Malusha M Fobler
- War-Related Illness and Injury Study Center, VA-New Jersey Healthcare SystemEast Orange, NJ, USA
| | - Drew A Helmer
- War-Related Illness and Injury Study Center, VA-New Jersey Healthcare SystemEast Orange, NJ, USA
| | - Gregory Elder
- Neurology Service, James J. Peters Department of Veterans Affairs Medical CenterBronx, New York, USA
| | - Michael C Shaughness
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research CenterSilver Spring, MD, USA
| | - Stephen T Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research CenterSilver Spring, MD, USA
| | - Scott J Russo
- Department of Neuroscience, Icahn School of Medicine at Mount SinaiNew York, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount SinaiNew York, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical CenterBronx, New York, USA
- Geriatric Research and Clinical Center, James J. Peter Veterans Affairs Medical CenterBronx, NY, USA
| |
Collapse
|
1003
|
Abstract
Witnessing a traumatic event but not directly experiencing it can be psychologically quite damaging. In North America alone, ∼30% of individuals who witness a traumatic event develop post-traumatic stress disorder (PTSD). While effects of direct trauma are evident, consequences of indirect or secondary trauma are often ignored. Also unclear is the role of social support in the consequences of these experiences. The social defeat paradigm, which involves aggressive encounters by a large Long-Evans male rat (resident) towards a smaller Sprague-Dawley male rat (intruder), is considered a rodent model of PTSD. We have modified this model to create a trauma witness model (TWM) and have used our TWM model to also evaluate social support effects. Basically, when an intruder rat is placed into the home cage of a resident rat, it encounters an agonistic behavior resulting in intruder subordination. The socially defeated intruder is designated the SD rat. A second rat, the cage mate of the SD, is positioned to witness the event and is the trauma witnessing (TW) rat. Experiments were performed in two different experimental conditions. In one, the SD and TW rats were cagemates and acclimatized together. Then, one SD rat was subjected to three sessions of social defeat for 7 d. TW rat witnessed these events. After each social defeat exposure, the TW and SD rats were housed together. In the second, the TW and SD rats were housed separately starting after the first defeat. At the end of each protocol, depression-anxiety-like behavior and memory tests were conducted on the SD and TW rats, blood withdrawn and specific organs collected. Witnessing traumatic events led to depression- and anxiety-like behavior and produced memory deficits in TW rats associated with elevated corticosterone levels.
Collapse
|
1004
|
Banerjee A, Engineer CT, Sauls BL, Morales AA, Kilgard MP, Ploski JE. Abnormal emotional learning in a rat model of autism exposed to valproic acid in utero. Front Behav Neurosci 2014; 8:387. [PMID: 25429264 PMCID: PMC4228846 DOI: 10.3389/fnbeh.2014.00387] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/17/2014] [Indexed: 01/30/2023] Open
Abstract
Autism Spectrum Disorders (ASD) are complex neurodevelopmental disorders characterized by repetitive behavior and impaired social communication and interactions. Apart from these core symptoms, a significant number of ASD individuals display higher levels of anxiety and some ASD individuals exhibit impaired emotional learning. We therefore sought to further examine anxiety and emotional learning in an environmentally induced animal model of ASD that utilizes the administration of the known teratogen, valproic acid (VPA) during gestation. Specifically we exposed dams to one of two different doses of VPA (500 and 600 mg/kg) or vehicle on day 12.5 of gestation and examined the resultant progeny. Our data indicate that animals exposed to VPA in utero exhibit enhanced anxiety in the open field test and normal object recognition memory compared to control animals. Animals exposed to 500 mg/kg of VPA displayed normal acquisition of auditory fear conditioning, and exhibited reduced extinction of fear memory and normal litter survival rates as compared to control animals. We observed that animals exposed to 600 mg/kg of VPA exhibited a significant reduction in the acquisition of fear conditioning, a significant reduction in social interaction and a significant reduction in litter survival rates as compared to control animals. VPA (600 mg/kg) exposed animals exhibited similar shock sensitivity and hearing as compared to control animals indicating the fear conditioning deficit observed in these animals was not likely due to sensory deficits, but rather due to deficits in learning or memory retrieval. In conclusion, considering that progeny from dams exposed to rather similar doses of VPA exhibit striking differences in emotional learning, the VPA model may serve as a useful tool to explore the molecular and cellular mechanisms that contribute to not only ASD, but also emotional learning.
Collapse
Affiliation(s)
- Anwesha Banerjee
- School of Behavioral and Brain Sciences, University of Texas at Dallas Richardson, TX, USA
| | - Crystal T Engineer
- School of Behavioral and Brain Sciences, University of Texas at Dallas Richardson, TX, USA
| | - Bethany L Sauls
- School of Behavioral and Brain Sciences, University of Texas at Dallas Richardson, TX, USA
| | - Anna A Morales
- School of Behavioral and Brain Sciences, University of Texas at Dallas Richardson, TX, USA
| | - Michael P Kilgard
- School of Behavioral and Brain Sciences, University of Texas at Dallas Richardson, TX, USA
| | - Jonathan E Ploski
- School of Behavioral and Brain Sciences, University of Texas at Dallas Richardson, TX, USA
| |
Collapse
|
1005
|
Affiliation(s)
- Gerhard Schratt
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, 35032 Marburg, Germany
| |
Collapse
|
1006
|
Hodes GE, Pfau ML, Leboeuf M, Golden SA, Christoffel DJ, Bregman D, Rebusi N, Heshmati M, Aleyasin H, Warren BL, Lebonté B, Horn S, Lapidus KA, Stelzhammer V, Wong EHF, Bahn S, Krishnan V, Bolaños-Guzman CA, Murrough JW, Merad M, Russo SJ. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc Natl Acad Sci U S A 2014. [PMID: 25331895 DOI: 10.1073/pnas.1415191111/-/dcsupplemental] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Depression and anxiety disorders are associated with increased release of peripheral cytokines; however, their functional relevance remains unknown. Using a social stress model in mice, we find preexisting individual differences in the sensitivity of the peripheral immune system that predict and promote vulnerability to social stress. Cytokine profiles were obtained 20 min after the first social stress exposure. Of the cytokines regulated by stress, IL-6 was most highly up-regulated only in mice that ultimately developed a susceptible behavioral phenotype following a subsequent chronic stress, and levels remained elevated for at least 1 mo. We confirmed a similar elevation of serum IL-6 in two separate cohorts of patients with treatment-resistant major depressive disorder. Before any physical contact in mice, we observed individual differences in IL-6 levels from ex vivo stimulated leukocytes that predict susceptibility versus resilience to a subsequent stressor. To shift the sensitivity of the peripheral immune system to a pro- or antidepressant state, bone marrow (BM) chimeras were generated by transplanting hematopoietic progenitor cells from stress-susceptible mice releasing high IL-6 or from IL-6 knockout (IL-6(-/-)) mice. Stress-susceptible BM chimeras exhibited increased social avoidance behavior after exposure to either subthreshold repeated social defeat stress (RSDS) or a purely emotional stressor termed witness defeat. IL-6(-/-) BM chimeric and IL-6(-/-) mice, as well as those treated with a systemic IL-6 monoclonal antibody, were resilient to social stress. These data establish that preexisting differences in stress-responsive IL-6 release from BM-derived leukocytes functionally contribute to social stress-induced behavioral abnormalities.
Collapse
Affiliation(s)
- Georgia E Hodes
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Madeline L Pfau
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Marylene Leboeuf
- Departments of Gene and Cell Medicine, Oncological Sciences, and
| | - Sam A Golden
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Daniel J Christoffel
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Dana Bregman
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Nicole Rebusi
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Mitra Heshmati
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Hossein Aleyasin
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Brandon L Warren
- Department of Psychology, Florida State University, Tallahassee, FL 32306-4301
| | - Benoit Lebonté
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sarah Horn
- Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Kyle A Lapidus
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Viktoria Stelzhammer
- Institute of Biotechnology, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 1QT, United Kingdom; Department of Neuroscience, Erasmus Medical Center, NL 3000 CA Rotterdam, The Netherlands
| | - Erik H F Wong
- CNS-Pain Innovative Medicine Unit, External Science, AstraZeneca Pharmaceuticals, Wilmington, DE 19850; and
| | - Sabine Bahn
- Institute of Biotechnology, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 1QT, United Kingdom; Department of Neuroscience, Erasmus Medical Center, NL 3000 CA Rotterdam, The Netherlands
| | - Vaishnav Krishnan
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | | | - James W Murrough
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Miriam Merad
- Departments of Gene and Cell Medicine, Oncological Sciences, and
| | - Scott J Russo
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029;
| |
Collapse
|
1007
|
Gassen NC, Hartmann J, Zschocke J, Stepan J, Hafner K, Zellner A, Kirmeier T, Kollmannsberger L, Wagner KV, Dedic N, Balsevich G, Deussing JM, Kloiber S, Lucae S, Holsboer F, Eder M, Uhr M, Ising M, Schmidt MV, Rein T. Association of FKBP51 with priming of autophagy pathways and mediation of antidepressant treatment response: evidence in cells, mice, and humans. PLoS Med 2014; 11:e1001755. [PMID: 25386878 PMCID: PMC4227651 DOI: 10.1371/journal.pmed.1001755] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 09/30/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND FK506 binding protein 51 (FKBP51) is an Hsp90 co-chaperone and regulator of the glucocorticoid receptor, and consequently of stress physiology. Clinical studies suggest a genetic link between FKBP51 and antidepressant response in mood disorders; however, the underlying mechanisms remain elusive. The objective of this study was to elucidate the role of FKBP51 in the actions of antidepressants, with a particular focus on pathways of autophagy. METHODS AND FINDINGS Established cell lines, primary neural cells, human blood cells of healthy individuals and patients with depression, and mice were treated with antidepressants. Mice were tested for several neuroendocrine and behavioral parameters. Protein interactions and autophagic pathway activity were mainly evaluated by co-immunoprecipitation and Western blots. We first show that the effects of acute antidepressant treatment on behavior are abolished in FKBP51 knockout (51KO) mice. Autophagic markers, such as the autophagy initiator Beclin1, were increased following acute antidepressant treatment in brains from wild-type, but not 51KO, animals. FKBP51 binds to Beclin1, changes decisive protein interactions and phosphorylation of Beclin1, and triggers autophagic pathways. Antidepressants and FKBP51 exhibited synergistic effects on these pathways. Using chronic social defeat as a depression-relevant stress model in combination with chronic paroxetine (PAR) treatment revealed that the stress response, as well as the effects of antidepressants on behavior and autophagic markers, depends on FKBP51. In human blood cells of healthy individuals, FKBP51 levels correlated with the potential of antidepressants to induce autophagic pathways. Importantly, the clinical antidepressant response of patients with depression (n = 51) could be predicted by the antidepressant response of autophagic markers in patient-derived peripheral blood lymphocytes cultivated and treated ex vivo (Beclin1/amitriptyline: r = 0.572, p = 0.003; Beclin1/PAR: r = 0.569, p = 0.004; Beclin1/fluoxetine: r = 0.454, p = 0.026; pAkt/amitriptyline: r = -0.416, p = 0.006; pAkt/PAR: r = -0.355, p = 0.021; LC3B-II/PAR: r = 0.453, p = 0.02), as well as by the lymphocytic expression levels of FKBP51 (r = 0.631, p<0.0001), pAkt (r = -0.515, p = 0.003), and Beclin1 (r = 0.521, p = 0.002) at admission. Limitations of the study include the use of male mice only and the relatively low number of patients for protein analyses. CONCLUSIONS To our knowledge, these findings provide the first evidence for the molecular mechanism of FKBP51 in priming autophagic pathways; this process is linked to the potency of at least some antidepressants. These newly discovered functions of FKBP51 also provide novel predictive markers for treatment outcome, consistent with physiological and potential clinical relevance. Please see later in the article for the Editors' Summary.
Collapse
Affiliation(s)
- Nils C. Gassen
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- * E-mail: (NCG); (TR)
| | - Jakob Hartmann
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jürgen Zschocke
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jens Stepan
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Andreas Zellner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Thomas Kirmeier
- Department of Clinical Research, Max Planck Institute of Psychiatry, Munich, Germany
| | - Lorenz Kollmannsberger
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Klaus V. Wagner
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Nina Dedic
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Georgia Balsevich
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M. Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Stefan Kloiber
- Department of Clinical Research, Max Planck Institute of Psychiatry, Munich, Germany
| | - Susanne Lucae
- Department of Clinical Research, Max Planck Institute of Psychiatry, Munich, Germany
| | - Florian Holsboer
- Department of Clinical Research, Max Planck Institute of Psychiatry, Munich, Germany
| | - Matthias Eder
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Manfred Uhr
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Marcus Ising
- Department of Clinical Research, Max Planck Institute of Psychiatry, Munich, Germany
| | - Mathias V. Schmidt
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Theo Rein
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- * E-mail: (NCG); (TR)
| |
Collapse
|
1008
|
Abstract
Repeated aggression is a frequent symptom of many psychiatric and neurological disorders, including obsessive-compulsive and attention deficit hyperactivity disorders, bipolar and post-traumatic stress disorders, epilepsy, autism, schizophrenia and drug abuse. However, repeated aggression is insufficiently studied because there is a lack of adequate models in animals. The sensory contact model (SCM), widely used to study the effects of chronic social defeat stress, can also be used to investigate the effects of repeated aggression. Mice with repeated positive fighting experience in daily agonistic interactions in this model develop pronounced aggressiveness, anxiety and impulsivity, disturbances in motivated and cognitive behaviors, and impairments of sociability; they also demonstrate hyperactivity, attention-deficit behavior, motor dysfunctions and repetitive stereotyped behaviors, such as jerks, rotations and head twitches. In this protocol, we describe how to apply the SCM to study repeated aggression in mice. Severe neuropathology develops in male mice after 20-21 d of agonistic interactions.
Collapse
|
1009
|
Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc Natl Acad Sci U S A 2014; 111:16136-41. [PMID: 25331895 DOI: 10.1073/pnas.1415191111] [Citation(s) in RCA: 498] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Depression and anxiety disorders are associated with increased release of peripheral cytokines; however, their functional relevance remains unknown. Using a social stress model in mice, we find preexisting individual differences in the sensitivity of the peripheral immune system that predict and promote vulnerability to social stress. Cytokine profiles were obtained 20 min after the first social stress exposure. Of the cytokines regulated by stress, IL-6 was most highly up-regulated only in mice that ultimately developed a susceptible behavioral phenotype following a subsequent chronic stress, and levels remained elevated for at least 1 mo. We confirmed a similar elevation of serum IL-6 in two separate cohorts of patients with treatment-resistant major depressive disorder. Before any physical contact in mice, we observed individual differences in IL-6 levels from ex vivo stimulated leukocytes that predict susceptibility versus resilience to a subsequent stressor. To shift the sensitivity of the peripheral immune system to a pro- or antidepressant state, bone marrow (BM) chimeras were generated by transplanting hematopoietic progenitor cells from stress-susceptible mice releasing high IL-6 or from IL-6 knockout (IL-6(-/-)) mice. Stress-susceptible BM chimeras exhibited increased social avoidance behavior after exposure to either subthreshold repeated social defeat stress (RSDS) or a purely emotional stressor termed witness defeat. IL-6(-/-) BM chimeric and IL-6(-/-) mice, as well as those treated with a systemic IL-6 monoclonal antibody, were resilient to social stress. These data establish that preexisting differences in stress-responsive IL-6 release from BM-derived leukocytes functionally contribute to social stress-induced behavioral abnormalities.
Collapse
|
1010
|
Liu G, Rustom N, Litteljohn D, Bobyn J, Rudyk C, Anisman H, Hayley S. Use of induced pluripotent stem cell derived neurons engineered to express BDNF for modulation of stressor related pathology. Front Cell Neurosci 2014; 8:316. [PMID: 25352778 PMCID: PMC4196567 DOI: 10.3389/fncel.2014.00316] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/20/2014] [Indexed: 01/18/2023] Open
Abstract
Combined cell and gene-based therapeutic strategies offer potential in the treatment of neurodegenerative and psychiatric conditions that have been associated with structural brain disturbances. In the present investigation, we used a novel virus-free re-programming method to generate induced pluripotent stem cells (iPSCs), and then subsequently transformed these cells into neural cells which over-expressed brain derived neurotrophic factor (BDNF). Importantly, the infusion of iPSC derived neural cells (as a cell replacement and gene delivery tool) and BDNF (as a protective factor) influenced neuronal outcomes. Specifically, intracerebroventricular transplantation of iPSC-derived neural progenitors that over-expressed BDNF reversed the impact of immune (lipopolysaccharide) and chronic stressor challenges upon subventricular zone adult neurogenesis, and the iPSC-derived neural progenitor cells alone blunted the stressor-induced corticosterone response. Moreover, our findings indicate that mature dopamine producing neurons can be generated using iPSC procedures and appear to be viable when infused in vivo. Taken together, these data could have important implications for using gene-plus-cell replacement methods to modulate stressor related pathology.
Collapse
Affiliation(s)
- Gele Liu
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Nazneen Rustom
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Darcy Litteljohn
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Jessica Bobyn
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Chris Rudyk
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Hymie Anisman
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Shawn Hayley
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| |
Collapse
|
1011
|
Donahue RJ, Muschamp JW, Russo SJ, Nestler EJ, Carlezon WA. Effects of striatal ΔFosB overexpression and ketamine on social defeat stress-induced anhedonia in mice. Biol Psychiatry 2014; 76:550-8. [PMID: 24495460 PMCID: PMC4087093 DOI: 10.1016/j.biopsych.2013.12.014] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 12/19/2013] [Accepted: 12/30/2013] [Indexed: 12/27/2022]
Abstract
BACKGROUND Chronic social defeat stress (CSDS) produces persistent behavioral adaptations in mice. In many behavioral assays, it can be difficult to determine if these adaptations reflect core signs of depression. We designed studies to characterize the effects of CSDS on sensitivity to reward because anhedonia (reduced sensitivity to reward) is a defining characteristic of depressive disorders in humans. We also examined the effects of striatal ΔFosB overexpression and the N-methyl-D-aspartate receptor antagonist ketamine, both of which promote resilience, on CSDS-induced alterations in reward function and social interaction. METHODS Intracranial self-stimulation (ICSS) was used to quantify CSDS-induced changes in reward function. Mice were implanted with lateral hypothalamic electrodes, and ICSS thresholds were measured after each of 10 daily CSDS sessions and during a 5-day recovery period. We also examined if acute intraperitoneal administration of ketamine (2.5-20 mg/kg) reverses CSDS-induced effects on reward or, in separate mice, social interaction. RESULTS ICSS thresholds were increased by CSDS, indicating decreases in the rewarding impact of lateral hypothalamic stimulation (anhedonia). This effect was attenuated in mice overexpressing ∆FosB in striatum, consistent with pro-resilient actions of this transcription factor. High, but not low, doses of ketamine administered after completion of the CSDS regimen attenuated social avoidance in defeated mice, although this effect was transient. Ketamine did not block CSDS-induced anhedonia in the ICSS test. CONCLUSIONS This study found that CSDS triggers persistent anhedonia and confirms that ΔFosB overexpression produces stress resilience. The findings of this study also indicate that acute administration of ketamine fails to attenuate CSDS-induced anhedonia despite reducing other depression-related behavioral abnormalities.
Collapse
Affiliation(s)
- Rachel J. Donahue
- Behavioral Genetics Laboratory, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - John W. Muschamp
- Behavioral Genetics Laboratory, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Scott J. Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Eric J. Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - William A. Carlezon
- Behavioral Genetics Laboratory, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| |
Collapse
|
1012
|
The stress-inducible actin-interacting protein DRR1 shapes social behavior. Psychoneuroendocrinology 2014; 48:98-110. [PMID: 24998413 DOI: 10.1016/j.psyneuen.2014.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 01/18/2023]
Abstract
Understanding the molecular mechanisms by which stress is translated into changes in complex behavior may help to identify novel treatment strategies for stress-associated psychiatric disorders. The tumor suppressor gene down-regulated in renal cell carcinoma 1 (DRR1) was recently characterized as a new molecular link between stress, synaptic efficacy and behavioral performance, most likely through its ability to modulate actin dynamics. The lateral septum is one of the brain regions prominently involved in the stress response. This brain region features high DRR1 expression in adult mice, even under basal conditions. We therefore aimed to characterize and dissect the functional role of septal DRR1 in modulating complex behavior. DRR1 protein expression was shown to be expressed in both neurons and astrocytes of the lateral septum of adult mice. Septal DRR1 mRNA expression increased after acute defeat stress and glucocorticoid receptor activation. To mimic the stress-induced DRR1 increase in the lateral septum of mice, we performed adenovirus-mediated region-specific overexpression of DRR1 and characterized the behavior of these mice. Overexpression of DRR1 in the septal region increased sociability, but did not change cognitive, anxiety-like or anhedonic behavior. The observed changes in social behavior did not involve alterations of the expression of vasopressin or oxytocin receptors, the canonical social neuropeptidergic circuits of the lateral septum. In summary, our data suggest that the stress-induced increase of DRR1 expression in the lateral septum could be a protective mechanism to buffer or counterbalance negative consequences of stress exposure on social behavior.
Collapse
|
1013
|
Illuminating circuitry relevant to psychiatric disorders with optogenetics. Curr Opin Neurobiol 2014; 30:9-16. [PMID: 25215625 DOI: 10.1016/j.conb.2014.08.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 08/22/2014] [Indexed: 12/11/2022]
Abstract
The brain's remarkable capacity to generate cognition and behavior is mediated by an extraordinarily complex set of neural interactions that remain largely mysterious. This complexity poses a significant challenge in developing therapeutic interventions to ameliorate psychiatric disease. Accordingly, few new classes of drugs have been made available for patients with mental illness since the 1950s. Optogenetics offers the ability to selectively manipulate individual neural circuit elements that underlie disease-relevant behaviors and is currently accelerating the pace of preclinical research into neurobiological mechanisms of disease. In this review, we highlight recent findings from studies that employ optogenetic approaches to gain insight into normal and aberrant brain function relevant to mental illness. Emerging data from these efforts offers an exquisitely detailed picture of disease-relevant neural circuits in action, and hints at the potential of optogenetics to open up entirely new avenues in the treatment of psychiatric disorders.
Collapse
|
1014
|
Disrupted MEK/ERK signaling in the medial orbital cortex and dorsal endopiriform nuclei of the prefrontal cortex in a chronic restraint stress mouse model of depression. Neurosci Lett 2014; 580:163-8. [DOI: 10.1016/j.neulet.2014.08.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 07/24/2014] [Accepted: 08/01/2014] [Indexed: 01/15/2023]
|
1015
|
Kirshenbaum GS, Lieberman SR, Briner TJ, Leonardo ED, Dranovsky A. Adolescent but not adult-born neurons are critical for susceptibility to chronic social defeat. Front Behav Neurosci 2014; 8:289. [PMID: 25221485 PMCID: PMC4147831 DOI: 10.3389/fnbeh.2014.00289] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/07/2014] [Indexed: 11/26/2022] Open
Abstract
Recent evidence implicates adult hippocampal neurogenesis in regulating behavioral and physiologic responses to stress. Hippocampal neurogenesis occurs across the lifespan, however the rate of cell birth is up to 300% higher in adolescent mice compared to adults. Adolescence is a sensitive period in development where emotional circuitry and stress reactivity undergo plasticity establishing life-long set points. Therefore neurogenesis occurring during adolescence may be particularly important for emotional behavior. However, little is known about the function of hippocampal neurons born during adolescence. In order to assess the contribution of neurons born in adolescence to the adult stress response and depression-related behavior, we transiently reduced cell proliferation either during adolescence, or during adulthood in GFAP-Tk mice. We found that the intervention in adolescence did not change adult baseline behavioral response in the forced swim test, sucrose preference test or social affiliation test, and did not change adult corticosterone responses to an acute stressor. However following chronic social defeat, adult mice with reduced adolescent neurogenesis showed a resilient phenotype. A similar transient reduction in adult neurogenesis did not affect depression-like behaviors or stress induced corticosterone. Our study demonstrates that hippocampal neurons born during adolescence, but not in adulthood are important to confer susceptibility to chronic social defeat.
Collapse
Affiliation(s)
- Greer S Kirshenbaum
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| | - Sophie R Lieberman
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| | - Tamara J Briner
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| | - E David Leonardo
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| | - Alex Dranovsky
- Dranovsky-Leonardo Lab, Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, Columbia University New York, NY, USA
| |
Collapse
|
1016
|
Kulesskaya N, Karpova NN, Ma L, Tian L, Voikar V. Mixed housing with DBA/2 mice induces stress in C57BL/6 mice: implications for interventions based on social enrichment. Front Behav Neurosci 2014; 8:257. [PMID: 25147512 PMCID: PMC4123727 DOI: 10.3389/fnbeh.2014.00257] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 07/13/2014] [Indexed: 12/11/2022] Open
Abstract
Several behavioral interventions, based on social enrichment and observational learning are applied in treatment of neuropsychiatric disorders. However, the mechanism of such modulatory effect and the safety of applied methods on individuals involved in social support need further investigation. We took advantage of known differences between inbred mouse strains to reveal the effect of social enrichment on behavior and neurobiology of animals with different behavioral phenotypes. C57BL/6 and DBA/2 female mice displaying multiple differences in cognitive, social, and emotional behavior were group-housed either in same-strain or in mixed-strain conditions. Comprehensive behavioral phenotyping and analysis of expression of several plasticity- and stress-related genes were done to measure the reciprocal effects of social interaction between the strains. Contrary to our expectation, mixed housing did not change the behavior of DBA/2 mice. Nevertheless, the level of serum corticosterone and the expression of glucocorticoid receptor Nr3c1 in the brain were increased in mixed housed DBA/2 as compared with those of separately housed DBA/2 mice. In contrast, socially active C57BL/6 animals were more sensitive to the mixed housing, displaying several signs of stress: alterations in learning, social, and anxiety-like behavior and anhedonia. These behavioral impairments were accompanied by the elevated serum corticosterone and the reduced expression of Nr3c1, as well as the elevated Bdnf levels in the cortex and hippocampus. Our results demonstrate the importance of social factors in modulation of both behavior and the underlying neurobiological mechanisms in stress response, and draw attention to the potential negative impact of social interventions for individuals involved in social support.
Collapse
Affiliation(s)
| | - Nina N Karpova
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Li Ma
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Li Tian
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Vootele Voikar
- Neuroscience Center, University of Helsinki Helsinki, Finland
| |
Collapse
|
1017
|
Antidepressant-like effects of cortical deep brain stimulation coincide with pro-neuroplastic adaptations of serotonin systems. Biol Psychiatry 2014; 76:203-12. [PMID: 24503468 PMCID: PMC4072754 DOI: 10.1016/j.biopsych.2013.12.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 11/27/2013] [Accepted: 12/16/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cortical deep brain stimulation (DBS) is a promising therapeutic option for treatment-refractory depression, but its mode of action remains enigmatic. Serotonin (5-HT) systems are engaged indirectly by ventromedial prefrontal cortex (vmPFC) DBS. Resulting neuroplastic changes in 5-HT systems could thus coincide with the long-term therapeutic activity of vmPFC DBS. METHODS We tested this hypothesis by evaluating the antidepressant-like activity of vmPFC DBS in the chronic social defeat stress (CSDS) model of depression (n = 8-13 mice/group). Circuit-wide activation induced by vmPFC DBS was mapped with c-Fos immunolabeling. The effects of chronic vmPFC DBS on the physiology and morphology of genetically identified 5-HT cells from the dorsal raphe nucleus (DRN) were examined with whole-cell recording, somatodendritic three-dimensional reconstructions and morphometric analyses of presynaptic boutons along 5-HT axons. RESULTS Acute DBS drove c-Fos expression locally in the vmPFC and in several distal monosynaptically connected regions, including the DRN. Chronic DBS reversed CSDS-induced social avoidance, restored the disrupted balance of excitatory/inhibitory inputs onto 5-HT neurons, and reversed 5-HT hypoexcitability observed after CSDS. Furthermore, vmPFC DBS reversed CSDS-induced arborization of 5-HT dendrites in the DRN and increased the size and density of 5-HT presynaptic terminals in the dentate gyrus and vmPFC. CONCLUSIONS We validate a new preclinical paradigm to examine cellular mechanisms underlying the antidepressant-like activity of vmPFC DBS and identify dramatic circuit-mediated cellular adaptations that coincide with this treatment. These neuroplastic changes of 5-HT neurons might contribute to the progressive mood improvements reported in patients treated with chronic courses of cortical DBS.
Collapse
|
1018
|
Subchronic and mild social defeat stress accelerates food intake and body weight gain with polydipsia-like features in mice. Behav Brain Res 2014; 270:339-48. [DOI: 10.1016/j.bbr.2014.05.040] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/28/2014] [Accepted: 05/19/2014] [Indexed: 01/04/2023]
|
1019
|
|
1020
|
Warren BL, Sial OK, Alcantara LF, Greenwood MA, Brewer JS, Rozofsky JP, Parise EM, Bolaños-Guzmán CA. Altered gene expression and spine density in nucleus accumbens of adolescent and adult male mice exposed to emotional and physical stress. Dev Neurosci 2014; 36:250-60. [PMID: 24943326 PMCID: PMC4125435 DOI: 10.1159/000362875] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 04/14/2014] [Indexed: 12/16/2022] Open
Abstract
Stressful early life experiences are implicated in lifelong health. However, little is known about the consequences of emotional stress (ES) or physical stress (PS) on neurobiology. Therefore, the following set of experiments was designed to assess changes in transcription and translation of key proteins within the nucleus accumbens (NAc). Male adolescent (postnatal day 35) or adult (8-week-old) mice were exposed to ES or PS using a witness social defeat paradigm. Then, 24 h after the last stress session, we measured levels of specific mRNAs and proteins within the NAc. Spine density was also assessed in separate groups of mice. Exposure to ES or PS disrupted extracellular signal-related kinase 2 (ERK2), reduced transcription of ΔFosB and had no effect on cAMP response element-binding protein (CREB) mRNA. Western blots revealed that exposure to ES or PS decreased ERK2 phosphorylation in adolescents, whereas the same stress regimen increased ERK2 phosphorylation in adults. Exposure to ES or PS had no effect on ΔFosB or CREB phosphorylation. ES and PS increased spine density in the NAc of adolescent exposed mice, but only exposure to PS increased spine density in adults. Together, these findings demonstrate that exposure to ES or PS is a potent stressor in adolescent and adult mice and can disturb the integrity of the NAc by altering transcription and translation of important signaling molecules in an age-dependent manner. Furthermore, exposure to ES and PS induces substantial synaptic plasticity of the NAc.
Collapse
Affiliation(s)
- Brandon L Warren
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301
| | - Omar K. Sial
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301
| | - Lyonna F. Alcantara
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301
| | - Maria A. Greenwood
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301
| | - Jacob S. Brewer
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301
| | - John P. Rozofsky
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301
| | - Eric M. Parise
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301
| | - Carlos A. Bolaños-Guzmán
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-4301
| |
Collapse
|
1021
|
Mouse social stress induces increased fear conditioning, helplessness and fatigue to physical challenge together with markers of altered immune and dopamine function. Neuropharmacology 2014; 85:328-41. [PMID: 24907589 DOI: 10.1016/j.neuropharm.2014.05.039] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/24/2014] [Accepted: 05/26/2014] [Indexed: 12/24/2022]
Abstract
In neuropsychiatry, animal studies demonstrating causal effects of environmental manipulations relevant to human aetiology on behaviours relevant to human psychopathologies are valuable. Such valid models can improve understanding of aetio-pathophysiology and preclinical discovery and development of new treatments. In depression, specific uncontrollable stressful life events are major aetiological factors, and subsequent generalized increases in fearfulness, helplessness and fatigue are core symptoms or features. Here we exposed adult male C57BL/6 mice to 15-day psychosocial stress with loss of social control but minimal physical wounding. One cohort was assessed in a 3-day test paradigm of motor activity, fear conditioning and 2-way avoid-escape behaviour on days 16-18, and a second cohort was assessed in a treadmill fatigue paradigm on days 19 and 29, followed by the 3-day paradigm on days 30-32. All tests used a physical aversive stimulus, namely mild, brief electroshocks. Socially stressed mice displayed decreased motor activity, increased fear acquisition, decreased 2-way avoid-escape responding (increased helplessness) and increased fatigue. They also displayed increased plasma TNF and spleen hypertrophy, and adrenal hypertrophy without hyper-corticoidism. In a third cohort, psychosocial stress effects on brain gene expression were assessed using next generation sequencing. Gene expression was altered in pathways of inflammation and G-protein coupled receptors in prefrontal cortex and amygdala; in the latter, expression of genes important in dopamine function were de-regulated including down-regulated Drd2, Adora2a and Darpp-32. This model can be applied to identify targets for treating psychopathologies such as helplessness or fatigue, and to screen compounds/biologics developed to act at these targets.
Collapse
|
1022
|
Krishnan V. Defeating the fear: new insights into the neurobiology of stress susceptibility. Exp Neurol 2014; 261:412-6. [PMID: 24852100 DOI: 10.1016/j.expneurol.2014.05.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 05/08/2014] [Accepted: 05/12/2014] [Indexed: 12/17/2022]
Abstract
The psychopathological impact of emotional stress on a specific individual varies markedly: while most escape the development of post-traumatic stress disorder and/or major depression, a select group of individuals demonstrate a vulnerability to succumb to these conditions. The past decade has witnessed an explosion in animal research into the underlying neurobiological mechanisms that govern both vulnerability and resilience to such stressors. In the May 2014 issue, Chou and colleagues employ the mouse social defeat model of chronic stress to demonstrate that defeated susceptible mice display an exaggerated conditioned fear response associated with more pronounced autonomic changes. These physiological alterations were found to be mediated via local increases in the levels of brain derived neurotrophic factor (BDNF) within the basolateral amygdala and could be inhibited by the systemic administration of a beta adrenergic antagonist. This mini-review critically examines this manuscript's new mechanistic insights in light of previous results employing similar approaches. The strengths and limitations of the social defeat model, as well as the relevance of these findings to neurologic illness are discussed briefly.
Collapse
Affiliation(s)
- Vaishnav Krishnan
- Department of Neurology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
1023
|
Iñiguez SD, Riggs LM, Nieto SJ, Dayrit G, Zamora NN, Shawhan KL, Cruz B, Warren BL. Social defeat stress induces a depression-like phenotype in adolescent male c57BL/6 mice. Stress 2014; 17:247-55. [PMID: 24689732 PMCID: PMC5534169 DOI: 10.3109/10253890.2014.910650] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Abstract Exposure to stress is highly correlated with the emergence of mood-related illnesses. Because major depressive disorder often emerges in adolescence, we assessed the effects of social defeat stress on responses to depressive-like behaviors in juvenile mice. To do this, postnatal day (PD) 35 male c57BL/6 mice were exposed to 10 days of social defeat stress (PD35-44), while control mice were handled daily. Twenty-four hours after the last episode of defeat (PD45), separate groups of mice were tested in the social interaction, forced swimming, sucrose preference, and elevated plus-maze behavioral assays (n = 7-12 per group). Also, we examined body weight gain across days of social defeat and levels of blood serum corticosterone 40 min after the last episode of defeat stress. Our data indicates that defeated mice exhibited a depressive-like phenotype as inferred from increased social avoidance, increased immobility in the forced swim test, and reduced sucrose preference (a measure of anhedonia), when compared to non-defeated controls. Defeated mice also displayed an anxiogenic-like phenotype when tested on the elevated plus-maze. Lastly, stressed mice displayed lower body weight gain, along with increased blood serum corticosterone levels, when compared to non-stressed controls. Overall, we show that in adolescent male c57BL/6 mice, social defeat stress induces a depression- and anxiety-like phenotype 24 h after the last episode of stress. These data suggest that the social defeat paradigm may be used to examine the etiology of stress-induced mood-related disorders during adolescence.
Collapse
Affiliation(s)
- Sergio D. Iñiguez
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Lace M. Riggs
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Steven J. Nieto
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Genesis Dayrit
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Norma N. Zamora
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Kristi L. Shawhan
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Bryan Cruz
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Brandon L. Warren
- Intramural Research Program, National Institute on Drug Abuse–National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
1024
|
Friedman AK, Walsh JJ, Juarez B, Ku SM, Chaudhury D, Wang J, Li X, Dietz DM, Pan N, Vialou VF, Neve RL, Yue Z, Han MH. Enhancing depression mechanisms in midbrain dopamine neurons achieves homeostatic resilience. Science 2014; 344:313-9. [PMID: 24744379 PMCID: PMC4334447 DOI: 10.1126/science.1249240] [Citation(s) in RCA: 332] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Typical therapies try to reverse pathogenic mechanisms. Here, we describe treatment effects achieved by enhancing depression-causing mechanisms in ventral tegmental area (VTA) dopamine (DA) neurons. In a social defeat stress model of depression, depressed (susceptible) mice display hyperactivity of VTA DA neurons, caused by an up-regulated hyperpolarization-activated current (I(h)). Mice resilient to social defeat stress, however, exhibit stable normal firing of these neurons. Unexpectedly, resilient mice had an even larger I(h), which was observed in parallel with increased potassium (K(+)) channel currents. Experimentally further enhancing Ih or optogenetically increasing the hyperactivity of VTA DA neurons in susceptible mice completely reversed depression-related behaviors, an antidepressant effect achieved through resilience-like, projection-specific homeostatic plasticity. These results indicate a potential therapeutic path of promoting natural resilience for depression treatment.
Collapse
Affiliation(s)
- Allyson K. Friedman
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jessica J. Walsh
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Barbara Juarez
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Stacy M. Ku
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dipesh Chaudhury
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jing Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xianting Li
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David M. Dietz
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nina Pan
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vincent F. Vialou
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rachael L. Neve
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zhenyu Yue
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ming-Hu Han
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
1025
|
Takahashi A, Nagayasu K, Nishitani N, Kaneko S, Koide T. Control of intermale aggression by medial prefrontal cortex activation in the mouse. PLoS One 2014; 9:e94657. [PMID: 24740241 PMCID: PMC3989250 DOI: 10.1371/journal.pone.0094657] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/19/2014] [Indexed: 11/30/2022] Open
Abstract
Aggressive behavior is widely observed throughout the animal kingdom because of its adaptiveness for social animals. However, when aggressive behavior exceeds the species-typical level, it is no longer adaptive, so there should be a mechanism to control excessive aggression to keep it within the adaptive range. Using optogenetics, we demonstrate that activation of excitatory neurons in the medial prefrontal cortex (mPFC), but not the orbitofrontal cortex (OFC), inhibits inter-male aggression in mice. At the same time, optogenetic silencing of mPFC neurons causes an escalation of aggressive behavior both quantitatively and qualitatively. Activation of the mPFC suppresses aggressive bursts and reduces the intensity of aggressive behavior, but does not change the duration of the aggressive bursts. Our findings suggest that mPFC activity has an inhibitory role in the initiation and execution, but not the termination, of aggressive behavior, and maintains such behavior within the adaptive range.
Collapse
Affiliation(s)
- Aki Takahashi
- Mouse Genomics Resource Laboratory, National Institute of Genetics (NIG), Mishima, Shizuoka, Japan
- Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka, Japan
- * E-mail:
| | - Kazuki Nagayasu
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto, Japan
| | - Naoya Nishitani
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto, Japan
| | - Tsuyoshi Koide
- Mouse Genomics Resource Laboratory, National Institute of Genetics (NIG), Mishima, Shizuoka, Japan
- Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka, Japan
| |
Collapse
|
1026
|
Patki G, Solanki N, Atrooz F, Ansari A, Allam F, Jannise B, Maturi J, Salim S. Novel mechanistic insights into treadmill exercise based rescue of social defeat-induced anxiety-like behavior and memory impairment in rats. Physiol Behav 2014; 130:135-44. [PMID: 24732411 DOI: 10.1016/j.physbeh.2014.04.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 01/17/2014] [Accepted: 04/04/2014] [Indexed: 12/12/2022]
Abstract
Social defeat (SD) induced stress causes physiological and behavioral deficits in rodents, including depression and anxiety-like behaviors, as well as memory impairment. Anxiolytic and mood elevating effects of physical exercise are also known. However, rescue effect of physical exercise in social defeat-induced anxiety, depression or memory impairment has not been addressed. The role of epigenetic mechanisms that potentially contribute to these rescue or protective effects is also not known. The present study investigated the effect of moderate treadmill exercise on anxiety-like behavior and memory function in rats subjected to SD using a modified version of the resident-intruder model for social stress (defeat). Changes in histone acetylation and histone-modifying enzymes were examined in hippocampus, amygdala and frontal cortex which are considered critical for anxiety, depression and cognition. Sprague Dawley rats were randomly assigned in four groups; control, exercised, social defeat, social defeat and exercise. At the end of the SD or control exposure lasting 30 min daily for 7 days, one group of SD rats was subjected to treadmill exercise for 2 weeks, whereas the other SD group was handled without exercise. Anxiety-like behavior tests and radial arm water maze test suggested that moderate treadmill exercise rescued social defeat induced anxiety-like behavior and memory impairment. Moreover, exercise normalized SD-induced increase in oxidative stress, most likely by adjusting antioxidant response. Our data suggests involvement of epigenetic mechanisms including histone acetylation of H3 and modulation of methyl-CpG-binding in the hippocampus that might contribute to the rescue effects of exercise in SD-induced behavioral deficits in rats.
Collapse
Affiliation(s)
- Gaurav Patki
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, TX, USA
| | - Naimesh Solanki
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, TX, USA
| | - Fatin Atrooz
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, TX, USA
| | - Amber Ansari
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, TX, USA
| | - Farida Allam
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, TX, USA
| | - Brittany Jannise
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, TX, USA
| | - Jaganmohan Maturi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, TX, USA
| | - Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, TX, USA.
| |
Collapse
|
1027
|
Riga D, Schmitz LJM, van der Harst JE, van Mourik Y, Hoogendijk WJG, Smit AB, De Vries TJ, Spijker S. A sustained depressive state promotes a guanfacine reversible susceptibility to alcohol seeking in rats. Neuropsychopharmacology 2014; 39:1115-24. [PMID: 24192553 PMCID: PMC3957105 DOI: 10.1038/npp.2013.311] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 10/08/2013] [Accepted: 10/10/2013] [Indexed: 12/29/2022]
Abstract
High rates of comorbidity between alcohol use disorder (AUD) and major depressive disorder (MDD) are reported. Preclinical models examining effects of primary depression on secondary AUD are currently absent, preventing adequate testing of drug treatment. Here, we combined social defeat-induced persistent stress (SDPS) and operant alcohol self-administration (SA) paradigms to assess causality between these two neuropsychiatric disorders. We then exploited guanfacine, an FDA-approved adrenergic agent reported to reduce drug craving in humans, against SDPS-induced modulation of operant alcohol SA. Wistar rats were socially defeated and isolated for a period of ≥9 weeks, during which depression-like symptomatology (cognitive and social behavioral symptoms) was assessed. Subsequently, animals were subjected to a 5-month operant alcohol SA paradigm, examining acquisition, motivation, extinction, and cue-induced reinstatement of alcohol seeking. The effects of guanfacine on motivation and relapse were measured at >6 months following defeat. SDPS rats exhibited significant disruption of social and cognitive behavior, including short-term spatial and long-term social memory, several months following defeat. Notably, SDPS increased motivation to obtain alcohol, and cue-induced relapse vulnerability. Guanfacine reversed the SDPS-induced effects on motivation and relapse. Together, our model mimics core symptomatology of a sustained depressive-like state and a subsequent vulnerability to alcohol abuse. We show that SDPS is strongly associated with an enhanced motivation for alcohol intake and relapse. Finally, we show that the clinically employed drug guanfacine has potential as a novel treatment option in comorbid patients, as it effectively reduced the enhanced sensitivity to alcohol and alcohol-associated stimuli.
Collapse
Affiliation(s)
- Danai Riga
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Leanne JM Schmitz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | | | - Yvar van Mourik
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Witte JG Hoogendijk
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Taco J De Vries
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands,Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Sabine Spijker
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands,Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands, Tel: +31 20 598 7111, Fax: +31 20 598 9281, E-mail:
| |
Collapse
|
1028
|
Abstract
INTRODUCTION The success of antidepressant research has long been challenged by a limited mechanistic understanding of depression pathogenesis and antidepressant treatment response. Progress in this field has thereby consistently been hindered by a lack of novel conceptual approaches and sophisticated experimental techniques to dissect the highly intricate neurobiology of depression. Using fresh approaches to investigate the cellular and molecular mechanisms underlying depression will thus be vital for discovery of novel antidepressant targets. AREAS COVERED This article provides an overview of some fundamental problems that depression research is currently facing and critically evaluates how these issues could be addressed by future research. It also discusses novel conceptual and technological advances in the field of neuroscience, particularly in regard to how they may help in providing unprecedented insight into the molecular mechanisms of depression pathogenesis. EXPERT OPINION Although progress in antidepressant drug discovery has been limited over recent years, modern innovations in neuroscience, molecular biology, genetics and bioinformatics are just beginning to revolutionize depression research and to reveal novel and promising treatment targets. Integrating findings from a range of relevant experimental models and using the most advanced technology will be vital for the future success of antidepressant drug discovery.
Collapse
Affiliation(s)
- Christoph Anacker
- McGill University, Douglas Mental Health University Institute , 6875 Boulevard La Salle, Montreal, Quebec, H4H 1R3 , Canada +1 514 761 6131 - 2503 ;
| |
Collapse
|
1029
|
Challis C, Beck SG, Berton O. Optogenetic modulation of descending prefrontocortical inputs to the dorsal raphe bidirectionally bias socioaffective choices after social defeat. Front Behav Neurosci 2014; 8:43. [PMID: 24596546 PMCID: PMC3925846 DOI: 10.3389/fnbeh.2014.00043] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/29/2014] [Indexed: 12/31/2022] Open
Abstract
It has been well established that modulating serotonin (5-HT) levels in humans and animals affects perception and response to social threats, however the circuit mechanisms that control 5-HT output during social interaction are not well understood. A better understanding of these systems could provide groundwork for more precise and efficient therapeutic interventions. Here we examined the organization and plasticity of microcircuits implicated in top-down control of 5-HT neurons in the dorsal raphe nucleus (DRN) by excitatory inputs from the ventromedial prefrontal cortex (vmPFC) and their role in social approach-avoidance decisions. We did this in the context of a social defeat model that induces a long lasting form of social aversion that is reversible by antidepressants. We first used viral tracing and Cre-dependent genetic identification of vmPFC glutamatergic synapses in the DRN to determine their topographic distribution in relation to 5-HT and GABAergic subregions and found that excitatory vmPFC projections primarily localized to GABA-rich areas of the DRN. We then used optogenetics in combination with cFos mapping and slice electrophysiology to establish the functional effects of repeatedly driving vmPFC inputs in DRN. We provide the first direct evidence that vmPFC axons drive synaptic activity and immediate early gene expression in genetically identified DRN GABA neurons through an AMPA receptor-dependent mechanism. In contrast, we did not detect vmPFC-driven synaptic activity in 5-HT neurons and cFos induction in 5-HT neurons was limited. Finally we show that optogenetically increasing or decreasing excitatory vmPFC input to the DRN during sensory exposure to an aggressor's cues enhances or diminishes avoidance bias, respectively. These results clarify the functional organization of vmPFC-DRN pathways and identify GABAergic neurons as a key cellular element filtering top-down vmPFC influences on affect-regulating 5-HT output.
Collapse
Affiliation(s)
- Collin Challis
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine Philadelphia, PA, USA ; Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine Philadelphia, PA, USA
| | - Sheryl G Beck
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine Philadelphia, PA, USA ; Department of Anesthesiology, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine Philadelphia, PA, USA
| | - Olivier Berton
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine Philadelphia, PA, USA ; Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine Philadelphia, PA, USA
| |
Collapse
|
1030
|
Resiliency to social defeat stress relates to the inter-strain social interaction and is influenced by season variation. Neurosci Lett 2014; 561:13-7. [DOI: 10.1016/j.neulet.2013.12.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 12/20/2013] [Indexed: 01/25/2023]
|
1031
|
Kim JM, Zheng HM, Lee BY, Lee WK, Lee DH. Anti-Helicobacter pylori Properties of GutGard™. Prev Nutr Food Sci 2014; 18:104-10. [PMID: 24471118 PMCID: PMC3892500 DOI: 10.3746/pnf.2013.18.2.104] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 04/15/2013] [Indexed: 12/13/2022] Open
Abstract
Presence of Helicobacter pylori is associated with an increased risk of developing upper gastrointestinal tract diseases. Antibiotic therapy and a combination of two or three drugs have been widely used to eradicate H. pylori infections. Due to antibiotic resistant drugs, new drug resources are needed such as plants which contain antibacterial compounds. The aim of this study was to investigate the ability of GutGard™ to inhibit H. pylori growth both in Mongolian gerbils and C57BL/6 mouse models. Male Mongolian gerbils were infected with the bacteria by intragastric inoculation (2×109 CFU/gerbil) 3 times over 5 days and then orally treated once daily 6 times/week for 8 weeks with 15, 30 and 60 mg/kg GutGard™. After the final administration, biopsy samples of the gastric mucosa were assayed for bacterial identification via urease, catalase and ELISA assays as well as immunohistochemistry (IHC). In the Mongolian gerbil model, IHC and ELISA assays revealed that GutGard™ inhibited H. pylori colonization in gastric mucosa in a dose dependent manner. The anti-H. pylori effects of GutGard™ in H. pylori-infected C57BL/6 mice were also examined. We found that treatment with 25 mg/kg GutGard™ significantly reduced H. pylori colonization in mice gastric mucosa. Our results suggest that GutGard™ may be useful as an agent to prevent H. pylori infection.
Collapse
Affiliation(s)
- Jae Min Kim
- National Center of Efficacy Evaluation for the Development of Health Products Targeting Digestive Disorder, Inha Hospital, Incheon 400-712, Korea ; Laboratory of Developmental Genetics, Inha University School of Medicine, Incheon 400-712, Korea
| | - Hong Mei Zheng
- National Center of Efficacy Evaluation for the Development of Health Products Targeting Digestive Disorder, Inha Hospital, Incheon 400-712, Korea
| | - Boo Yong Lee
- Department of Food Science & Biotechnology, CHA University, Gyeonggi 487-010, Korea
| | - Woon Kyu Lee
- National Center of Efficacy Evaluation for the Development of Health Products Targeting Digestive Disorder, Inha Hospital, Incheon 400-712, Korea ; Laboratory of Developmental Genetics, Inha University School of Medicine, Incheon 400-712, Korea
| | - Don Haeng Lee
- National Center of Efficacy Evaluation for the Development of Health Products Targeting Digestive Disorder, Inha Hospital, Incheon 400-712, Korea
| |
Collapse
|
1032
|
Icariin attenuates glucocorticoid insensitivity mediated by repeated psychosocial stress on an ovalbumin-induced murine model of asthma. Int Immunopharmacol 2014; 19:381-90. [PMID: 24462390 DOI: 10.1016/j.intimp.2014.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 01/10/2014] [Accepted: 01/10/2014] [Indexed: 01/23/2023]
Abstract
Evidence shows that psychosocial stress exacerbates asthma, but there is little intervention to alleviate negative effects of psychosocial stress on asthma. We investigated the role of icariin in anti-inflammation and anti-anxiety potential in a murine model combined psychosocial stress with allergic exposure. The results indicated that icariin administered remarkable increased activity in the center of the open field, reversed airway hyperresponsivenesss, reduced inflammatory cytokine infiltration to the lung and whole body and also in part recovered glucocorticoid responsiveness. Furthermore, our data also showed that icariin significantly inhibited increases of corticosterone and markedly increased glucocorticoid receptor mRNA and protein expression in the lungs of mice exposed to both stress and allergen. Collectively, we speculate that inducing glucocorticoid receptor modulation might be the potential mechanisms of icariin to facilitate corticosteroid responsiveness of cytokine production.
Collapse
|
1033
|
Iñiguez SD, Alcantara LF, Warren BL, Riggs LM, Parise EM, Vialou V, Wright KN, Dayrit G, Nieto SJ, Wilkinson MB, Lobo MK, Neve RL, Nestler EJ, Bolaños-Guzmán CA. Fluoxetine exposure during adolescence alters responses to aversive stimuli in adulthood. J Neurosci 2014; 34:1007-21. [PMID: 24431458 PMCID: PMC3891944 DOI: 10.1523/jneurosci.5725-12.2014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 11/30/2013] [Accepted: 12/06/2013] [Indexed: 11/21/2022] Open
Abstract
The mechanisms underlying the enduring neurobiological consequences of antidepressant exposure during adolescence are poorly understood. Here, we assessed the long-term effects of exposure to fluoxetine (FLX), a selective serotonin reuptake inhibitor, during adolescence on behavioral reactivity to emotion-eliciting stimuli. We administered FLX (10 mg/kg, bi-daily, for 15 d) to male adolescent [postnatal day 35 (P35) to P49] C57BL/6 mice. Three weeks after treatment (P70), reactivity to aversive stimuli (i.e., social defeat stress, forced swimming, and elevated plus maze) was assessed. We also examined the effects of FLX on the expression of extracellular signal-regulated kinase (ERK) 1/2-related signaling within the ventral tegmental area (VTA) of adolescent mice and Sprague Dawley rats. Adolescent FLX exposure suppressed depression-like behavior, as measured by the social interaction and forced swim tests, while enhancing anxiety-like responses in the elevated plus maze in adulthood. This complex behavioral profile was accompanied by decreases in ERK2 mRNA and protein phosphorylation within the VTA, while stress alone resulted in opposite neurobiological effects. Pharmacological (U0126) inhibition, as well as virus-mediated downregulation of ERK within the VTA mimicked the antidepressant-like profile observed after juvenile FLX treatment. Conversely, overexpression of ERK2 induced a depressive-like response, regardless of FLX pre-exposure. These findings demonstrate that exposure to FLX during adolescence modulates responsiveness to emotion-eliciting stimuli in adulthood, at least partially, via long-lasting adaptations in ERK-related signaling within the VTA. Our results further delineate the role ERK plays in regulating mood-related behaviors across the lifespan.
Collapse
Affiliation(s)
- Sergio D. Iñiguez
- Department of Psychology, California State University, San Bernardino, California 92407
| | - Lyonna F. Alcantara
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306
| | - Brandon L. Warren
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306
| | - Lace M. Riggs
- Department of Psychology, California State University, San Bernardino, California 92407
| | - Eric M. Parise
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306
| | - Vincent Vialou
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - Katherine N. Wright
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306
| | - Genesis Dayrit
- Department of Psychology, California State University, San Bernardino, California 92407
| | - Steven J. Nieto
- Department of Psychology, California State University, San Bernardino, California 92407
| | - Matthew B. Wilkinson
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - Mary K. Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, and
| | - Rachael L. Neve
- Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Eric J. Nestler
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - Carlos A. Bolaños-Guzmán
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306
| |
Collapse
|
1034
|
Antidepressant-like properties of novel HDAC6-selective inhibitors with improved brain bioavailability. Neuropsychopharmacology 2014; 39:389-400. [PMID: 23954848 PMCID: PMC3870780 DOI: 10.1038/npp.2013.207] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/15/2013] [Accepted: 08/02/2013] [Indexed: 12/13/2022]
Abstract
HDAC inhibitors have been reported to produce antidepressant and pro-cognitive effects in animal models, however, poor brain bioavailability or lack of isoform selectivity of current probes has limited our understanding of their mode of action. We report the characterization of novel pyrimidine hydroxyl amide small molecule inhibitors of HDAC6, brain bioavailable upon systemic administration. We show that two compounds in this family, ACY-738 and ACY-775, inhibit HDAC6 with low nanomolar potency and a selectivity of 60- to 1500-fold over class I HDACs. In contrast to tubastatin A, a reference HDAC6 inhibitor with similar potency and peripheral activity, but more limited brain bioavailability, ACY-738 and ACY-775 induce dramatic increases in α-tubulin acetylation in brain and stimulate mouse exploratory behaviors in novel, but not familiar environments. Interestingly, despite a lack of detectable effect on histone acetylation, we show that ACY-738 and ACY-775 share the antidepressant-like properties of other HDAC inhibitors, such as SAHA and MS-275, in the tail suspension test and social defeat paradigm. These effects of ACY-738 and ACY-775 are directly attributable to the inhibition of HDAC6 expressed centrally, as they are fully abrogated in mice with a neural-specific loss of function of HDAC6. Furthermore, administered in combination, a behaviorally inactive dose of ACY-738 markedly potentiates the anti-immobility activity of a subactive dose of the selective serotonin reuptake inhibitor citalopram. Our results validate new isoform-selective probes for in vivo pharmacological studies of HDAC6 in the CNS and reinforce the viability of this HDAC isoform as a potential target for antidepressant development.
Collapse
|
1035
|
Daskalakis NP, Yehuda R. Principles for developing animal models of military PTSD. Eur J Psychotraumatol 2014; 5:23825. [PMID: 25206946 PMCID: PMC4138703 DOI: 10.3402/ejpt.v5.23825] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/13/2014] [Accepted: 04/29/2014] [Indexed: 01/28/2023] Open
Abstract
The extent to which animal studies can be relevant to military posttraumatic stress disorder (PTSD) continues to be a matter of discussion. Some features of the clinical syndrome are more easily modeled than others. In the animal literature, a great deal of attention is focused on modeling the characteristics of military exposures and their impact on measurable behaviors and biological parameters. There are many issues to consider regarding the ecological validity of predator, social defeat or immobilization stress to combat-related experience. In contrast, less attention has been paid to individual variation following these exposures. Such variation is critical to understand how individual differences in the response to military trauma exposure may result to PTSD or resilience. It is important to consider potential differences in biological findings when comparing extremely exposed to non-exposed animals, versus those that result from examining individual differences. Animal models of military PTSD are also critical in advancing efforts in clinical treatment. In an ideal translational approach to study deployment related outcomes, information from humans and animals, blood and brain, should be carefully considered in tandem, possibly even computed simultaneously, to identify molecules, pathways and networks that are likely to be the key drivers of military PTSD symptoms. With the use novel biological methodologies (e.g., optogenetics) in the animal models, critical genes and pathways can be tuned up or down (rather than over-expressed or ablated completely) in discrete brain regions. Such techniques together with pre-and post-deployment human imaging will accelerate the identification of novel pharmacological and non-pharmacological intervention strategies.
Collapse
Affiliation(s)
- Nikolaos P Daskalakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA ; Mental Health Care Center, James J. Peters Veterans Affairs Medical Center, Bronx, USA
| | - Rachel Yehuda
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA ; Mental Health Care Center, James J. Peters Veterans Affairs Medical Center, Bronx, USA
| |
Collapse
|
1036
|
Abstract
Serotonin (5-HT) modulates neural responses to socioaffective cues and can bias approach or avoidance behavioral decisions, yet the cellular mechanisms underlying its contribution to the regulation of social experiences remain poorly understood. We hypothesized that GABAergic neurons in the dorsal raphe nucleus (DRN) may participate in socioaffective regulation by controlling serotonergic tone during social interaction. We tested this hypothesis using whole-cell recording techniques in genetically identified DRN GABA and 5-HT neurons in mice exposed to social defeat, a model that induces long-lasting avoidance behaviors in a subset of mice responsive to serotonergic antidepressants. Our results revealed that social defeat engaged DRN GABA neurons and drove GABAergic sensitization that strengthened inhibition of 5-HT neurons in mice that were susceptible, but not resilient to social defeat. Furthermore, optogenetic silencing of DRN GABA neurons disinhibited neighboring 5-HT neurons and prevented the acquisition of social avoidance in mice exposed to a social threat, but did not affect a previously acquired avoidance phenotype. We provide the first characterization of GABA neurons in the DRN that monosynaptically inhibit 5-HT neurons and reveal their key role in neuroplastic processes underlying the development of social avoidance.
Collapse
|
1037
|
Campi KL, Greenberg GD, Kapoor A, Ziegler TE, Trainor BC. Sex differences in effects of dopamine D1 receptors on social withdrawal. Neuropharmacology 2013; 77:208-16. [PMID: 24120838 DOI: 10.1016/j.neuropharm.2013.09.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 09/24/2013] [Accepted: 09/29/2013] [Indexed: 01/19/2023]
Abstract
Dopamine signaling in the nucleus accumbens (NAc) plays a critical role in the regulation of motivational states. Recent studies in male rodents show that social defeat stress increases the activity of ventral tegmental dopamine neurons projecting to the NAc, and that this increased activity is necessary for stress-induced social withdrawal. Domestic female mice are not similarly aggressive, which has hindered complementary studies in females. Using the monogamous California mouse (Peromyscus californicus), we found that social defeat increased total dopamine, DOPAC, and HVA content in the NAc in both males and females. These results are generally consistent with previous studies in Mus, and suggest defeat stress also increases NAc dopamine signaling in females. However, these results do not explain our previous observations that defeat stress induces social withdrawal in female but not male California mice. Pharmacological manipulations provided more insights. When 500 ng of the D1 agonist SKF38393 was infused in the NAc shell of females that were naïve to defeat, social interaction behavior was reduced. This same dose of SKF38393 had no effect in males, suggesting that D1 receptor activation is sufficient to induce social withdrawal in females but not males. Intra-accumbens infusion of the D1 antagonist SCH23390 increased social approach behavior in females exposed to defeat but not in females naïve to defeat. This result suggests that D1 receptors are necessary for defeat-induced social withdrawal. Overall, our results suggest that sex differences in molecular pathways that are regulated by D1 receptors contribute to sex differences in social withdrawal behavior.
Collapse
Affiliation(s)
- Katharine L Campi
- Department of Psychology, University of California, Davis, CA 95616, USA
| | - Gian D Greenberg
- Department of Psychology, University of California, Davis, CA 95616, USA; Neuroscience Graduate Group, University of California, Davis, CA 95616, USA
| | - Amita Kapoor
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53706, USA
| | - Toni E Ziegler
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53706, USA
| | - Brian C Trainor
- Department of Psychology, University of California, Davis, CA 95616, USA; Neuroscience Graduate Group, University of California, Davis, CA 95616, USA; Center for Neuroscience, University of California, Davis, CA 95616, USA.
| |
Collapse
|
1038
|
Patki G, Solanki N, Atrooz F, Allam F, Salim S. Depression, anxiety-like behavior and memory impairment are associated with increased oxidative stress and inflammation in a rat model of social stress. Brain Res 2013; 1539:73-86. [PMID: 24096214 DOI: 10.1016/j.brainres.2013.09.033] [Citation(s) in RCA: 286] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 09/20/2013] [Accepted: 09/22/2013] [Indexed: 01/11/2023]
Abstract
In the present study, we have examined the behavioral and biochemical effect of induction of psychological stress using a modified version of the resident-intruder model for social stress (social defeat). At the end of the social defeat protocol, body weights, food and water intake were recorded, depression and anxiety-like behaviors as well as memory function was examined. Biochemical analysis including oxidative stress measurement, inflammatory markers and other molecular parameters, critical to behavioral effects were examined. We observed a significant decrease in the body weight in the socially defeated rats as compared to the controls. Furthermore, social defeat increased anxiety-like behavior and caused memory impairment in rats (P<0.05). Socially defeated rats made significantly more errors in long term memory tests (P<0.05) as compared to control rats. Furthermore, brain extracellular signal-regulated kinase-1/2 (ERK1/2), and an inflammatory marker, interleukin (IL)-6 were activated (P<0.05), while the protein levels of glyoxalase (GLO)-1, glutathione reductase (GSR)-1, calcium/calmodulin-dependent protein kinase type (CAMK)-IV, cAMP-response-element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) were significantly less (P<0.05) in the hippocampus, but not in the prefrontal cortex and amygdala of socially defeated rats, when compared to control rats. We suggest that social defeat stress alters ERK1/2, IL-6, GLO1, GSR1, CAMKIV, CREB, and BDNF levels in specific brain areas, leading to oxidative stress-induced anxiety-depression-like behaviors and as well as memory impairment in rats.
Collapse
Affiliation(s)
- Gaurav Patki
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | | | | | | | | |
Collapse
|
1039
|
Meduri JD, Farnbauch LA, Jasnow AM. Paradoxical enhancement of fear expression and extinction deficits in mice resilient to social defeat. Behav Brain Res 2013; 256:580-90. [PMID: 24029700 DOI: 10.1016/j.bbr.2013.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 08/07/2013] [Accepted: 09/02/2013] [Indexed: 01/19/2023]
Abstract
The exposure to stress has been associated with increased depressive and anxiety symptoms, yet not all individuals respond negatively to the experience of stress. Recent rodent social defeat models demonstrate similar individual differences in response to social stress. In particular, mice subjected to chronic social defeat have been characterized as being either "susceptible" or "resilient" by the level of social interaction following social defeat. Susceptibility is associated with lasting social avoidance as well as increased anxiety-like behavior, and depressive-like symptoms. Resilient animals, however, do not show social avoidance or increased depressive-like symptoms, but retain increased anxiety-like behavior. Thus, it is unclear what "resilience" as measured by social interaction represents in terms of an overall behavioral and physiological phenotype. Here, we use an acute social defeat procedure, which produces distinct behavioral phenotypes in social interaction with no apparent changes in anxiety-like behavior. Susceptible mice display lasting social avoidance, whereas resilient mice display normal social interaction. Susceptible mice also displayed deficits in fear extinction retention but had normal within-session extinction. Paradoxically, resilience was associated with enhanced fear expression, and severe deficits in fear extinction and extinction retention beyond that observed in susceptible mice. These effects in resilient mice were only apparent after the experience of social stress and were not due to impaired behavioral flexibility. These data suggest that mechanisms controlling resilience to acute social defeat as characterized by social interaction leave animals vulnerable to maladaptive fear behavior.
Collapse
Affiliation(s)
- Jeremy D Meduri
- Department of Psychology, Kent State University, 230 Kent Hall, Kent, OH 44242, USA.
| | | | | |
Collapse
|
1040
|
Abstract
Mood disorders are common and debilitating conditions characterized in part by profound deficits in reward-related behavioural domains. A recent literature has identified important structural and functional alterations within the brain's reward circuitry--particularly in the ventral tegmental area-nucleus accumbens pathway--that are associated with symptoms such as anhedonia and aberrant reward-associated perception and memory. This Review synthesizes recent data from human and rodent studies from which emerges a circuit-level framework for understanding reward deficits in depression. We also discuss some of the molecular and cellular underpinnings of this framework, ranging from adaptations in glutamatergic synapses and neurotrophic factors to transcriptional and epigenetic mechanisms.
Collapse
Affiliation(s)
- Scott J Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA. scott.russo@mssm. edu
| | | |
Collapse
|
1041
|
Rutten BPF, Hammels C, Geschwind N, Menne-Lothmann C, Pishva E, Schruers K, van den Hove D, Kenis G, van Os J, Wichers M. Resilience in mental health: linking psychological and neurobiological perspectives. Acta Psychiatr Scand 2013; 128:3-20. [PMID: 23488807 PMCID: PMC3746114 DOI: 10.1111/acps.12095] [Citation(s) in RCA: 220] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/17/2013] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To review the literature on psychological and biological findings on resilience (i.e. the successful adaptation and swift recovery after experiencing life adversities) at the level of the individual, and to integrate findings from animal and human studies. METHOD Electronic and manual literature search of MEDLINE, EMBASE and PSYCHINFO, using a range of search terms around biological and psychological factors influencing resilience as observed in human and experimental animal studies, complemented by review articles and cross-references. RESULTS The term resilience is used in the literature for different phenomena ranging from prevention of mental health disturbance to successful adaptation and swift recovery after experiencing life adversities, and may also include post-traumatic psychological growth. Secure attachment, experiencing positive emotions and having a purpose in life are three important psychological building blocks of resilience. Overlap between psychological and biological findings on resilience in the literature is most apparent for the topic of stress sensitivity, although recent results suggest a crucial role for reward experience in resilience. CONCLUSION Improving the understanding of the links between genetic endowment, environmental impact and gene-environment interactions with developmental psychology and biology is crucial for elucidating the neurobiological and psychological underpinnings of resilience.
Collapse
Affiliation(s)
- B P F Rutten
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical Centre, Maastricht, the Netherlands.
| | - C Hammels
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical CentreMaastricht, the Netherlands
| | - N Geschwind
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical CentreMaastricht, the Netherlands,Research Group on Health Psychology, CLEP, Department of Psychology, University of LeuvenLeuven, Belgium
| | - C Menne-Lothmann
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical CentreMaastricht, the Netherlands
| | - E Pishva
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical CentreMaastricht, the Netherlands
| | - K Schruers
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical CentreMaastricht, the Netherlands,Center for Learning and Experimental Psychology, Catholic University of LeuvenLeuven, Belgium
| | - D van den Hove
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical CentreMaastricht, the Netherlands,Department of Psychiatry, Psychosomatics and Psychotherapy, University of WürzburgWürzburg, Germany
| | - G Kenis
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical CentreMaastricht, the Netherlands
| | - J van Os
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical CentreMaastricht, the Netherlands,King's Health Partners, Department of Psychosis Studies, Institute of Psychiatry, King's College LondonLondon, UK
| | - M Wichers
- Department of Psychiatry and Psychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), European Graduate School of Neuroscience (EURON), Maastricht University Medical CentreMaastricht, the Netherlands
| |
Collapse
|
1042
|
Olfaction and olfactory-mediated behaviour in psychiatric disease models. Cell Tissue Res 2013; 354:69-80. [DOI: 10.1007/s00441-013-1617-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/12/2013] [Indexed: 12/26/2022]
|
1043
|
Social stress models in depression research: what do they tell us? Cell Tissue Res 2013; 354:179-90. [PMID: 23532563 DOI: 10.1007/s00441-013-1606-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/04/2013] [Indexed: 12/18/2022]
Abstract
Interest has recently surged in the use of social stress models, especially social defeat. Such interest lies both in the recognition that stressors of social origin play a major role in human psychopathologies and in the acknowledgement that natural and hence ethologically-based stress models have important translational value. The use of the most recent technology has allowed the recognition of the mechanisms through which social defeat might have enduring psychoneuroendocrine effects, especially social avoidance and anhedonia, two behaviours relevant to human depression. In view of the sensitivity of these behavioural outcomes to repeated antidepressant treatments, the social defeat model has been proposed as a possible animal model of depression. The present survey is aimed at examining the limits of such an interpretation and focuses on methodological aspects and on the relevance of social defeat to the study of anxiety-related pathologies.
Collapse
|
1044
|
Trainor BC, Takahashi EY, Campi KL, Florez SA, Greenberg GD, Laman-Maharg A, Laredo SA, Orr VN, Silva AL, Steinman MQ. Sex differences in stress-induced social withdrawal: independence from adult gonadal hormones and inhibition of female phenotype by corncob bedding. Horm Behav 2013; 63:543-50. [PMID: 23384773 PMCID: PMC3637973 DOI: 10.1016/j.yhbeh.2013.01.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 01/22/2013] [Accepted: 01/27/2013] [Indexed: 01/19/2023]
Abstract
There is compelling evidence for important sex differences in behavioral and hormonal responses to psychosocial stress. Here we examined the effects of gonadal hormones on behavioral responses to social defeat stress in monogamous California mice (Peromyscus californicus). Three episodes of social defeat induced social withdrawal in intact females but not males. Gonadectomy blocked corticosterone responses to defeat in females and sensitized male corticosterone responses. However, gonadectomy had no effects on social interaction behavior, suggesting that social withdrawal is not dependent on gonadal hormones in the adult California mouse. In contrast, defeat reduced exploratory behavior in the open field test for intact but not castrated males. We also examined the effects of social defeat on social interaction behavior when California mice were raised on corncob bedding, which has estrogenic properties. In this dataset of over 300 mice, we observed that social defeat did not induce social withdrawal when females were raised on corncob bedding. This finding suggests that the use of corncob in rodent studies could mask important sex differences in the effects of stress on brain and behavior. Although gonadal hormones do not affect social withdrawal behavior in adults, our data suggest that hormones may act earlier in development to induce a more resilient social phenotype.
Collapse
Affiliation(s)
- Brian C Trainor
- Department of Psychology, University of California, Davis, CA 95616, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1045
|
Epigenetic regulation of RAC1 induces synaptic remodeling in stress disorders and depression. Nat Med 2013; 19:337-44. [PMID: 23416703 PMCID: PMC3594624 DOI: 10.1038/nm.3090] [Citation(s) in RCA: 258] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 01/14/2013] [Indexed: 02/07/2023]
Abstract
Depression induces structural and functional synaptic plasticity in brain reward circuits, although the mechanisms promoting these changes and their relevance to behavioral outcomes are unknown. Transcriptional profiling of the nucleus accumbens (NAc) for Rho GTPase-related genes, which are known regulators of synaptic structure, revealed a sustained reduction in RAS-related C3 botulinum toxin substrate 1 (Rac1) expression after chronic social defeat stress. This was associated with a repressive chromatin state surrounding the proximal promoter of Rac1. Inhibition of class 1 histone deacetylases (HDACs) with MS-275 rescued both the decrease in Rac1 transcription after social defeat stress and depression-related behavior, such as social avoidance. We found a similar repressive chromatin state surrounding the RAC1 promoter in the NAc of subjects with depression, which corresponded with reduced RAC1 transcription. Viral-mediated reduction of Rac1 expression or inhibition of Rac1 activity in the NAc increases social defeat-induced social avoidance and anhedonia in mice. Chronic social defeat stress induces the formation of stubby excitatory spines through a Rac1-dependent mechanism involving the redistribution of synaptic cofilin, an actin-severing protein downstream of Rac1. Overexpression of constitutively active Rac1 in the NAc of mice after chronic social defeat stress reverses depression-related behaviors and prunes stubby spines. Taken together, our data identify epigenetic regulation of RAC1 in the NAc as a disease mechanism in depression and reveal a functional role for Rac1 in rodents in regulating stress-related behaviors.
Collapse
|
1046
|
Sex, stress, and epigenetics: regulation of behavior in animal models of mood disorders. Biol Sex Differ 2013; 4:1. [PMID: 23331332 PMCID: PMC3586570 DOI: 10.1186/2042-6410-4-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 12/31/2012] [Indexed: 01/30/2023] Open
Abstract
Women have a higher incidence of stress related disorders including depression and generalized anxiety disorder, and epigenetic mechanisms likely contribute to this sex difference. Evidence from preclinical research suggests that epigenetic mechanisms are responsible for both sexual dimorphism of brain regions and sensitivity of the stress response. Epigenetic modifications such as DNA methylation and histone modifications can occur transgenerationally, developmentally, or in response to environmental stimuli such as stress exposure. This review will provide an overview of the various forms of epigenetic modifications observed in the central nervous system and will explain how these mechanisms contribute to a sexually dimorphic brain. It will also discuss the ways in which epigenetic alterations coincide with, and functionally contribute to, the behavioral response to stress across the lifespan. Ultimately, this review will focus on novel research utilizing animal models to investigate sex differences in epigenetic mechanisms that influence susceptibility to stress. Exploration of this relationship reveals epigenetic mechanisms with the potential to explain sexual dimorphism in the occurrence of stress related disorders.
Collapse
|
1047
|
Abstract
Each of us has felt afraid, and we can all recognize fear in many animal species. Yet there is no consensus in the scientific study of fear. Some argue that 'fear' is a psychological construct rather than something discoverable through scientific investigation. Others argue that the term 'fear' cannot properly be applied to animals because we cannot know whether they feel afraid. Studies in rodents show that there are highly specific brain circuits for fear, whereas findings from human neuroimaging seem to make the opposite claim. Here, I review the field and urge three approaches that could reconcile the debates. For one, we need a broadly comparative approach that would identify core components of fear conserved across phylogeny. This also pushes us towards the second point of emphasis: an ecological theory of fear that is essentially functional. Finally, we should aim even to incorporate the conscious experience of being afraid, reinvigorating the study of feelings across species.
Collapse
Affiliation(s)
- Ralph Adolphs
- Division of Humanities and Social Sciences, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
1048
|
Abstract
Although tricyclic antidepressants rapidly activate monoaminergic neurotransmission, these drugs must be administered chronically to alleviate symptoms of depression. This observation suggests that molecular mechanisms downstream of monoamine receptor activation, which include the induction of gene transcription, underlie chronic antidepressant-induced changes in behavior. Here we show that methyl-CpG-binding protein 2 (MeCP2) regulates behavioral responses to chronic antidepressant treatment. Imipramine administration induces phosphorylation of MeCP2 at Ser421 (pMeCP2) selectively in the nucleus accumbens and the lateral habenula, two brain regions important for depressive-like behaviors. To test the role of pMeCP2 in depressive-like behaviors, we used male mice that bear a germ-line mutation knocked into the X-linked Mecp2 locus that changes Ser421 to a nonphosphorylatable Ala residue (S421A). MeCP2 S421A knock-in (KI) mice showed increased immobility in forced-swim and tail-suspension tests compared with their wild-type (WT) littermates. However, immobility of both MeCP2 WT and KI mice in forced swim was reduced by acute administration of imipramine, demonstrating that loss of pMeCP2 does not impair acute pharmacological sensitivity to this drug. After chronic social defeat stress, chronic administration of imipramine significantly improved social interaction in the MeCP2 WT mice. In contrast, the MeCP2 KI mice did not respond to chronic imipramine administration. These data suggest novel roles for pMeCP2 in the sensitivity to stressful stimuli and demonstrate that pMeCP2 is required for the effects of chronic imipramine on depressive-like behaviors induced by chronic social defeat stress.
Collapse
|
1049
|
Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons. Nature 2012; 493:532-6. [PMID: 23235832 PMCID: PMC3554860 DOI: 10.1038/nature11713] [Citation(s) in RCA: 800] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 10/25/2012] [Indexed: 12/12/2022]
Abstract
Ventral tegmental area (VTA) dopamine (DA) neurons in the brain’s reward circuit play a crucial role in mediating stress responses1–4 including determining susceptibility vs. resilience to social stress-induced behavioural abnormalities5. VTA DA neurons exhibit two in vivo patterns of firing: low frequency tonic firing and high frequency phasic firing6–8. Phasic firing of the neurons, which is well known to encode reward signals6,7,9, is upregulated by repeated social defeat stress, a highly validated mouse model of depression5,8,10–13. Surprisingly, this pathophysiological effect is seen in susceptible mice only, with no change in firing rate apparent in resilient individuals5,8. However, direct evidence linking—in real-time—DA neuron phasic firing in promoting the susceptible (depression-like) phenotype is lacking. Here, we took advantage of the temporal precision and cell type- and projection pathway-specificity of optogenetics to demonstrate that enhanced phasic firing of these neurons mediates susceptibility to social defeat stress in freely behaving mice. We show that optogenetic induction of phasic, but not tonic, firing, in VTA DA neurons of mice undergoing a subthreshold social defeat paradigm rapidly induced a susceptible phenotype as measured by social avoidance and decreased sucrose preference. Optogenetic phasic stimulation of these neurons also quickly induced a susceptible phenotype in previously resilient mice that had been subjected to repeated social defeat stress. Furthermore, we show differences in projection pathway-specificity in promoting stress susceptibility: phasic activation of VTA neurons projecting to the nucleus accumbens (NAc), but not to the medial prefrontal cortex (mPFC), induced susceptibility to social defeat stress. Conversely, optogenetic inhibition of the VTA-NAc projection induced resilience, while inhibition of the VTA-mPFC projection promoted susceptibility. Overall, these studies reveal novel firing pattern- and neural circuit-specific mechanisms of depression.
Collapse
|
1050
|
Hodes GE, Russo SJ. The Neurobiology of Depression and Anxiety: How Do We Change from Models of Drug Efficacy to Understanding Mood and Anxiety Disorders? DRUG DISCOVERY FOR PSYCHIATRIC DISORDERS 2012. [DOI: 10.1039/9781849734943-00159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Current treatments for depression and anxiety disorders are only effective in approximately half of the patient population. Effective treatments have negative side-effects including sexual dysfunction, weight gain and gastrointestinal problems. Furthermore, even when patients achieve remission, they often need to increase dosage or change treatment across their lifetime as efficacy weakens. The majority of treatments being used today are based on the monoamine hypothesis of depression, a theory of depression that was based on the effectiveness of drugs discovered by chance to alleviate the symptoms of depression. This chapter provides an overview of the neurobiology of depression and anxiety disorders within the context of drug discovery. The chapter starts with what we currently know about these disorders through the lens of the monoamine hypothesis of depression. We then provide a background into the animal models of depression and anxiety that are being used to understand the underlying biology of these disorders and test new treatments. Work conducted using these animal models has directed human imagining and has provided us with new information about both the molecular and cellular mechanism of depression and anxiety as well as the neural circuitry controlling these disorders. Finally, we will conclude with a discussion of new treatments being developed based on empirical evidence about the neurobiology of depression and anxiety and the need to develop more personalized treatments in the future. It is hoped that these new drugs will be able to provide effective treatment for more people, with fewer negative side-effects.
Collapse
Affiliation(s)
- Georgia E. Hodes
- Fishberg Department of Neuroscience and Freidman Brain Institute Mount Sinai School of Medicine 1425 Madison Ave., New York, NY 10029 USA
| | - Scott J. Russo
- Fishberg Department of Neuroscience and Freidman Brain Institute Mount Sinai School of Medicine 1425 Madison Ave., New York, NY 10029 USA
| |
Collapse
|