251
|
Abstract
Pain has a strong emotional component and is defined by its unpleasantness. Chronic pain represents a complex disorder with anxio-depressive symptoms and cognitive deficits. Underlying mechanisms are still not well understood but an important role for interactions between prefrontal cortical areas and subcortical limbic structures has emerged. Evidence from preclinical studies in the rodent brain suggests that neuroplastic changes in prefrontal (anterior cingulate, prelimbic and infralimbic) cortical and subcortical (amygdala and nucleus accumbens) brain areas and their interactions (corticolimbic circuitry) contribute to the complexity and persistence of pain and may be predetermining factors as has been proposed in recent human neuroimaging studies.
Collapse
Affiliation(s)
- Jeremy M Thompson
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, United States; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States.
| |
Collapse
|
252
|
LaGamma CT, Tang WW, Morgan AA, McGowan JC, Brachman RA, Denny CA. Antidepressant but Not Prophylactic Ketamine Administration Alters Calretinin and Calbindin Expression in the Ventral Hippocampus. Front Mol Neurosci 2018; 11:404. [PMID: 30459554 PMCID: PMC6232342 DOI: 10.3389/fnmol.2018.00404] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/15/2018] [Indexed: 01/20/2023] Open
Abstract
Ketamine has been found to have rapid, long-lasting antidepressant effects in treatment-resistant (TR) patients with major depressive disorder (MDD). Recently, we have also shown that ketamine acts as a prophylactic to protect against the development of stress-induced depressive-like behavior in mice, indicating that a preventative treatment against mental illness using ketamine is possible. While there is significant investigation into ketamine’s antidepressant mechanism of action, little is known about ketamine’s underlying prophylactic mechanism. More specifically, whether ketamine’s prophylactic action is molecularly similar to or divergent from its antidepressant action is entirely unknown. Here, we sought to characterize immunohistochemical signatures of cell populations governing ketamine’s antidepressant and prophylactic effects. 129S6/SvEv mice were treated with saline (Sal) or ketamine (K) either before a social defeat (SD) stressor as a prophylactic, or after SD as an antidepressant, then subsequently assessed for depressive-like behavior. Post-fixed brains were processed for doublecortin (DCX), calretinin (CR) and calbindin (CB) expression. The number of DCX+ neurons in the dentate gyrus (DG) of the hippocampus (HPC) was not affected by prophylactic or antidepressant ketamine treatment, while the number of CR+ neurons in the ventral hilus increased with antidepressant ketamine under SD conditions. Moreover, antidepressant, but not prophylactic ketamine administration significantly altered CR and CB expression in the ventral HPC (vHPC). These data show that while antidepressant ketamine treatment mediates some of its effects via adult hippocampal markers, prophylactic ketamine administration does not, at least in 129S6/SvEv mice. These data suggest that long-lasting behavioral effects of prophylactic ketamine are independent of hippocampal DCX, CR and CB expression in stress-susceptible mice.
Collapse
Affiliation(s)
- Christina T LaGamma
- Division of Integrative Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, United States
| | - William W Tang
- Department of Psychiatry, Columbia University, New York, NY, United States
| | - Ashlea A Morgan
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, NY, United States
| | | | - Rebecca A Brachman
- Department of Psychiatry, Columbia University, New York, NY, United States
| | - Christine A Denny
- Division of Integrative Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, United States.,Department of Psychiatry, Columbia University, New York, NY, United States
| |
Collapse
|
253
|
Park JY, Park SY, Kwon H, Song Y, Yun B, Lee Y, Cho Y, Joo A, Han PL. A Group of Descending Glutamatergic Neurons Activated by Stress in Corticolimbic Regions Project to the Nucleus Accumbens. Exp Neurobiol 2018; 27:387-396. [PMID: 30429648 PMCID: PMC6221842 DOI: 10.5607/en.2018.27.5.387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/02/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022] Open
Abstract
The nucleus accumbens (NAc) is the major component of the ventral striatum that regulates stress-induced depression. The NAc receives dopaminergic inputs from the ventral tegmental area (VTA), and the role of VTA-NAc neurons in stress response has been recently characterized. The NAc also receives glutamatergic inputs from various forebrain structures including the prelimbic cortex (PL), basolateral amygdala (BLA), and ventral hippocampus (vHIP), whereas the role of those glutamatergic afferents in stress response remains underscored. In the present study, we investigated the extent to which descending glutamatergic neurons activated by stress in the PL, BLA, and vHIP project to the NAc. To specifically label the input neurons into the NAc, fluorescent-tagged cholera toxin subunit B (CTB), which can be used as a retrograde neuronal tracer, was injected into the NAc. After two weeks, the mice were placed under restraint for 1 h. Subsequent histological analyses indicated that CTB-positive cells were detected in 170~680 cells/mm2 in the PL, BLA, and vHIP, and those CTB-positive cells were mostly glutamatergic. In the PL, BLA, and vHIP regions analyzed, stress-induced c-Fos expression was found in 20~100 cells/mm2. Among the CTB-positive cells, 2.6% in the PL, 4.2% in the BLA, and 1.1% in the vHIP were co-labeled by c-Fos, whereas among c-Fos-positive cells, 7.7% in the PL, 19.8% in the BLA, and 8.5% in the vHIP were co-labeled with CTB. These results suggest that the NAc receives a significant but differing proportion of glutamatergic inputs from the PL, BLA, and vHIP in stress response.
Collapse
Affiliation(s)
- Jin-Young Park
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - So Young Park
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Hyejin Kwon
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Yumi Song
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Boin Yun
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Yubin Lee
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Yeryung Cho
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Ahran Joo
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Pyung-Lim Han
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea.,Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
254
|
Schafer M, Schiller D. Navigating Social Space. Neuron 2018; 100:476-489. [PMID: 30359610 PMCID: PMC6226014 DOI: 10.1016/j.neuron.2018.10.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/20/2018] [Accepted: 10/03/2018] [Indexed: 01/17/2023]
Abstract
Cognitive maps are encoded in the hippocampal formation and related regions and range from the spatial to the purely conceptual. Neural mechanisms that encode information into relational structures, up to an arbitrary level of abstraction, may explain such a broad range of representation. Research now indicates that social life can also be mapped by these mechanisms: others' spatial locations, social memory, and even a two-dimensional social space framed by social power and affiliation. The systematic mapping of social life onto a relational social space facilitates adaptive social decision making, akin to social navigation. This emerging line of research has implications for cognitive mapping research, clinical disorders that feature hippocampal dysfunction, and the field of social cognitive neuroscience.
Collapse
Affiliation(s)
- Matthew Schafer
- Department of Psychiatry, Department of Neuroscience, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniela Schiller
- Department of Psychiatry, Department of Neuroscience, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
255
|
García‐Pardo MP, Miñarro J, Llansola M, Felipo V, Aguilar MA. Role ofNMDAandAMPAglutamatergic receptors in the effects of social defeat on the rewarding properties ofMDMAin mice. Eur J Neurosci 2018; 50:2623-2634. [DOI: 10.1111/ejn.14190] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Affiliation(s)
- M. P. García‐Pardo
- Unidad de Investigación Psicobiología de las Drogodependencias Departamento de Psicobiología Facultad de Psicología Universidad de Valencia Avda. Blasco Ibáñez, 21 46010 Valencia Spain
- Unitat Predepartamental de Medicina Facultat de Ciències de la Salut Universitat Jaume I. Castelló de la Plana Castelló Spain
| | - J. Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias Departamento de Psicobiología Facultad de Psicología Universidad de Valencia Avda. Blasco Ibáñez, 21 46010 Valencia Spain
| | - M. Llansola
- Laboratory of Neurobiology Centro Investigación Príncipe Felipe Valencia Spain
| | - V. Felipo
- Laboratory of Neurobiology Centro Investigación Príncipe Felipe Valencia Spain
| | - M. A. Aguilar
- Unidad de Investigación Psicobiología de las Drogodependencias Departamento de Psicobiología Facultad de Psicología Universidad de Valencia Avda. Blasco Ibáñez, 21 46010 Valencia Spain
| |
Collapse
|
256
|
McAllister BB, Wright DK, Wortman RC, Shultz SR, Dyck RH. Elimination of vesicular zinc alters the behavioural and neuroanatomical effects of social defeat stress in mice. Neurobiol Stress 2018; 9:199-213. [PMID: 30450385 PMCID: PMC6234281 DOI: 10.1016/j.ynstr.2018.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/26/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic stress can have deleterious effects on mental health, increasing the risk of developing depression or anxiety. But not all individuals are equally affected by stress; some are susceptible while others are more resilient. Understanding the mechanisms that lead to these differing outcomes has been a focus of considerable research. One unexplored mechanism is vesicular zinc – zinc that is released by neurons as a neuromodulator. We examined how chronic stress, induced by repeated social defeat, affects mice that lack vesicular zinc due to genetic deletion of zinc transporter 3 (ZnT3). These mice, unlike wild type mice, did not become socially avoidant of a novel conspecific, suggesting resilience to stress. However, they showed enhanced sensitivity to the potentiating effect of stress on cued fear memory. Thus, the contribution of vesicular zinc to stress susceptibility is not straightforward. Stress also increased anxiety-like behaviour but produced no deficits in a spatial Y-maze test. We found no evidence that microglial activation or hippocampal neurogenesis accounted for the differences in behavioural outcome. Volumetric analysis revealed that ZnT3 KO mice have larger corpus callosum and parietal cortex volumes, and that corpus callosum volume was decreased by stress in ZnT3 KO, but not wild type, mice.
Collapse
Key Words
- BLA, Basolateral amygdala
- CC, Corpus callosum
- Chronic stress
- Depression
- EPM, Elevated plus-maze
- Fear memory
- LV, Lateral ventricles
- Magnetic resonance imaging (MRI)
- NAc, Nucleus accumbens
- NSF, Novelty-suppressed feeding
- PBS, Phosphate-buffered saline
- PFA, Paraformaldehyde
- PFC, Prefrontal cortex
- RSD, Repeated social defeat
- SLC30A3
- Synaptic zinc
- ZnT3, Zinc transporter 3
- dHPC, Dorsal hippocampus
- vHPC, Ventral hippocampus
Collapse
Affiliation(s)
- Brendan B McAllister
- Department of Psychology & Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - David K Wright
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Ryan C Wortman
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Richard H Dyck
- Department of Psychology & Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
257
|
Camporeze B, Manica BA, Bonafé GA, Ferreira JJC, Diniz AL, de Oliveira CTP, Mathias Junior LR, de Aguiar PHP, Ortega MM. Optogenetics: the new molecular approach to control functions of neural cells in epilepsy, depression and tumors of the central nervous system. Am J Cancer Res 2018; 8:1900-1918. [PMID: 30416844 PMCID: PMC6220144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/14/2018] [Indexed: 06/09/2023] Open
Abstract
The optogenetic tools have been described as valuable techniques to study neural activity through light stimulation, as well as potential neuromodulator approaches in the management of several central nervous system (CNS) diseases. Since the first bacteriorhodopsin protein described as a single-component light-activated regulator of transmembrane ion flow description, in 1980's, the focus has been on channel proteins for neurobiology; however, the advances in engineering techniques showed involvement changes in cellular biological behavior in several types of proteins involved in cell cytoskeleton regulation, motility and gene expression. Although the use of this technology has been published in many papers, a question still remains regarding real results and potential clinical applicability in CNS diseases, as well as the publications scarcity that systematically analyses the published results. Lastly, the aim of this review is to discuss the experimental results, molecular mechanisms and potential clinical applications of optogenetic tools in epilepsy and depression treatment, as well as its applicability in the treatment of CNS tumors.
Collapse
Affiliation(s)
- Bruno Camporeze
- Postgraduate Program in Health Science, Laboratory of Cellular and Molecular Biology and Bioactive Compounds, São Francisco University (USF)Bragança Paulista-SP, Brazil
- Postgraduate Program in Health Science, Department of Neurosurgery, Institute of Medical Assistance of The State Public Servant (IAMSPE)São Paulo-SP, Brazil
| | - Bruno Alcântara Manica
- Departament of Neurology, Medical School University Pontifical University Catholic of São Paulo (PUCSP)Sorocaba-SP, Brazil
| | - Gabriel Alves Bonafé
- Postgraduate Program in Health Science, Laboratory of Cellular and Molecular Biology and Bioactive Compounds, São Francisco University (USF)Bragança Paulista-SP, Brazil
| | | | - Aurélio Lourenço Diniz
- Departament of Neurology, Medical School University Pontifical University Catholic of São Paulo (PUCSP)Sorocaba-SP, Brazil
| | | | | | - Paulo Henrique Pires de Aguiar
- Postgraduate Program in Health Science, Department of Neurosurgery, Institute of Medical Assistance of The State Public Servant (IAMSPE)São Paulo-SP, Brazil
- Departament of Neurology, Medical School University Pontifical University Catholic of São Paulo (PUCSP)Sorocaba-SP, Brazil
- Departament of Neurosurgery, Hospital Santa PaulaSão Paulo-SP, Brazil
- Department of Research and Innovation, Laboratory of Cellular and Molecular Biology, Medical School of ABC (FMABC)Santo André-SP, Brazil
| | - Manoela Marques Ortega
- Postgraduate Program in Health Science, Laboratory of Cellular and Molecular Biology and Bioactive Compounds, São Francisco University (USF)Bragança Paulista-SP, Brazil
| |
Collapse
|
258
|
Ketamine and its metabolite (2R,6R)-hydroxynorketamine induce lasting alterations in glutamatergic synaptic plasticity in the mesolimbic circuit. Mol Psychiatry 2018; 23:2066-2077. [PMID: 29158578 DOI: 10.1038/mp.2017.239] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 08/24/2017] [Accepted: 09/07/2017] [Indexed: 12/17/2022]
Abstract
Low doses of ketamine trigger rapid and lasting antidepressant effects after one injection in treatment-resistant patients with major depressive disorder. Modulation of AMPA receptors (AMPARs) in the hippocampus and prefrontal cortex is suggested to mediate the antidepressant action of ketamine and of one of its metabolites (2R,6R)-hydroxynorketamine ((2R,6R)-HNK). We have examined whether ketamine and (2R,6R)-HNK affect glutamatergic transmission and plasticity in the mesolimbic system, brain regions known to have key roles in reward-motivated behaviors, mood and hedonic drive. We found that one day after the injection of a low dose of ketamine, long-term potentiation (LTP) in the nucleus accumbens (NAc) was impaired. Loss of LTP was maintained for 7 days and was not associated with an altered basal synaptic transmission mediated by AMPARs and N-methyl-D-aspartate receptors (NMDARs). Inhibition of mammalian target of rapamycin signaling with rapamycin did not prevent the ketamine-induced loss of LTP but inhibited LTP in saline-treated mice. However, ketamine blunted the increase in the phosphorylation of the GluA1 subunit of AMPARs at a calcium/calmodulin-dependent protein kinase II/protein kinase C site induced by an LTP induction protocol. Moreover, ketamine caused a persistent increased phosphorylation of GluA1 at a protein kinase A site. (2R,6R)-HNK also impaired LTP in the NAc. In dopaminergic neurons of the ventral tegmental area from ketamine- or (2R,6R)-HNK-treated mice, AMPAR-mediated responses were depressed, while those mediated by NMDARs were unaltered, which resulted in a reduced AMPA/NMDA ratio, a measure of long-term synaptic depression. These results demonstrate that a single injection of ketamine or (2R,6R)-HNK induces enduring alterations in the function of AMPARs and synaptic plasticity in brain regions involved in reward-related behaviors.
Collapse
|
259
|
Mitchell SJ, Maguire EP, Cunningham L, Gunn BG, Linke M, Zechner U, Dixon CI, King SL, Stephens DN, Swinny JD, Belelli D, Lambert JJ. Early-life adversity selectively impairs α2-GABA A receptor expression in the mouse nucleus accumbens and influences the behavioral effects of cocaine. Neuropharmacology 2018; 141:98-112. [PMID: 30138693 PMCID: PMC6178871 DOI: 10.1016/j.neuropharm.2018.08.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/02/2018] [Accepted: 08/19/2018] [Indexed: 01/20/2023]
Abstract
Haplotypes of the Gabra2 gene encoding the α2-subunit of the GABAA receptor (GABAAR) are associated with drug abuse, suggesting that α2-GABAARs may play an important role in the circuitry underlying drug misuse. The genetic association of Gabra2 haplotypes with cocaine addiction appears to be evident primarily in individuals who had experienced childhood trauma. Given this association of childhood trauma, cocaine abuse and the Gabra2 haplotypes, we have explored in a mouse model of early life adversity (ELA) whether such events influence the behavioral effects of cocaine and if, as suggested by the human studies, α2-GABAARs in the nucleus accumbens (NAc) are involved in these perturbed behaviors. In adult mice prior ELA caused a selective decrease of accumbal α2-subunit mRNA, resulting in a selective decrease in the number and size of the α2-subunit (but not the α1-subunit) immunoreactive clusters in NAc core medium spiny neurons (MSNs). Functionally, in adult MSNs ELA decreased the amplitude and frequency of GABAAR-mediated miniature inhibitory postsynaptic currents (mIPSCs), a profile similar to that of α2 "knock-out" (α2-/-) mice. Behaviourally, adult male ELA and α2-/- mice exhibited an enhanced locomotor response to acute cocaine and blunted sensitisation upon repeated cocaine administration, when compared to their appropriate controls. Collectively, these findings reveal a neurobiological mechanism which may relate to the clinical observation that early trauma increases the risk for substance abuse disorder (SAD) in individuals harbouring haplotypic variations in the Gabra2 gene.
Collapse
Affiliation(s)
- Scott J Mitchell
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee DD1 9SY, Scotland, United Kingdom
| | - Edward P Maguire
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee DD1 9SY, Scotland, United Kingdom
| | - Linda Cunningham
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee DD1 9SY, Scotland, United Kingdom
| | - Benjamin G Gunn
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee DD1 9SY, Scotland, United Kingdom
| | - Matthias Linke
- Institute of Human Genetics, Mainz University, Medical Center, Mainz, Germany
| | - Ulrich Zechner
- Institute of Human Genetics, Mainz University, Medical Center, Mainz, Germany
| | - Claire I Dixon
- School of Psychology, University of Sussex, Falmer, Brighton, BN 9QG, United Kingdom
| | - Sarah L King
- School of Psychology, University of Sussex, Falmer, Brighton, BN 9QG, United Kingdom
| | - David N Stephens
- School of Psychology, University of Sussex, Falmer, Brighton, BN 9QG, United Kingdom
| | - Jerome D Swinny
- Institute for Biomedical & Biomolecular Sciences, School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, United Kingdom.
| | - Delia Belelli
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee DD1 9SY, Scotland, United Kingdom.
| | - Jeremy J Lambert
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital & Medical School, Dundee University, Dundee DD1 9SY, Scotland, United Kingdom.
| |
Collapse
|
260
|
Parkinson's Disease-Linked LRRK2-G2019S Mutation Alters Synaptic Plasticity and Promotes Resilience to Chronic Social Stress in Young Adulthood. J Neurosci 2018; 38:9700-9711. [PMID: 30249796 DOI: 10.1523/jneurosci.1457-18.2018] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/06/2018] [Accepted: 09/13/2018] [Indexed: 12/20/2022] Open
Abstract
The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is a prevalent cause of late-onset Parkinson's disease, producing psychiatric and motor symptoms, including depression, that are indistinguishable from sporadic cases. Here we tested how this mutation impacts depression-related behaviors and associated synaptic responses and plasticity in mice expressing a Lrrk2-G2019S knock-in mutation. Young adult male G2019S knock-in and wild-type mice were subjected to chronic social defeat stress (CSDS), a validated depression model, and other tests of anhedonia, anxiety, and motor learning. We found that G2019S mice were highly resilient to CSDS, failing to exhibit social avoidance compared to wild-type mice, many of which exhibited prominent social avoidance and were thus susceptible to CSDS. In the absence of CSDS, no behavioral differences between genotypes were found. Whole-cell recordings of spiny projection neurons (SPNs) in the nucleus accumbens revealed that glutamatergic synapses in G2019S mice lacked functional calcium-permeable AMPARs, and following CSDS, failed to accumulate inwardly rectifying AMPAR responses characteristic of susceptible mice. Based on this abnormal AMPAR response profile, we asked whether long-term potentiation (LTP) of corticostriatal synaptic strength was affected. We found that both D1 receptor (D1R)- and D2R-SPNs in G2019S mutants were unable to express LTP, with D2R-SPNs abnormally expressing long-term depression following an LTP-induction protocol. Thus, G2019S promotes resilience to chronic social stress in young adulthood, likely reflecting synapses constrained in their ability to undergo experience-dependent plasticity. These unexpected findings may indicate early adaptive coping mechanisms imparted by the G2019S mutation.SIGNIFICANCE STATEMENT The G2019S mutation in LRRK2 causes late-onset Parkinson's disease (PD). LRRK2 is highly expressed in striatal neurons throughout life, but it is unclear how mutant LRRK2 affects striatal neuron function and behaviors in young adulthood. We addressed this question using Lrrk2-G2019S knock-in mice. The data show that young adult G2019S mice were unusually resilient to a depression-like syndrome resulting from chronic social stress. Further, mutant striatal synapses were incapable of forms of synaptic plasticity normally accompanying depression-like behavior and important for supporting the full range of cognitive function. These data suggest that in humans, LRRK2 mutation may affect striatal circuit function in ways that alter normal responses to stress and could be relevant for treatment strategies for non-motor PD symptoms.
Collapse
|
261
|
Russell AL, Handa RJ, Wu TJ. Sex-Dependent Effects of Mild Blast-induced Traumatic Brain Injury on Corticotropin-releasing Factor Receptor Gene Expression: Potential Link to Anxiety-like Behaviors. Neuroscience 2018; 392:1-12. [PMID: 30248435 DOI: 10.1016/j.neuroscience.2018.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/18/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) affects 1.7 million people in the United States every year, resulting in increased risk of death and disabilities. A significant portion of TBIs experienced by military personnel are induced by explosive blast devices. Active duty military personnel are especially vulnerable to mild blast-induced (mb)TBI and the associated long-term effects, such as anxiety disorders. Additionally, females are at an increased risk of being diagnosed with anxiety-related disorders. The mechanism by which mbTBI results in anxiety disorders in males and females is unknown. The sexually dimorphic corticotropin-releasing factor (CRF) is a brain signaling system linked to anxiety. CRF and its family of related peptides modulate anxiety-related behaviors by binding to CRF receptor subtypes 1 and 2 (CRFR1, CRFR2, respectively). These receptors are distributed throughout limbic structures that control behaviors related to emotion, memory, and arousal. Therefore, the aim of this study was to understand the link between mbTBI and anxiety by examining the impact of mbTBI on the CRFR system in male and female mice. mbTBI increased anxiety-like behaviors in both males and females (p < 0.05). In the present study, mbTBI did not alter CRFR1 gene expression in males or females. However, mbTBI disrupted CRFR2 gene expression in different limbic structures in males and females. In males, mbTBI increased baseline CRFR2 gene expression in the ventral hippocampus (p < 0.05) and decreased restraint-induced expression in the anterior bed nucleus of the stria terminalis (aBNST) and amygdala (p < 0.05). In females, mbTBI decreased restraint-induced CRFR2 gene expression in the dorsal hippocampus (p < 0.05). The inherent sex differences and the mbTBI-induced decrease in restraint-induced CRFR2 gene expression may contribute to anxiety-like behaviors. The results of the present study show that the response to mbTBI within the limbic structures modulates anxiety in a sex-dependent manner. The studies further suggest that CRFR2 may serve as a potential target to mitigate mbTBI effects.
Collapse
Affiliation(s)
- Ashley L Russell
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, United States; Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - T John Wu
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, United States; Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States; Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| |
Collapse
|
262
|
Turner BD, Kashima DT, Manz KM, Grueter CA, Grueter BA. Synaptic Plasticity in the Nucleus Accumbens: Lessons Learned from Experience. ACS Chem Neurosci 2018; 9:2114-2126. [PMID: 29280617 DOI: 10.1021/acschemneuro.7b00420] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Synaptic plasticity contributes to behavioral adaptations. As a key node in the reward pathway, the nucleus accumbens (NAc) is important for determining motivation-to-action outcomes. Across animal models of motivation including addiction, depression, anxiety, and hedonic feeding, selective recruitment of neuromodulatory signals and plasticity mechanisms have been a focus of physiologists and behaviorists alike. Experience-dependent plasticity mechanisms within the NAc vary depending on the distinct afferents and cell-types over time. A greater understanding of molecular mechanisms determining how these changes in synaptic strength track with behavioral adaptations will provide insight into the process of learning and memory along with identifying maladaptations underlying pathological behavior. Here, we summarize recent findings detailing how changes in NAc synaptic strength and mechanisms of plasticity manifest in various models of motivational disorders.
Collapse
Affiliation(s)
- Brandon D. Turner
- Vanderbilt Brain Institute, Nashville, Tennessee 37232, United States
| | - Daniel T. Kashima
- Vanderbilt Brain Institute, Nashville, Tennessee 37232, United States
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kevin M. Manz
- Vanderbilt Brain Institute, Nashville, Tennessee 37232, United States
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Carrie A. Grueter
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Brad A. Grueter
- Vanderbilt Brain Institute, Nashville, Tennessee 37232, United States
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
263
|
Lindenbach D, Seamans JK, Phillips AG. Activation of the ventral subiculum reinvigorates behavior after failure to achieve a goal: Implications for dopaminergic modulation of motivational processes. Behav Brain Res 2018; 356:266-270. [PMID: 30201390 DOI: 10.1016/j.bbr.2018.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 08/30/2018] [Accepted: 09/05/2018] [Indexed: 11/20/2022]
Abstract
Previous studies confirm that brief electrical stimulation of glutamatergic afferents from the ventral subiculum (vSub) can significantly enhance dopamine release in the ventral striatum for an extended duration (>20 min). However, the functional significance of this effect on motivated behavior remains to be specified. Here we tested the hypothesis that brief electrical stimulation of the ventral subiculum (20 Hz for 10 s) might increase effort expenditure for food rewards. Motivation was assessed by a progressive ratio lever pressing task, which requires continuous escalation of the numbers of lever presses to receive each subsequent sucrose pellet, eventually resulting in the failure to achieve the required ratio for a food reward. vSub stimulation at the start of a session did not affect the rate or total number of lever presses prior to reaching the "break point". In contrast, stimulation of the vSub with identical parameters on a post break point trial resulted in a significant increase in total responses. These findings demonstrate that activation of the vSub with parameters that modulate dopamine efflux in the nucleus accumbens can re-activate goal-directed behavior after failure to achieve a goal. Our data highlight a possible role for the vSub in the pathophysiology and potential treatment of motivational processes linked to psychiatric disease.
Collapse
Affiliation(s)
- David Lindenbach
- Djavad Mowafaghian Centre for Brain Health, Department of Psychiatry, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Jeremy K Seamans
- Djavad Mowafaghian Centre for Brain Health, Department of Psychiatry, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Anthony G Phillips
- Djavad Mowafaghian Centre for Brain Health, Department of Psychiatry, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
264
|
Reed SJ, Lafferty CK, Mendoza JA, Yang AK, Davidson TJ, Grosenick L, Deisseroth K, Britt JP. Coordinated Reductions in Excitatory Input to the Nucleus Accumbens Underlie Food Consumption. Neuron 2018; 99:1260-1273.e4. [DOI: 10.1016/j.neuron.2018.07.051] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 06/14/2018] [Accepted: 07/27/2018] [Indexed: 12/21/2022]
|
265
|
Huang P, Gao T, Dong Z, Zhou C, Lai Y, Pan T, Liu Y, Zhao X, Sun X, Hua H, Wen G, Gao L, Lv Z. Neural circuitry among connecting the hippocampus, prefrontal cortex and basolateral amygdala in a mouse depression model: Associations correlations between BDNF levels and BOLD – fMRI signals. Brain Res Bull 2018; 142:107-115. [DOI: 10.1016/j.brainresbull.2018.06.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 06/21/2018] [Accepted: 06/28/2018] [Indexed: 01/16/2023]
|
266
|
Abstract
Depression and anxiety are the most common mood disorders affecting 300 million sufferers worldwide. Maladaptive changes in the neuroendocrine stress response is cited as the most common underlying cause, though how the circuits underlying this response are controlled at the molecular level, remains largely unknown. Approximately 40% of patients do not respond to current treatments, indicating that untapped mechanisms exist. Here we review recent evidence implicating JNK in the control of anxiety and depressive-like behavior with a particular focus on its action in immature granule cells of the hippocampal neurogenic niche and the potential for therapeutic targeting for affective disorders.
Collapse
Affiliation(s)
- Patrik Hollos
- Turku Centre for Biotechnology, Åbo Akademi and University of Turku, BioCity, Turku FIN, Finland
| | - Francesca Marchisella
- Turku Centre for Biotechnology, Åbo Akademi and University of Turku, BioCity, Turku FIN, Finland
| | - Eleanor T Coffey
- Turku Centre for Biotechnology, Åbo Akademi and University of Turku, BioCity, Turku FIN, Finland
| |
Collapse
|
267
|
Targeting the Mouse Ventral Hippocampus in the Intrahippocampal Kainic Acid Model of Temporal Lobe Epilepsy. eNeuro 2018; 5:eN-NWR-0158-18. [PMID: 30131968 PMCID: PMC6102375 DOI: 10.1523/eneuro.0158-18.2018] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/08/2018] [Accepted: 06/29/2018] [Indexed: 11/21/2022] Open
Abstract
Here we describe a novel mouse model of temporal lobe epilepsy (TLE) that moves the site of kainate injection from the rodent dorsal hippocampus (corresponding to the human posterior hippocampus) to the ventral hippocampus (corresponding to the human anterior hippocampus). We compare the phenotypes of this new model—with respect to seizures, cognitive impairment, affective deficits, and histopathology—to the standard dorsal intrahippocampal kainate model. Our results demonstrate that histopathological measures of granule cell dispersion and mossy fiber sprouting maximize near the site of kainate injection. Somewhat surprisingly, both the dorsal and ventral models exhibit similar spatial memory impairments in addition to similar electrographic and behavioral seizure burdens. In contrast, we find a more pronounced affective (anhedonic) phenotype specifically in the ventral model. These results demonstrate that the ventral intrahippocampal kainic acid model recapitulates critical pathologies of the dorsal model while providing a means to further study affective phenotypes such as depression in TLE.
Collapse
|
268
|
The Innate Immune Receptors TLR2/4 Mediate Repeated Social Defeat Stress-Induced Social Avoidance through Prefrontal Microglial Activation. Neuron 2018; 99:464-479.e7. [DOI: 10.1016/j.neuron.2018.06.035] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/29/2018] [Accepted: 06/21/2018] [Indexed: 12/17/2022]
|
269
|
Godoy LD, Rossignoli MT, Delfino-Pereira P, Garcia-Cairasco N, de Lima Umeoka EH. A Comprehensive Overview on Stress Neurobiology: Basic Concepts and Clinical Implications. Front Behav Neurosci 2018; 12:127. [PMID: 30034327 PMCID: PMC6043787 DOI: 10.3389/fnbeh.2018.00127] [Citation(s) in RCA: 418] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022] Open
Abstract
Stress is recognized as an important issue in basic and clinical neuroscience research, based upon the founding historical studies by Walter Canon and Hans Selye in the past century, when the concept of stress emerged in a biological and adaptive perspective. A lot of research after that period has expanded the knowledge in the stress field. Since then, it was discovered that the response to stressful stimuli is elaborated and triggered by the, now known, stress system, which integrates a wide diversity of brain structures that, collectively, are able to detect events and interpret them as real or potential threats. However, different types of stressors engage different brain networks, requiring a fine-tuned functional neuroanatomical processing. This integration of information from the stressor itself may result in a rapid activation of the Sympathetic-Adreno-Medullar (SAM) axis and the Hypothalamus-Pituitary-Adrenal (HPA) axis, the two major components involved in the stress response. The complexity of the stress response is not restricted to neuroanatomy or to SAM and HPA axes mediators, but also diverge according to timing and duration of stressor exposure, as well as its short- and/or long-term consequences. The identification of neuronal circuits of stress, as well as their interaction with mediator molecules over time is critical, not only for understanding the physiological stress responses, but also to understand their implications on mental health.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Polianna Delfino-Pereira
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Norberto Garcia-Cairasco
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Eduardo Henrique de Lima Umeoka
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
270
|
Anacker C, Luna VM, Stevens GS, Millette A, Shores R, Jimenez JC, Chen B, Hen R. Hippocampal neurogenesis confers stress resilience by inhibiting the ventral dentate gyrus. Nature 2018; 559:98-102. [PMID: 29950730 PMCID: PMC6118212 DOI: 10.1038/s41586-018-0262-4] [Citation(s) in RCA: 383] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 05/16/2018] [Indexed: 12/22/2022]
Abstract
Adult neurogenesis in the dentate gyrus of the hippocampus is highly regulated by environmental influences, and functionally implicated in behavioral responses to stress and antidepressants1,2,3,4. However, how adult-born neurons regulate dentate gyrus information processing to protect from stress-induced anxiety-like behavior is unknown. Here we show that neurogenesis confers resilience to chronic stress by inhibiting the activity of mature granule cells in the ventral dentate gyrus (vDG), a subregion implicated in mood regulation. We found that chemogenetic inhibition of adult-born neurons in the vDG promotes susceptibility to social defeat stress while increasing neurogenesis confers resilience to chronic stress. Using in vivo calcium (Ca2+) imaging to record neuronal activity from large cell populations in the vDG, we show that increased neurogenesis results in a decrease in the activity of “stress-responsive cells” that are active preferentially during attacks or while mice explore anxiogenic environments. These effects on dentate gyrus activity are necessary and sufficient for stress resilience, as direct silencing of the vDG confers resilience, while excitation promotes susceptibility. Our results suggest that the activity of the vDG may be a key factor in determining individual levels of vulnerability to stress and related psychiatric disorders.
Collapse
Affiliation(s)
- Christoph Anacker
- Department of Psychiatry, Division of Systems Neuroscience, Columbia University and Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA.
| | - Victor M Luna
- Department of Psychiatry, Division of Systems Neuroscience, Columbia University and Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA
| | - Gregory S Stevens
- Department of Psychiatry, Division of Systems Neuroscience, Columbia University and Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA
| | - Amira Millette
- Department of Psychiatry, Division of Systems Neuroscience, Columbia University and Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA
| | - Ryan Shores
- Department of Psychiatry, Division of Systems Neuroscience, Columbia University and Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA
| | - Jessica C Jimenez
- Department of Psychiatry, Division of Systems Neuroscience, Columbia University and Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA
| | - Briana Chen
- Department of Psychiatry, Division of Systems Neuroscience, Columbia University and Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA
| | - René Hen
- Department of Psychiatry, Division of Systems Neuroscience, Columbia University and Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA. .,Department of Neuroscience, Columbia University, New York, NY, USA. .,Department of Pharmacology, Columbia University, New York, NY, USA.
| |
Collapse
|
271
|
Carbamoylated erythropoietin modulates cognitive outcomes of social defeat and differentially regulates gene expression in the dorsal and ventral hippocampus. Transl Psychiatry 2018; 8:113. [PMID: 29884778 PMCID: PMC5993867 DOI: 10.1038/s41398-018-0168-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 05/01/2018] [Indexed: 02/07/2023] Open
Abstract
Cognitive deficits are widespread in psychiatric disorders and frequently as debilitating as the affective component. Widely prescribed antidepressants for treating depressive disorders have limited efficacy in normalizing cognitive function. Erythropoietin (Epo) has been shown to improve cognitive function in schizophrenia and treatment resistant depressed patients. However, the potent elevation of red blood cell counts by Epo can cause hematological complications in non-anemic patients. We investigated a chemically engineered, posttranslational modification of Epo, carbamoylation, which renders it non-erythropoietic. We conducted mass-spectrometry-based peptide mapping of carbamoylated Epo (Cepo) and tested its ability to improve cognitive function after social defeat stress. Gene expression analysis in discrete brain regions was performed to obtain mechanistic insight of Cepo action. Cepo reversed stress-induced spatial working memory deficits while affecting long-term (24 h) novel object recognition in these rats. Contextual fear conditioning following defeat was enhanced by Cepo, but attenuated in controls. However, Cepo improved fear extinction in all rats compared to vehicle treatment. Cepo induced differential gene expression of BDNF, VGF, Arc, TH. and neuritin in the mPFC and discrete hippocampal subfields, with strongest induction in the dorsal hippocampus. Analysis of gene-brain region-behavior interactions showed that Cepo-induced neurotrophic mechanisms influence cognitive function. Carbamoylated erythropoietin can be developed as a therapeutic neurotrophic agent to treat cognitive dysfunction in neuropsychiatric diseases. Due to its distinct mechanism of action, it is unlikely to cross react with the activity of currently prescribed small molecule drugs and can be used as an add-on biologic drug.
Collapse
|
272
|
Larrieu T, Sandi C. Stress-Induced Depression: Is Social Rank a Predictive Risk Factor? Bioessays 2018; 40:e1800012. [PMID: 29869396 DOI: 10.1002/bies.201800012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/27/2018] [Indexed: 12/17/2022]
Abstract
An intriguing question in the field of stress is what makes an individual more likely to be susceptible or resilient to stress-induced depression. Predisposition to stress susceptibility is believed to be influenced by genetic factors and early adversity. However, beyond genetics and life experiences, recent evidence has highlighted social rank as a key determinant of susceptibility to stress, underscoring dominant individuals as the vulnerable ones. This evidence is in conflict with epidemiological, clinical, and animal work pointing at a link between social subordination and depression. Here, we review and analyze rodent protocols addressing the relevance of social rank to predict vulnerability to chronic social stress. We also discuss whether a specific social status (i.e., dominance or subordination) is the appropriate predictor of vulnerability to develop stress-induced depression or rather, the loss of social rank and resources.
Collapse
Affiliation(s)
- Thomas Larrieu
- Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne 1015, Switzerland
| | - Carmen Sandi
- Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne 1015, Switzerland
| |
Collapse
|
273
|
Rusconi F, Battaglioli E. Acute Stress-Induced Epigenetic Modulations and Their Potential Protective Role Toward Depression. Front Mol Neurosci 2018; 11:184. [PMID: 29904343 PMCID: PMC5990609 DOI: 10.3389/fnmol.2018.00184] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/14/2018] [Indexed: 11/16/2022] Open
Abstract
Psychiatric disorders entail maladaptive processes impairing individuals’ ability to appropriately interface with environment. Among them, depression is characterized by diverse debilitating symptoms including hopelessness and anhedonia, dramatically impacting the propensity to live a social and active life and seriously affecting working capability. Relevantly, besides genetic predisposition, foremost risk factors are stress-related, such as experiencing chronic psychosocial stress—including bullying, mobbing and abuse—, and undergoing economic crisis or chronic illnesses. In the last few years the field of epigenetics promised to understand core mechanisms of gene-environment crosstalk, contributing to get into pathogenic processes of many disorders highly influenced by stressful life conditions. However, still very little is known about mechanisms that tune gene expression to adapt to the external milieu. In this Perspective article, we discuss a set of protective, functionally convergent epigenetic processes induced by acute stress in the rodent hippocampus and devoted to the negative modulation of stress-induced immediate early genes (IEGs) transcription, hindering stress-driven morphostructural modifications of corticolimbic circuitry. We also suggest that chronic stress damaging protective epigenetic mechanisms, could bias the functional trajectory of stress-induced neuronal morphostructural modification from adaptive to maladaptive, contributing to the onset of depression in vulnerable individuals. A better understanding of the epigenetic response to stress will be pivotal to new avenues of therapeutic intervention to treat depression, especially in light of limited efficacy of available antidepressant drugs.
Collapse
Affiliation(s)
- Francesco Rusconi
- Department of Medical Biotechnologies and Translational Medicine, University of Milan Via Fratelli Cervi, Segrate, Italy
| | - Elena Battaglioli
- Department of Medical Biotechnologies and Translational Medicine, University of Milan Via Fratelli Cervi, Segrate, Italy.,CNR Institute of Neuroscience Via Vanvitelli, Milan, Italy
| |
Collapse
|
274
|
Deslauriers J, Toth M, Der-Avakian A, Risbrough VB. Current Status of Animal Models of Posttraumatic Stress Disorder: Behavioral and Biological Phenotypes, and Future Challenges in Improving Translation. Biol Psychiatry 2018; 83:895-907. [PMID: 29338843 PMCID: PMC6085893 DOI: 10.1016/j.biopsych.2017.11.019] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/05/2017] [Accepted: 11/03/2017] [Indexed: 12/23/2022]
Abstract
Increasing predictability of animal models of posttraumatic stress disorder (PTSD) has required active collaboration between clinical and preclinical scientists. Modeling PTSD is challenging, as it is a heterogeneous disorder with ≥20 symptoms. Clinical research increasingly utilizes objective biological measures (e.g., imaging, peripheral biomarkers) or nonverbal behaviors and/or physiological responses to complement verbally reported symptoms. This shift toward more-objectively measurable phenotypes enables refinement of current animal models of PTSD, and it supports the incorporation of homologous measures across species. We reviewed >600 articles to examine the ability of current rodent models to probe biological phenotypes of PTSD (e.g., sleep disturbances, hippocampal and fear-circuit dysfunction, inflammation, glucocorticoid receptor hypersensitivity) in addition to behavioral phenotypes. Most models reliably produced enduring generalized anxiety-like or depression-like behaviors, as well as hyperactive fear circuits, glucocorticoid receptor hypersensitivity, and response to long-term selective serotonin reuptake inhibitors. Although a few paradigms probed fear conditioning/extinction or utilized peripheral immune, sleep, and noninvasive imaging measures, we argue that these should be incorporated more to enhance translation. Data on female subjects, on subjects at different ages across the life span, or on temporal trajectories of phenotypes after stress that can inform model validity and treatment study design are needed. Overall, preclinical (and clinical) PTSD researchers are increasingly incorporating homologous biological measures to assess markers of risk, response, and treatment outcome. This shift is exciting, as we and many others hope it not only will support translation of drug efficacy from animal models to clinical trials but also will potentially improve predictability of stage II for stage III clinical trials.
Collapse
Affiliation(s)
- Jessica Deslauriers
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, California
| | - Mate Toth
- Department of Behavioural Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andre Der-Avakian
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, California.
| |
Collapse
|
275
|
Generalization of Conditioned Auditory Fear is Regulated by Maternal Effects on Ventral Hippocampal Synaptic Plasticity. Neuropsychopharmacology 2018; 43:1297-1307. [PMID: 29154366 PMCID: PMC5916357 DOI: 10.1038/npp.2017.281] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 09/30/2017] [Accepted: 11/12/2017] [Indexed: 01/09/2023]
Abstract
Maternal care shapes individual differences in fear-associated neural circuitry. In rats, maternal licking and grooming (LG) in early life regulates ventral hippocampal (VH) function and plasticity in adulthood, but its consequent effect on the regulation of fear memories remains unknown. We report an effect of maternal care on generalization of learned fear, such that offspring of high LG mothers express generalized fear responses when confronted with neutral stimuli following auditory fear conditioning. These animals simultaneously display a reduction in the magnitude of VH long-term potentiation (LTP) expressed and reduced input-output transformation in Schaffer collateral synapses. Inhibition of VH-LTP during learning specifically increases fear generalization in offspring of low LG mothers during recall, suggesting a role for VH synaptic plasticity in the specification of fear memories. These findings suggest that rearing by low LG dams enhances the efficacy of fear-related neural systems to support accurate encoding of fear memories through effects on the VH.
Collapse
|
276
|
Peña CJ, Nestler EJ. Progress in Epigenetics of Depression. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:41-66. [PMID: 29933956 DOI: 10.1016/bs.pmbts.2017.12.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Depression is a prevalent and complex psychiatric syndrome. Epigenetic mechanisms bridge the genetic and environmental factors that contribute to the pathophysiology of depression. A surge of research over the last decade has identified changes in DNA methylation, histone modifications, histone organization, and noncoding RNAs associated with depression and stress-induced depression-like behavior in animal models. We focus here on associations of epigenetic factors concurrent with depression and depression-like behavior, although risk for depression and some of the associated epigenetic changes are known to have developmental origins. Finally, emerging technology may enable breakthroughs in the ability to rescue depression-associated epigenetic modifications at specific genes, greatly enhancing specificity of future potential therapeutic treatments.
Collapse
Affiliation(s)
- Catherine J Peña
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eric J Nestler
- Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
277
|
Lorsch ZS, Loh YHE, Purushothaman I, Walker DM, Parise EM, Salery M, Cahill ME, Hodes GE, Pfau ML, Kronman H, Hamilton PJ, Issler O, Labonté B, Symonds AE, Zucker M, Zhang TY, Meaney MJ, Russo SJ, Shen L, Bagot RC, Nestler EJ. Estrogen receptor α drives pro-resilient transcription in mouse models of depression. Nat Commun 2018; 9:1116. [PMID: 29549264 PMCID: PMC5856766 DOI: 10.1038/s41467-018-03567-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 02/22/2018] [Indexed: 11/30/2022] Open
Abstract
Most people exposed to stress do not develop depression. Animal models have shown that stress resilience is an active state that requires broad transcriptional adaptations, but how this homeostatic process is regulated remains poorly understood. In this study, we analyze upstream regulators of genes differentially expressed after chronic social defeat stress. We identify estrogen receptor α (ERα) as the top regulator of pro-resilient transcriptional changes in the nucleus accumbens (NAc), a key brain reward region implicated in depression. In accordance with these findings, nuclear ERα protein levels are altered by stress in male and female mice. Further, overexpression of ERα in the NAc promotes stress resilience in both sexes. Subsequent RNA-sequencing reveals that ERα overexpression in NAc reproduces the transcriptional signature of resilience in male, but not female, mice. These results indicate that NAc ERα is an important regulator of pro-resilient transcriptional changes, but with sex-specific downstream targets.
Collapse
Affiliation(s)
- Zachary S Lorsch
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Yong-Hwee Eddie Loh
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Deena M Walker
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Eric M Parise
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Marine Salery
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Georgia E Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| | - Madeline L Pfau
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Hope Kronman
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Peter J Hamilton
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Orna Issler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Benoit Labonté
- Department of Neuroscience and Psychiatry, Faculty of Medicine, Laval University, 2601 Chemin de la Canardière Québec, QC, G1J 2G3, Canada
| | - Ann E Symonds
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Matthew Zucker
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Tie Yuan Zhang
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Institute, Sackler Program for Epigenetics and Psychobiology, Departments of Psychiatry and Neurology and Neurosurgery, McGill University, 6875 Boulevard Lasalle, Montréal, QC, H4H 1R3, Canada
| | - Michael J Meaney
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Institute, Sackler Program for Epigenetics and Psychobiology, Departments of Psychiatry and Neurology and Neurosurgery, McGill University, 6875 Boulevard Lasalle, Montréal, QC, H4H 1R3, Canada
| | - Scott J Russo
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Li Shen
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Rosemary C Bagot
- Departments of Psychology & Psychiatry, Ludmer Centre for Neuroinformatics and Mental Health, McGill University, 1205 Avenue Dr Penfield, Montréal, QC, H3A 1B1, Canada
| | - Eric J Nestler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA.
| |
Collapse
|
278
|
Boutros N, Der-Avakian A, Kesby JP, Lee S, Markou A, Semenova S. Effects of adolescent alcohol exposure on stress-induced reward deficits, brain CRF, monoamines and glutamate in adult rats. Psychopharmacology (Berl) 2018; 235:737-747. [PMID: 29181815 DOI: 10.1007/s00213-017-4789-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 11/13/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Adolescent alcohol exposure may increase depression vulnerability in adulthood by increasing the anhedonic response to stress. METHODS Male Wistar rats (postnatal days 28-53) were exposed to binge-like adolescent intermittent ethanol (AIE) or water. In adulthood, rats were exposed to social defeat, consisting of daily confrontations with an aggressive conspecific, followed by testing of brain reward function in a discrete-trial current-intensity intracranial self-stimulation procedure for 10 consecutive days. Neurochemistry and corticotropin-releasing factor (CRF) and CRF receptor 1 (CRFR1) mRNA levels were assessed in corticolimbic brain areas on day 11 of social defeat stress. RESULTS Social defeat elevated reward thresholds in both AIE- and water-exposed rats indicating stress-induced anhedonia. However, AIE-exposed rats were more likely to show threshold elevations after repeated stress compared to water-exposed rats. AIE exposure decreased CRF mRNA levels in the nucleus accumbens and increased CRFR1 mRNA levels in the prefrontal cortex, while stress increased CRF mRNA levels in the central amygdala. In the caudate putamen, AIE exposure decreased dopamine turnover, while stress increased glutamate and serotonin metabolism and turnover. CONCLUSIONS These results demonstrate increased risk of repeated stress-induced anhedonia after AIE exposure, an effect that may be due to alterations in brain CRF and dopamine systems. These results suggest that the increased rates of depression reported in people with a history of adolescent alcohol exposure may be related to alterations in brain reward and stress systems that may contribute to increased stress-induced anhedonia.
Collapse
Affiliation(s)
| | | | - James P Kesby
- University of California San Diego, La Jolla, CA, USA.,Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland, Australia
| | - Soon Lee
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Athina Markou
- University of California San Diego, La Jolla, CA, USA
| | - Svetlana Semenova
- University of California San Diego, La Jolla, CA, USA. .,PAREXEL International, 1560 E Chevy Chase Dr, Glendale, CA, 91206, USA.
| |
Collapse
|
279
|
Wei X, Ma T, Cheng Y, Huang CC, Wang X, Lu J, Wang J. Dopamine D1 or D2 receptor-expressing neurons in the central nervous system. Addict Biol 2018; 23:569-584. [PMID: 28436559 DOI: 10.1111/adb.12512] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/13/2017] [Accepted: 03/11/2017] [Indexed: 12/21/2022]
Abstract
Dopamine signals mainly through D1 receptors (D1Rs) and D2 receptors (D2Rs); D1R-expressing or D2R-expressing neurons contribute to distinct reward and addictive behaviors. Traditionally, transgenic mice expressing green fluorescent protein (GFP) under D1R or D2R promoters are used for fluorescent verification in electrophysiology studies, whereas Cre mice are employed for behavioral research. However, it is unknown whether the same neuronal populations are targeted in GFP and Cre mice. Additionally, while D1Rs and D2Rs are known to be expressed in different striatal neurons, their expression patterns outside the striatum remain unclear. The present study addressed these two questions by using several transgenic mouse lines expressing fluorescent proteins (GFP or tdTomato) or Cre under the control of D1R or D2R promoters. We found a high degree of overlap between GFP-positive and Cre-positive neurons in the striatum and hippocampus. Additionally, we discovered that D1Rs and D2Rs were highly segregated in the orbitofrontal cortex, prefrontal cortex, dorsal and ventral hippocampus, and amygdala: ~4-34 percent of neurons co-expressed these receptors. Importantly, slice electrophysiological studies demonstrated that D1R-positive and D1R-negative hippocampal neurons were functionally distinct in a mouse line generated by crossing Drd1a-Cre mice with a Cre reporter Ai14 line. Lastly, we discovered that chronic alcohol intake differentially altered D1R-positive and D2R-positive neuron excitability in the ventral CA1. These data suggest that GFP and Cre mice target the same populations of striatal neurons, D1R-expressing or D2R-expressing neurons are highly segregated outside the striatum, and these neurons in the ventral hippocampal may exert distinct roles in alcohol addiction.
Collapse
Affiliation(s)
- Xiaoyan Wei
- Department of Neuroscience and Experimental Therapeutics, College of Medicine; Texas A&M University Health Science Center; Bryan TX USA
| | - Tengfei Ma
- Department of Neuroscience and Experimental Therapeutics, College of Medicine; Texas A&M University Health Science Center; Bryan TX USA
| | - Yifeng Cheng
- Department of Neuroscience and Experimental Therapeutics, College of Medicine; Texas A&M University Health Science Center; Bryan TX USA
| | - Cathy C.Y. Huang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine; Texas A&M University Health Science Center; Bryan TX USA
| | - Xuehua Wang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine; Texas A&M University Health Science Center; Bryan TX USA
| | - Jiayi Lu
- Department of Neuroscience and Experimental Therapeutics, College of Medicine; Texas A&M University Health Science Center; Bryan TX USA
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine; Texas A&M University Health Science Center; Bryan TX USA
| |
Collapse
|
280
|
Bulin SE, Mendoza ML, Richardson DR, Song KH, Solberg TD, Yun S, Eisch AJ. Dentate gyrus neurogenesis ablation via cranial irradiation enhances morphine self-administration and locomotor sensitization. Addict Biol 2018. [PMID: 28626932 DOI: 10.1111/adb.12524] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adult dentate gyrus (DG) neurogenesis is important for hippocampal-dependent learning and memory, but the role of new neurons in addiction-relevant learning and memory is unclear. To test the hypothesis that neurogenesis is involved in the vulnerability to morphine addiction, we ablated adult DG neurogenesis and examined morphine self-administration (MSA) and locomotor sensitization. Male Sprague-Dawley rats underwent hippocampal-focused, image-guided X-ray irradiation (IRR) to eliminate new DG neurons or sham treatment (Sham). Six weeks later, rats underwent either MSA (Sham = 16, IRR = 15) or locomotor sensitization (Sham = 12, IRR = 12). Over 21 days of MSA, IRR rats self-administered ~70 percent more morphine than Sham rats. After 28 days of withdrawal, IRR rats pressed the active lever 40 percent more than Sham during extinction. This was not a general enhancement of learning or locomotion, as IRR and Sham groups had similar operant learning and inactive lever presses. For locomotor sensitization, both IRR and Sham rats sensitized, but IRR rats sensitized faster and to a greater extent. Furthermore, dose-response revealed that IRR rats were more sensitive at a lower dose. Importantly, these increases in locomotor activity were not apparent after acute morphine administration and were not a byproduct of irradiation or post-irradiation recovery time. Therefore, these data, along with other previously published data, indicate that reduced hippocampal neurogenesis confers vulnerability for multiple classes of drugs. Thus, therapeutics to specifically increase or stabilize hippocampal neurogenesis could aid in preventing initial addiction as well as future relapse.
Collapse
Affiliation(s)
- Sarah E. Bulin
- Department of Psychiatry; University of Texas Southwestern Medical Center; Dallas TX USA
- Department of Pharmacology; University of Texas Health Science Center; San Antonio TX USA
| | - Matthew L. Mendoza
- Department of Psychiatry; University of Texas Southwestern Medical Center; Dallas TX USA
| | - Devon R. Richardson
- Department of Psychiatry; University of Texas Southwestern Medical Center; Dallas TX USA
| | - Kwang H. Song
- Department of Radiology Oncology; University of Texas Southwestern Medical Center; Dallas TX USA
- Texas Oncology PA; Fort Worth TX USA
| | - Timothy D. Solberg
- Department of Radiology Oncology; University of Texas Southwestern Medical Center; Dallas TX USA
- Department of Radiation Oncology; University of California; San Francisco CA USA
| | - Sanghee Yun
- Mahoney Institute of Neurosciences; University of Pennsylvania Perelman School of Medicine; Philadelphia PA USA
- Department of Anesthesiology and Critical Care Medicine; Children's Hospital of Philadelphia; Philadelphia PA USA
| | - Amelia J. Eisch
- Department of Psychiatry; University of Texas Southwestern Medical Center; Dallas TX USA
- Mahoney Institute of Neurosciences; University of Pennsylvania Perelman School of Medicine; Philadelphia PA USA
- Department of Anesthesiology and Critical Care Medicine; Children's Hospital of Philadelphia; Philadelphia PA USA
| |
Collapse
|
281
|
Edelmann E, Lessmann V. Dopaminergic innervation and modulation of hippocampal networks. Cell Tissue Res 2018; 373:711-727. [PMID: 29470647 DOI: 10.1007/s00441-018-2800-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/17/2018] [Indexed: 02/06/2023]
Abstract
The catecholamine dopamine plays an important role in hippocampus-dependent plasticity and related learning and memory processes. Dopamine secretion in the hippocampus is activated by, e.g., salient or novel stimuli, thereby helping to establish and to stabilize hippocampus-dependent memories. Disturbed dopaminergic function in the hippocampus leads to severe pathophysiological conditions. While the role and importance of dopaminergic modulation of hippocampal networks have been unequivocally proven, there is still a lack of detailed molecular and cellular mechanistic understanding of how dopamine orchestrates these hippocampal processes. In this chapter of the special issue "Hippocampal structure and function," we will discuss the current understanding of dopaminergic modulation of basal synaptic transmission and long-lasting, activity-dependent potentiation or depression.
Collapse
Affiliation(s)
- Elke Edelmann
- Institut für Physiologie, Otto-von-Guericke-Universität, Medizinische Fakultät, Leipziger Str. 44, 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Otto-von-Guericke University, Universitätsplatz 2, 39106, Magdeburg, Germany.
| | - Volkmar Lessmann
- Institut für Physiologie, Otto-von-Guericke-Universität, Medizinische Fakultät, Leipziger Str. 44, 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Otto-von-Guericke University, Universitätsplatz 2, 39106, Magdeburg, Germany.
| |
Collapse
|
282
|
Calipari ES, Godino A, Peck EG, Salery M, Mervosh NL, Landry JA, Russo SJ, Hurd YL, Nestler EJ, Kiraly DD. Granulocyte-colony stimulating factor controls neural and behavioral plasticity in response to cocaine. Nat Commun 2018; 9:9. [PMID: 29339724 PMCID: PMC5770429 DOI: 10.1038/s41467-017-01881-x] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 10/20/2017] [Indexed: 01/27/2023] Open
Abstract
Cocaine addiction is characterized by dysfunction in reward-related brain circuits, leading to maladaptive motivation to seek and take the drug. There are currently no clinically available pharmacotherapies to treat cocaine addiction. Through a broad screen of innate immune mediators, we identify granulocyte-colony stimulating factor (G-CSF) as a potent mediator of cocaine-induced adaptations. Here we report that G-CSF potentiates cocaine-induced increases in neural activity in the nucleus accumbens (NAc) and prefrontal cortex. In addition, G-CSF injections potentiate cocaine place preference and enhance motivation to self-administer cocaine, while not affecting responses to natural rewards. Infusion of G-CSF neutralizing antibody into NAc blocks the ability of G-CSF to modulate cocaine's behavioral effects, providing a direct link between central G-CSF action in NAc and cocaine reward. These results demonstrate that manipulating G-CSF is sufficient to alter the motivation for cocaine, but not natural rewards, providing a pharmacotherapeutic avenue to manipulate addictive behaviors without abuse potential.
Collapse
Affiliation(s)
- Erin S Calipari
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Arthur Godino
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biology, École Normale Supérieure de Lyon, Lyon, France
| | - Emily G Peck
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marine Salery
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicholas L Mervosh
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph A Landry
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yasmin L Hurd
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric J Nestler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Drew D Kiraly
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
283
|
Knowland D, Lim BK. Circuit-based frameworks of depressive behaviors: The role of reward circuitry and beyond. Pharmacol Biochem Behav 2018; 174:42-52. [PMID: 29309799 PMCID: PMC6340396 DOI: 10.1016/j.pbb.2017.12.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/29/2017] [Accepted: 12/31/2017] [Indexed: 12/23/2022]
Abstract
Major depressive disorder (MDD) is a common but serious neuropsychiatric affliction that comprises a diverse set of symptoms such as the inability to feel pleasure, lack of motivation, changes in appetite, and cognitive difficulties. Given the patient to patient symptomatic variability in MDD and differing severities of individual symptoms, it is likely that maladaptive changes in distinct brain areas may mediate discrete symptoms in MDD. The advent and recent surge of studies using viral-genetic approaches have allowed for circuit-specific dissection of networks underlying motivational behavior. In particular, areas such as the ventral tegmental area (VTA), nucleus accumbens (NAc), and ventral pallidum (VP) are thought to generally promote reward, with the medial prefrontal cortex (mPFC) providing top-down control of reward seeking. On the contrary, the lateral habenula (LHb) is considered to be the aversive center of the brain as it has been shown to encode negative valence. The behavioral symptoms of MDD may arise from a disruption in the reward circuitry, hyperactivity of aversive centers, or a combination of the two. Thus, gaining access to specific circuits within the brain and how separate motivational-relevant regions transmit and encode information between each other in the context of separate depression-related symptoms can provide critical knowledge towards symptom-specific treatment of MDD. Here, we review published literature emphasizing circuit- and cell type-specific dissection of depressive-like behaviors in animal models of depression with a particular focus on the chronic social defeat stress model of MDD.
Collapse
Affiliation(s)
- Daniel Knowland
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Byung Kook Lim
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Neurobiology Section Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
284
|
Val‐Laillet D, Guérin S, Coquery N, Nogret I, Formal M, Romé V, Le Normand L, Meurice P, Randuineau G, Guilloteau P, Malbert C, Parnet P, Lallès J, Segain J. Oral sodium butyrate impacts brain metabolism and hippocampal neurogenesis, with limited effects on gut anatomy and function in pigs. FASEB J 2018; 32:2160-2171. [DOI: 10.1096/fj.201700547rr] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- David Val‐Laillet
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
- Centre de Recherche en Nutrition Humaine Ouest (CRNH‐Ouest)NantesFrance
| | - Sylvie Guérin
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Nicolas Coquery
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Isabelle Nogret
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Michèle Formal
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Véronique Romé
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Laurence Le Normand
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Paul Meurice
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Gwénaëlle Randuineau
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Paul Guilloteau
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | | | - Patricia Parnet
- Centre de Recherche en Nutrition Humaine Ouest (CRNH‐Ouest)NantesFrance
- INRA, Unité Mixte de Recherche (UMR) 1280INRA‐Université de Nantes, Physiologie des Adaptations Nutritionnelles (PhAN)NantesFrance
- Institut des Maladies de l'Appareil DigestifCentre Hospitalier Universitaire (CHU) Ho tel‐DieuNantesFrance
| | - Jean‐Paul Lallès
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
- Centre de Recherche en Nutrition Humaine Ouest (CRNH‐Ouest)NantesFrance
| | - Jean‐Pierre Segain
- Centre de Recherche en Nutrition Humaine Ouest (CRNH‐Ouest)NantesFrance
- INRA, Unité Mixte de Recherche (UMR) 1280INRA‐Université de Nantes, Physiologie des Adaptations Nutritionnelles (PhAN)NantesFrance
- Institut des Maladies de l'Appareil DigestifCentre Hospitalier Universitaire (CHU) Ho tel‐DieuNantesFrance
| |
Collapse
|
285
|
Akil H, Gordon J, Hen R, Javitch J, Mayberg H, McEwen B, Meaney MJ, Nestler EJ. Treatment resistant depression: A multi-scale, systems biology approach. Neurosci Biobehav Rev 2018; 84:272-288. [PMID: 28859997 PMCID: PMC5729118 DOI: 10.1016/j.neubiorev.2017.08.019] [Citation(s) in RCA: 285] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/21/2017] [Accepted: 08/26/2017] [Indexed: 01/10/2023]
Abstract
An estimated 50% of depressed patients are inadequately treated by available interventions. Even with an eventual recovery, many patients require a trial and error approach, as there are no reliable guidelines to match patients to optimal treatments and many patients develop treatment resistance over time. This situation derives from the heterogeneity of depression and the lack of biomarkers for stratification by distinct depression subtypes. There is thus a dire need for novel therapies. To address these known challenges, we propose a multi-scale framework for fundamental research on depression, aimed at identifying the brain circuits that are dysfunctional in several animal models of depression as well the changes in gene expression that are associated with these models. When combined with human genetic and imaging studies, our preclinical studies are starting to identify candidate circuits and molecules that are altered both in models of disease and in patient populations. Targeting these circuits and mechanisms can lead to novel generations of antidepressants tailored to specific patient populations with distinctive types of molecular and circuit dysfunction.
Collapse
Affiliation(s)
- Huda Akil
- Depression Task Force, Hope for Depression Research Foundation, New York, NY 10019, United States; University of Michigan, United States
| | - Joshua Gordon
- Depression Task Force, Hope for Depression Research Foundation, New York, NY 10019, United States; Columbia University, United States; New York State Psychiatric Institute, United States
| | - Rene Hen
- Depression Task Force, Hope for Depression Research Foundation, New York, NY 10019, United States; Columbia University, United States; New York State Psychiatric Institute, United States
| | - Jonathan Javitch
- Depression Task Force, Hope for Depression Research Foundation, New York, NY 10019, United States; Columbia University, United States; New York State Psychiatric Institute, United States
| | - Helen Mayberg
- Depression Task Force, Hope for Depression Research Foundation, New York, NY 10019, United States; Emory University, United States
| | - Bruce McEwen
- Depression Task Force, Hope for Depression Research Foundation, New York, NY 10019, United States; Rockefeller University, United States
| | - Michael J Meaney
- Depression Task Force, Hope for Depression Research Foundation, New York, NY 10019, United States; McGill University, United States; Singapore Institute for Clinical Science, Singapore
| | - Eric J Nestler
- Depression Task Force, Hope for Depression Research Foundation, New York, NY 10019, United States; Icahn School of Medicine at Mount Sinai, United States.
| |
Collapse
|
286
|
Neuromodulation Using Optogenetics and Related Technologies. Neuromodulation 2018. [DOI: 10.1016/b978-0-12-805353-9.00035-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
287
|
In Vivo Fiber Photometry Reveals Signature of Future Stress Susceptibility in Nucleus Accumbens. Neuropsychopharmacology 2018; 43:255-263. [PMID: 28589967 PMCID: PMC5729554 DOI: 10.1038/npp.2017.122] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/19/2017] [Accepted: 05/23/2017] [Indexed: 12/12/2022]
Abstract
Recognizing why chronic stress causes only a subset of individuals to become depressed is critical to understanding depression on a basic level and, also, to developing treatments that increase resilience. Stress-induced alterations in the activity of reward-related brain regions, such as the nucleus accumbens (NAc), are linked to the pathophysiology of depression. However, it has been difficult to determine if differences in stress susceptibility are pre-existing or merely an effect of chronic stress. The NAc consists largely of medium spiny neurons (MSNs), distinguished by their predominant expression of either D1 or D2 dopamine receptors. Mice that develop depressive-like symptoms after chronic social defeat stress show distinct changes in the activity of these two cell subtypes. Until now it has not been possible to determine whether such effects are merely a consequence of stress or in fact precede stress and, thus, have utility in pre-identifying stress-susceptible individuals. The goal of this study was to define a cell-type specific signature of stress susceptibility and resilience. Using fiber photometry calcium imaging, we recorded calcium transients in NAc D1- and D2-MSNs in awake behaving mice and found that D1-MSN activity is a predictive marker of depression susceptibility: prior to stress, mice that will later become resilient had increased baseline D1- MSN activity, and increased calcium transients specific to social interaction. Differences in D2- MSN activity were not specific to social interaction. Our findings identify a pre-existing mechanism of stress-induced susceptibility, creating the potential to target preventative interventions to the most relevant populations.
Collapse
|
288
|
Carrillo-Roa T, Labermaier C, Weber P, Herzog DP, Lareau C, Santarelli S, Wagner KV, Rex-Haffner M, Harbich D, Scharf SH, Nemeroff CB, Dunlop BW, Craighead WE, Mayberg HS, Schmidt MV, Uhr M, Holsboer F, Sillaber I, Binder EB, Müller MB. Common genes associated with antidepressant response in mouse and man identify key role of glucocorticoid receptor sensitivity. PLoS Biol 2017; 15:e2002690. [PMID: 29283992 PMCID: PMC5746203 DOI: 10.1371/journal.pbio.2002690] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 11/21/2017] [Indexed: 12/29/2022] Open
Abstract
Response to antidepressant treatment in major depressive disorder (MDD) cannot be predicted currently, leading to uncertainty in medication selection, increasing costs, and prolonged suffering for many patients. Despite tremendous efforts in identifying response-associated genes in large genome-wide association studies, the results have been fairly modest, underlining the need to establish conceptually novel strategies. For the identification of transcriptome signatures that can distinguish between treatment responders and nonresponders, we herein submit a novel animal experimental approach focusing on extreme phenotypes. We utilized the large variance in response to antidepressant treatment occurring in DBA/2J mice, enabling sample stratification into subpopulations of good and poor treatment responders to delineate response-associated signature transcript profiles in peripheral blood samples. As a proof of concept, we translated our murine data to the transcriptome data of a clinically relevant human cohort. A cluster of 259 differentially regulated genes was identified when peripheral transcriptome profiles of good and poor treatment responders were compared in the murine model. Differences in expression profiles from baseline to week 12 of the human orthologues selected on the basis of the murine transcript signature allowed prediction of response status with an accuracy of 76% in the patient population. Finally, we show that glucocorticoid receptor (GR)-regulated genes are significantly enriched in this cluster of antidepressant-response genes. Our findings point to the involvement of GR sensitivity as a potential key mechanism shaping response to antidepressant treatment and support the hypothesis that antidepressants could stimulate resilience-promoting molecular mechanisms. Our data highlight the suitability of an appropriate animal experimental approach for the discovery of treatment response-associated pathways across species. Major depression is the second leading cause of disability worldwide. However, only one-third of patients with depression benefit from the first antidepressant compound they are prescribed. It is a fundamental problem that the outcomes of individual antidepressant treatments are still highly unpredictable. In clinical studies, discovery of biomarkers for antidepressant response is hampered by confounding factors such as the heterogeneity of the disease phenotype and additional environmental factors, e.g., previous life events and different schedules of psychopharmacological treatment, which reduce the power to detect true response biomarkers. To overcome some of these limitations, we have established a conceptually novel approach that allows the selection of extreme phenotypes in an antidepressant-responsive mouse strain. In the first step, we identify signatures in the transcriptome of peripheral blood associated with responses following stratification into good and poor treatment responders. As proof of concept, we translate the murine data to a population of depressed patients. We show that differences in expression profiles from baseline to week 12 of the human orthologues predict response status in patients. We finally provide evidence that sensitivity of the glucocorticoid receptor could be a potential key mechanism shaping response to antidepressant treatment.
Collapse
Affiliation(s)
- Tania Carrillo-Roa
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | | | - Peter Weber
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - David P. Herzog
- Department of Psychiatry and Psychotherapy & German Resilience Center (DRZ), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Caleb Lareau
- Department of Biostatistics, Harvard University, Boston, Massachusetts, United States of America
| | - Sara Santarelli
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Klaus V. Wagner
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Monika Rex-Haffner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Daniela Harbich
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | | | - Charles B. Nemeroff
- Department of Psychiatry and Behavioral Sciences, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Boadie W. Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - W. Edward Craighead
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Psychology, Emory University, Atlanta, Georgia, United States of America
| | - Helen S. Mayberg
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mathias V. Schmidt
- 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
| | | | | | - Elisabeth B. Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Marianne B. Müller
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy & German Resilience Center (DRZ), Johannes Gutenberg University Medical Center, Mainz, Germany
- * E-mail:
| |
Collapse
|
289
|
Dendrosomal nanocurcumin prevents morphine self-administration behavior in rats despite CA1 damage. Behav Pharmacol 2017; 28:681-689. [DOI: 10.1097/fbp.0000000000000291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
290
|
Bath KG, Russo SJ, Pleil KE, Wohleb ES, Duman RS, Radley JJ. Circuit and synaptic mechanisms of repeated stress: Perspectives from differing contexts, duration, and development. Neurobiol Stress 2017; 7:137-151. [PMID: 29276735 PMCID: PMC5736942 DOI: 10.1016/j.ynstr.2017.05.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/03/2017] [Accepted: 05/04/2017] [Indexed: 12/12/2022] Open
Abstract
The current review is meant to synthesize research presented as part of a symposium at the 2016 Neurobiology of Stress workshop in Irvine California. The focus of the symposium was "Stress and the Synapse: New Concepts and Methods" and featured the work of several junior investigators. The presentations focused on the impact of various forms of stress (altered maternal care, binge alcohol drinking, chronic social defeat, and chronic unpredictable stress) on synaptic function, neurodevelopment, and behavioral outcomes. One of the goals of the symposium was to highlight the mechanisms accounting for how the nervous system responds to stress and their impact on outcome measures with converging effects on the development of pathological behavior. Dr. Kevin Bath's presentation focused on the impact of disruptions in early maternal care and its impact on the timing of hippocampus maturation in mice, finding that this form of stress drove accelerated synaptic and behavioral maturation, and contributed to the later emergence of risk for cognitive and emotional disturbance. Dr. Scott Russo highlighted the impact of chronic social defeat stress in adolescent mice on the development and plasticity of reward circuity, with a focus on glutamatergic development in the nucleus accumbens and mesolimbic dopamine system, and the implications of these changes for disruptions in social and hedonic response, key processes disturbed in depressive pathology. Dr. Kristen Pleil described synaptic changes in the bed nuclei of the stria terminalis that underlie the behavioral consequences of allostatic load produced by repeated cycles of alcohol binge drinking and withdrawal. Dr. Eric Wohleb and Dr. Ron Duman provided new data associating decreased mammalian target of rapamycin (mTOR) signaling and neurobiological changes in the synapses in response to chronic unpredictable stress, and highlighted the potential for the novel antidepressant ketamine to rescue synaptic and behavioral effects. In aggregate, these presentations showcased how divergent perspectives provide new insights into the ways in which stress impacts circuit development and function, with implications for understanding emergence of affective pathology.
Collapse
Affiliation(s)
- Kevin G. Bath
- Department of Cognitive Linguistic and Psychological Sciences, Brown University, Providence, RI 02912, United States
| | - Scott J. Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Kristen E. Pleil
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, United States
| | - Eric S. Wohleb
- Department of Psychiatry, University of Cincinnati College of Medicine, Cincinnati, OH 45237, United States
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06508, United States
| | - Ronald S. Duman
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06508, United States
| | - Jason J. Radley
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, United States
| |
Collapse
|
291
|
Adolescence and Reward: Making Sense of Neural and Behavioral Changes Amid the Chaos. J Neurosci 2017; 37:10855-10866. [PMID: 29118215 DOI: 10.1523/jneurosci.1834-17.2017] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/28/2017] [Accepted: 09/28/2017] [Indexed: 11/21/2022] Open
Abstract
Adolescence is a time of significant neural and behavioral change with remarkable development in social, emotional, and cognitive skills. It is also a time of increased exploration and risk-taking (e.g., drug use). Many of these changes are thought to be the result of increased reward-value coupled with an underdeveloped inhibitory control, and thus a hypersensitivity to reward. Perturbations during adolescence can alter the developmental trajectory of the brain, resulting in long-term alterations in reward-associated behaviors. This review highlights recent developments in our understanding of how neural circuits, pubertal hormones, and environmental factors contribute to adolescent-typical reward-associated behaviors with a particular focus on sex differences, the medial prefrontal cortex, social reward, social isolation, and drug use. We then introduce a new approach that makes use of natural adaptations of seasonally breeding species to investigate the role of pubertal hormones in adolescent development. This research has only begun to parse out contributions of the many neural, endocrine, and environmental changes to the heightened reward sensitivity and increased vulnerability to mental health disorders that characterize this life stage.
Collapse
|
292
|
Nasca C, Bigio B, Zelli D, de Angelis P, Lau T, Okamoto M, Soya H, Ni J, Brichta L, Greengard P, Neve RL, Lee FS, McEwen BS. Role of the Astroglial Glutamate Exchanger xCT in Ventral Hippocampus in Resilience to Stress. Neuron 2017; 96:402-413.e5. [PMID: 29024663 DOI: 10.1016/j.neuron.2017.09.020] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 08/10/2017] [Accepted: 09/13/2017] [Indexed: 12/23/2022]
Abstract
We demonstrate that stress differentially regulates glutamate homeostasis in the dorsal and ventral hippocampus and identify a role for the astroglial xCT in ventral dentate gyrus (vDG) in stress and antidepressant responses. We provide an RNA-seq roadmap for the stress-sensitive vDG. The transcription factor REST binds to xCT promoter in co-occupancy with the epigenetic marker H3K27ac to regulate expression of xCT, which is also reduced in a genetic mouse model of inherent susceptibility to depressive-like behavior. Pharmacologically, modulating histone acetylation with acetyl-L-carnitine (LAC) or acetyl-N-cysteine (NAC) rapidly increases xCT and activates a network with mGlu2 receptors to prime an enhanced glutamate homeostasis that promotes both pro-resilient and antidepressant-like responses. Pharmacological xCT blockage counteracts NAC prophylactic effects. GFAP+-Cre-dependent overexpression of xCT in vDG mimics pharmacological actions in promoting resilience. This work establishes a mechanism by which vDG protection leads to stress resilience and antidepressant responses via epigenetic programming of an xCT-mGlu2 network.
Collapse
Affiliation(s)
- Carla Nasca
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA.
| | - Benedetta Bigio
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Danielle Zelli
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Paolo de Angelis
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Timothy Lau
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Masahiro Okamoto
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA; Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki 305-8574, Japan
| | - Hideyo Soya
- Department of Sports Neuroscience, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, Tennodai, Tsukuba, Ibaraki 305-8574, Japan; Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki 305-8574, Japan
| | - Jason Ni
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Lars Brichta
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Francis S Lee
- Sackler Institute for Developmental Psychobiology, Department of Psychiatry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
293
|
Ostroumov A, Dani JA. Convergent Neuronal Plasticity and Metaplasticity Mechanisms of Stress, Nicotine, and Alcohol. Annu Rev Pharmacol Toxicol 2017; 58:547-566. [PMID: 28977763 DOI: 10.1146/annurev-pharmtox-010617-052735] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stress and tobacco smoking are risk factors for alcoholism, but the underlying neural mechanisms are not well understood. Although stress, nicotine, and alcohol have broad, individual effects in the brain, some of their actions converge onto the same mechanisms and circuits. Stress and nicotine augment alcohol-related behaviors, in part via modulation of alcohol-evoked neuronal plasticity and metaplasticity mechanisms. Stress modulates alcohol-evoked plasticity via the release of signaling molecules that influence synaptic transmission. Nicotine also activates some of the same signaling molecules, cells, and circuits, producing a convergence of both stress and nicotine onto common plasticity mechanisms that influence alcohol self-administration. We describe several forms of alcohol-induced plasticity, including classic Hebbian plasticity at glutamatergic synapses, and we highlight less appreciated forms, such as non-Hebbian and GABAergic synaptic plasticity. Risk factors such as stress and nicotine initiate lasting neural changes that modify subsequent alcohol-induced synaptic plasticity and increase the vulnerability to alcohol addiction.
Collapse
Affiliation(s)
- Alexey Ostroumov
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, Philadelphia, Pennsylvania 19104, USA; ,
| | - John A Dani
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, Philadelphia, Pennsylvania 19104, USA; ,
| |
Collapse
|
294
|
Millan EZ, Kim HA, Janak PH. Optogenetic activation of amygdala projections to nucleus accumbens can arrest conditioned and unconditioned alcohol consummatory behavior. Neuroscience 2017; 360:106-117. [PMID: 28757250 PMCID: PMC5752133 DOI: 10.1016/j.neuroscience.2017.07.044] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/13/2017] [Accepted: 07/19/2017] [Indexed: 11/25/2022]
Abstract
Following a Pavlovian pairing procedure, alcohol-paired cues come to elicit behavioral responses that lead to alcohol consumption. Here we used an optogenetic approach to activate basolateral amygdala (BLA) axonal terminals targeting the shell of nucleus accumbens (AcbSh) and investigated a possible influence over cue-conditioned alcohol seeking and alcohol drinking, based on the demonstrated roles of these areas in behavioral responding to Pavlovian cues and in feeding behavior. Rats were trained to anticipate alcohol or sucrose following the onset of a discrete conditioned stimulus (CS). Channelrhodopsin-mediated activation of the BLA-to-AcbSh pathway concurrent with each CS disrupted cued alcohol seeking. Activation of the same pathway caused rapid cessation of alcohol drinking from a sipper tube. Neither effect was accompanied by an overall change in locomotion. Finally, the suppressive effect of photoactivation on cued-triggered seeking was also evidenced in animals trained with sucrose. Together these findings suggest that photoactivation of BLA terminals in the AcbSh can override the conditioned motivational properties of reward-predictive cues as well as unconditioned consummatory responses necessary for alcohol drinking. The findings provide evidence for a limbic-striatal influence over motivated behavior for orally consumed rewards, including alcohol.
Collapse
Affiliation(s)
- E Zayra Millan
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore MD 21218, United States.
| | - H Amy Kim
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore MD 21218, United States
| | - Patricia H Janak
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore MD 21218, United States; Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore MD 21205, United States.
| |
Collapse
|
295
|
Montagrin A, Saiote C, Schiller D. The social hippocampus. Hippocampus 2017; 28:672-679. [DOI: 10.1002/hipo.22797] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 08/22/2017] [Accepted: 08/22/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Alison Montagrin
- Department of Psychiatry, Department of Neuroscience; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai; New York New York 10029
| | - Catarina Saiote
- Department of Psychiatry, Department of Neuroscience; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai; New York New York 10029
| | - Daniela Schiller
- Department of Psychiatry, Department of Neuroscience; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai; New York New York 10029
| |
Collapse
|
296
|
Temporal profiling of depression vulnerability in a preclinical model of sustained depression. Sci Rep 2017; 7:8570. [PMID: 28819243 PMCID: PMC5561121 DOI: 10.1038/s41598-017-06984-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/22/2017] [Indexed: 11/08/2022] Open
Abstract
Major Depression is a prevalent mental disorder that is characterized by negative mood and reduced motivation, and frequently results in social withdrawal and memory-related deficits. Repeated stressors, such as adverse life events, increase the risk for development of the disorder. Consequently, individual variability in stress response greatly weighs on depression-vulnerability and -resilience. Here, we employed the social defeat-induced persistent stress (SDPS) paradigm to identify depression-prone individuals and to examine the temporal development of depression in the months following exposure to brief defeat stress. Male Wistar rats were socially defeated (5 defeat episodes) and single-housed for a prolonged period of time (~24 weeks). We assessed the emergence of a sustained depressive-like state by repeatedly evaluating social motivation (social approach avoidance) and spatial memory (object place recognition) in SDPS rats during the isolation period. Individual variability in the effects of SDPS yielded two extreme subpopulations: an SDPS-prone group that showed gradual affective and cognitive deterioration in terms of social approach and memory retention, and a SDPS-resilient group that did not develop this phenotype. Notably, in SDPS-prone individuals, the affective deficits preceded later cognitive impairments, providing a novel temporal profile of the development of pathology in this preclinical model of sustained depression.
Collapse
|
297
|
Pathway- and Cell-Specific Kappa-Opioid Receptor Modulation of Excitation-Inhibition Balance Differentially Gates D1 and D2 Accumbens Neuron Activity. Neuron 2017; 93:147-163. [PMID: 28056342 DOI: 10.1016/j.neuron.2016.12.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/14/2016] [Accepted: 12/02/2016] [Indexed: 12/31/2022]
Abstract
Endogenous dynorphin signaling via the kappa-opioid receptor (KOR) in the nucleus accumbens (NAcc) powerfully mediates negative affective states and stress reactivity. Excitatory inputs from the hippocampus and amygdala play a fundamental role in shaping the activity of both NAcc D1 and D2 MSNs, which encode positive and negative motivational valences, respectively. However, a circuit-based mechanism by which KOR modulation of excitation-inhibition balance modifies D1 and D2 MSN activity is lacking. Here, we provide a comprehensive synaptic framework wherein presynaptic KOR inhibition decreases the excitatory drive of D1 MSN activity by the amygdala, but not the hippocampus. Conversely, presynaptic inhibition by KORs of inhibitory synapses on D2 MSNs enhances integration of excitatory drive by the amygdala and hippocampus. In conclusion, we describe a circuit-based mechanism showing differential gating of afferent control of D1 and D2 MSN activity by KORs in a pathway-specific manner.
Collapse
|
298
|
Hierarchical Status Predicts Behavioral Vulnerability and Nucleus Accumbens Metabolic Profile Following Chronic Social Defeat Stress. Curr Biol 2017; 27:2202-2210.e4. [PMID: 28712571 DOI: 10.1016/j.cub.2017.06.027] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/09/2017] [Accepted: 06/09/2017] [Indexed: 11/21/2022]
Abstract
Extensive data highlight the existence of major differences in individuals' susceptibility to stress [1-4]. While genetic factors [5, 6] and exposure to early life stress [7, 8] are key components for such neurobehavioral diversity, intriguing observations revealed individual differences in response to stress in inbred mice [9-12]. This raised the possibility that other factors might be critical in stress vulnerability. A key challenge in the field is to identify non-invasively risk factors for vulnerability to stress. Here, we investigated whether behavioral factors, emerging from preexisting dominance hierarchies, could predict vulnerability to chronic stress [9, 13-16]. We applied a chronic social defeat stress (CSDS) model of depression in C57BL/6J mice to investigate the predictive power of hierarchical status to pinpoint which individuals will exhibit susceptibility to CSDS. Given that the high social status of dominant mice would be the one particularly challenged by CSDS, we predicted and found that dominant individuals were the ones showing a strong susceptibility profile as indicated by strong social avoidance following CSDS, while subordinate mice were not affected. Data from 1H-NMR spectroscopy revealed that the metabolic profile in the nucleus accumbens (NAc) relates to social status and vulnerability to stress. Under basal conditions, subordinates show lower levels of energy-related metabolites compared to dominants. In subordinates, but not dominants, levels of these metabolites were increased after exposure to CSDS. To the best of our knowledge, this is the first study that identifies non-invasively the origin of behavioral risk factors predictive of stress-induced depression-like behaviors associated with metabolic changes.
Collapse
|
299
|
Knowland D, Lilascharoen V, Pacia CP, Shin S, Wang EHJ, Lim BK. Distinct Ventral Pallidal Neural Populations Mediate Separate Symptoms of Depression. Cell 2017; 170:284-297.e18. [PMID: 28689640 PMCID: PMC5621481 DOI: 10.1016/j.cell.2017.06.015] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 04/28/2017] [Accepted: 06/08/2017] [Indexed: 02/07/2023]
Abstract
Major depressive disorder (MDD) patients display a common but often variable set of symptoms making successful, sustained treatment difficult to achieve. Separate depressive symptoms may be encoded by differential changes in distinct circuits in the brain, yet how discrete circuits underlie behavioral subsets of depression and how they adapt in response to stress has not been addressed. We identify two discrete circuits of parvalbumin-positive (PV) neurons in the ventral pallidum (VP) projecting to either the lateral habenula or ventral tegmental area contributing to depression. We find that these populations undergo different electrophysiological adaptations in response to social defeat stress, which are normalized by antidepressant treatment. Furthermore, manipulation of each population mediates either social withdrawal or behavioral despair, but not both. We propose that distinct components of the VP PV circuit can subserve related, yet separate depressive-like phenotypes in mice, which could ultimately provide a platform for symptom-specific treatments of depression.
Collapse
Affiliation(s)
- Daniel Knowland
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA USA; Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA USA
| | - Varoth Lilascharoen
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA USA
| | - Christopher Pham Pacia
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA USA
| | - Sora Shin
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA USA
| | - Eric Hou-Jen Wang
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA USA
| | - Byung Kook Lim
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA USA; Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA USA.
| |
Collapse
|
300
|
Abstract
Major depressive disorder (MDD) is a chronic and potentially life threatening illness that carries a staggering global burden. Characterized by depressed mood, MDD is often difficult to diagnose and treat owing to heterogeneity of syndrome and complex etiology. Contemporary antidepressant treatments are based on improved monoamine-based formulations from serendipitous discoveries made > 60 years ago. Novel antidepressant treatments are necessary, as roughly half of patients using available antidepressants do not see long-term remission of depressive symptoms. Current development of treatment options focuses on generating efficacious antidepressants, identifying depression-related neural substrates, and better understanding the pathophysiological mechanisms of depression. Recent insight into the brain's mesocorticolimbic circuitry from animal models of depression underscores the importance of ionic mechanisms in neuronal homeostasis and dysregulation, and substantial evidence highlights a potential role for ion channels in mediating depression-related excitability changes. In particular, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are essential regulators of neuronal excitability. In this review, we describe seminal research on HCN channels in the prefrontal cortex and hippocampus in stress and depression-related behaviors, and highlight substantial evidence within the ventral tegmental area supporting the development of novel therapeutics targeting HCN channels in MDD. We argue that methods targeting the activity of reward-related brain areas have significant potential as superior treatments for depression.
Collapse
Affiliation(s)
- Stacy M Ku
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|