1
|
Buenrostro-Jáuregui MH, Muñóz-Sánchez S, Rojas-Hernández J, Alonso-Orozco AI, Vega-Flores G, Tapia-de-Jesús A, Leal-Galicia P. A Comprehensive Overview of Stress, Resilience, and Neuroplasticity Mechanisms. Int J Mol Sci 2025; 26:3028. [PMID: 40243691 PMCID: PMC11988468 DOI: 10.3390/ijms26073028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Stress is a core concept in the mental health field that expands upon the seminal definition of stress as an acute response to the disruption of homeostasis. Stress is a complex process that involves both environmental challenges and the triggering of internal responses and impacts physiological, psychological, and behavioral systems. The capacity of the human brain to cope with stress is particularly crucial in early life, when neurodevelopment is highly plastic. Early-life stress (ELS), defined as exposure to severe chronic stress during sensitive periods of development, has been shown to cause lasting changes in brain structure and function. However, not all individuals exposed to ELS develop pathological outcomes, suggesting the presence of resilience mechanisms: adaptive processes that allow an individual to cope with adverse situations while maintaining psychological and neurobiological health. The aim of this review was to synthesize recent advances in the understanding of the neuroplasticity mechanisms underlying resilience to ELS. We discussed the neurobiological pathways implicated in stress response and adaptation, including the roles of neurogenesis, synaptic plasticity, and neural circuit remodeling. By focusing on the interplay between stress-induced neuroplastic changes and resilience mechanisms, we aimed to provide insights into potential therapeutic targets for stress-related psychopathology.
Collapse
Affiliation(s)
- Mario Humberto Buenrostro-Jáuregui
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
| | - Sinuhé Muñóz-Sánchez
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
| | - Jorge Rojas-Hernández
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
| | - Adriana Ixel Alonso-Orozco
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
- Facultad de Psicología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - German Vega-Flores
- Ciencias de la Salud, Universidad Internacional de Valencia, 46002 Valencia, Spain;
- Educación, Universidad Internacional de La Rioja, 26006 Logroño, Spain
| | - Alejandro Tapia-de-Jesús
- Departamento de Salud, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico;
| | - Perla Leal-Galicia
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
| |
Collapse
|
2
|
Demin KA, Kolesnikova TO, Galstyan DS, Krotova NA, Ilyin NP, Derzhavina KA, Seredinskaya M, Nerush M, Pushkareva SA, Masharsky A, de Abreu MS, Kalueff AV. The Utility of Prolonged Chronic Unpredictable Stress to Study the Effects of Chronic Fluoxetine, Eicosapentaenoic Acid, and Lipopolysaccharide on Anxiety-Like Behavior and Hippocampal Transcriptomic Responses in Male Rats. J Neurosci Res 2025; 103:e70025. [PMID: 39907099 DOI: 10.1002/jnr.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 12/05/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025]
Abstract
Chronic stress is a common trigger of multiple neuropsychiatric illnesses. Animal models are widely used to study stress-induced brain disorders and their interplay with neuroinflammation and other neuroimmune processes. Here, we apply the prolonged 12-week chronic unpredictable stress (PCUS) model to examine rat behavioral and hippocampal transcriptomic responses to stress and to chronic 4-week treatment with a classical antidepressant fluoxetine, an anti-inflammatory agent eicosapentaenoic acid (EPA), a pro-inflammatory agent lipopolysaccharide and their combinations. Overall, PCUS evoked anxiety-like behavioral phenotype in rats, corrected by chronic fluoxetine (alone or combined with other drugs), and EPA. PCUS also evoked pronounced transcriptomic responses in rat hippocampi, involving > 200 differentially expressed genes. While pharmacological manipulations did not affect hippocampal gene expression markedly, Gpr6, Drd2 and Adora2a were downregulated in stressed rats treated with fluoxetine, EPA and fluoxetine + EPA, suggesting their respective protein products (G protein-coupled receptor 6, dopamine D2 receptor and adenosine A2A receptor) as potential evolutionarily conserved targets under chronic stress. Overall, these findings support the validity of rat PCUS paradigm as a useful model to study stress-related anxiety pathogenesis, and call for further research probing how various conventional and novel drugs may (co)modulate behavioral and neurotranscriptomic biomarkers of chronic stress.
Collapse
Affiliation(s)
- Konstantin A Demin
- Almazov National Medical Research Centre, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Tatiana O Kolesnikova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Neuroscience Program, Sirius University of Science and Technology, Sochi, Russia
| | - David S Galstyan
- Almazov National Medical Research Centre, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Natalia A Krotova
- Almazov National Medical Research Centre, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Nikita P Ilyin
- Almazov National Medical Research Centre, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | | | - Maria Seredinskaya
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Maria Nerush
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Sofia A Pushkareva
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Alexey Masharsky
- Core Facility Centre for Molecular and Cell Technologies, St. Petersburg State University, St. Petersburg, Russia
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
- Western Caspian University, Baku, Azerbaijan
| | - Allan V Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| |
Collapse
|
3
|
von Ziegler LM, Roessler FK, Sturman O, Waag R, Privitera M, Duss SN, O'Connor EC, Bohacek J. Analysis of behavioral flow resolves latent phenotypes. Nat Methods 2024; 21:2376-2387. [PMID: 39533008 PMCID: PMC11621029 DOI: 10.1038/s41592-024-02500-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
The accurate detection and quantification of rodent behavior forms a cornerstone of basic biomedical research. Current data-driven approaches, which segment free exploratory behavior into clusters, suffer from low statistical power due to multiple testing, exhibit poor transferability across experiments and fail to exploit the rich behavioral profiles of individual animals. Here we introduce a pipeline to capture each animal's behavioral flow, yielding a single metric based on all observed transitions between clusters. By stabilizing these clusters through machine learning, we ensure data transferability, while dimensionality reduction techniques facilitate detailed analysis of individual animals. We provide a large dataset of 771 behavior recordings of freely moving mice-including stress exposures, pharmacological and brain circuit interventions-to identify hidden treatment effects, reveal subtle variations on the level of individual animals and detect brain processes underlying specific interventions. Our pipeline, compatible with popular clustering methods, substantially enhances statistical power and enables predictions of an animal's future behavior.
Collapse
Affiliation(s)
- Lukas M von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Fabienne K Roessler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Oliver Sturman
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
- ETH 3R Hub, ETH Zurich, Zurich, Switzerland
| | - Rebecca Waag
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Mattia Privitera
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Sian N Duss
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Eoin C O'Connor
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, Basel, Switzerland
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland.
- ETH 3R Hub, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
4
|
Beck DW, Heaton CN, Davila LD, Rakocevic LI, Drammis SM, Tyulmankov D, Vara P, Giri A, Umashankar Beck S, Zhang Q, Pokojovy M, Negishi K, Batson SA, Salcido AA, Reyes NF, Macias AY, Ibanez-Alcala RJ, Hossain SB, Waller GL, O'Dell LE, Moschak TM, Goosens KA, Friedman A. Model of a striatal circuit exploring biological mechanisms underlying decision-making during normal and disordered states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605535. [PMID: 39211231 PMCID: PMC11361035 DOI: 10.1101/2024.07.29.605535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Decision-making requires continuous adaptation to internal and external contexts. Changes in decision-making are reliable transdiagnostic symptoms of neuropsychiatric disorders. We created a computational model demonstrating how the striosome compartment of the striatum constructs a mathematical space for decision-making computations depending on context, and how the matrix compartment defines action value depending on the space. The model explains multiple experimental results and unifies other theories like reward prediction error, roles of the direct versus indirect pathways, and roles of the striosome versus matrix, under one framework. We also found, through new analyses, that striosome and matrix neurons increase their synchrony during difficult tasks, caused by a necessary increase in dimensionality of the space. The model makes testable predictions about individual differences in disorder susceptibility, decision-making symptoms shared among neuropsychiatric disorders, and differences in neuropsychiatric disorder symptom presentation. The model reframes the role of the striosomal circuit in neuroeconomic and disorder-affected decision-making. Highlights Striosomes prioritize decision-related data used by matrix to set action values. Striosomes and matrix have different roles in the direct and indirect pathways. Abnormal information organization/valuation alters disorder presentation. Variance in data prioritization may explain individual differences in disorders. eTOC Beck et al. developed a computational model of how a striatal circuit functions during decision-making. The model unifies and extends theories about the direct versus indirect pathways. It further suggests how aberrant circuit function underlies decision-making phenomena observed in neuropsychiatric disorders.
Collapse
|
5
|
Talani G, Biggio F, Gorule AA, Licheri V, Saolini E, Colombo D, Sarigu G, Petrella M, Vedele F, Biggio G, Sanna E. Sex-dependent changes of hippocampal synaptic plasticity and cognitive performance in C57BL/6J mice exposed to neonatal repeated maternal separation. Neuropharmacology 2023; 222:109301. [PMID: 36336069 DOI: 10.1016/j.neuropharm.2022.109301] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
The repeated maternal separation (RMS) is a useful experimental model useful in rodents to study the long-term influence of early-life stress on brain neurophysiology. We here investigated the influence of RMS exposure on hippocampal inhibitory and excitatory synaptic transmission, long-term synaptic plasticity and the related potential alterations in learning and memory performance in adult male and female C57Bl/6J mice. Mice were separated daily from their dam for 360 min, from postnatal day 2 (PND2) to PND17, and experiments were performed at PND 60. Patch-clamp recordings in hippocampal CA1 pyramidal neurons revealed a significant enhancement of GABAergic miniature IPSC (mIPSC) frequency, and a decrease in the amplitude of glutamatergic mEPSCs in male mice exposed to RMS. Only a slight but significant reduction in the amplitude of GABAergic mIPSCs was observed in females exposed to RMS compared to the relative controls. A marked increase in long-term depression (LTD) at CA3-CA1 glutamatergic synapses and in the response to the CB1r agonist win55,212 were detected in RMS male, but not female mice. An impaired spatial memory and a reduced preference for novelty was observed in males exposed to RMS but not in females. A single injection of β-ethynyl estradiol at PND2, prevented the changes observed in RMS male mice, suggesting that estrogens may play a protective role early in life against the exposure to stressful conditions. Our findings strengthen the idea of a sex-dependent influence of RMS on long-lasting modifications in synaptic transmission, effects that may be relevant for cognitive performance.
Collapse
Affiliation(s)
- Giuseppe Talani
- CNR Institute of Neuroscience, National Research Council, Monserrato, Italy.
| | - Francesca Biggio
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Ashish Avinash Gorule
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Valentina Licheri
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Eleonora Saolini
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Daniele Colombo
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Gabriele Sarigu
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Michele Petrella
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Francescangelo Vedele
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Giovanni Biggio
- CNR Institute of Neuroscience, National Research Council, Monserrato, Italy; Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Enrico Sanna
- CNR Institute of Neuroscience, National Research Council, Monserrato, Italy; Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| |
Collapse
|
6
|
Demin KA, Krotova NA, Ilyin NP, Galstyan DS, Kolesnikova TO, Strekalova T, de Abreu MS, Petersen EV, Zabegalov KN, Kalueff AV. Evolutionarily conserved gene expression patterns for affective disorders revealed using cross-species brain transcriptomic analyses in humans, rats and zebrafish. Sci Rep 2022; 12:20836. [PMID: 36460699 PMCID: PMC9718822 DOI: 10.1038/s41598-022-22688-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
Widespread, debilitating and often treatment-resistant, depression and other stress-related neuropsychiatric disorders represent an urgent unmet biomedical and societal problem. Although animal models of these disorders are commonly used to study stress pathogenesis, they are often difficult to translate across species into valuable and meaningful clinically relevant data. To address this problem, here we utilized several cross-species/cross-taxon approaches to identify potential evolutionarily conserved differentially expressed genes and their sets. We also assessed enrichment of these genes for transcription factors DNA-binding sites down- and up- stream from their genetic sequences. For this, we compared our own RNA-seq brain transcriptomic data obtained from chronically stressed rats and zebrafish with publicly available human transcriptomic data for patients with major depression and their respective healthy control groups. Utilizing these data from the three species, we next analyzed their differential gene expression, gene set enrichment and protein-protein interaction networks, combined with validated tools for data pooling. This approach allowed us to identify several key brain proteins (GRIA1, DLG1, CDH1, THRB, PLCG2, NGEF, IKZF1 and FEZF2) as promising, evolutionarily conserved and shared affective 'hub' protein targets, as well as to propose a novel gene set that may be used to further study affective pathogenesis. Overall, these approaches may advance cross-species brain transcriptomic analyses, and call for further cross-species studies into putative shared molecular mechanisms of affective pathogenesis.
Collapse
Affiliation(s)
- Konstantin A Demin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
| | - Nataliya A Krotova
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Nikita P Ilyin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | | | | | | | | | | | - Allan V Kalueff
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia.
- Institute of Neurosciences and Medicine, Novosibirsk, Russia.
- Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
7
|
Cahill S, Chandola T, Hager R. Genetic Variants Associated With Resilience in Human and Animal Studies. Front Psychiatry 2022; 13:840120. [PMID: 35669264 PMCID: PMC9163442 DOI: 10.3389/fpsyt.2022.840120] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
Resilience is broadly defined as the ability to maintain or regain functioning in the face of adversity and is influenced by both environmental and genetic factors. The identification of specific genetic factors and their biological pathways underpinning resilient functioning can help in the identification of common key factors, but heterogeneities in the operationalisation of resilience have hampered advances. We conducted a systematic review of genetic variants associated with resilience to enable the identification of general resilience mechanisms. We adopted broad inclusion criteria for the definition of resilience to capture both human and animal model studies, which use a wide range of resilience definitions and measure very different outcomes. Analyzing 158 studies, we found 71 candidate genes associated with resilience. OPRM1 (Opioid receptor mu 1), NPY (neuropeptide Y), CACNA1C (calcium voltage-gated channel subunit alpha1 C), DCC (deleted in colorectal carcinoma), and FKBP5 (FKBP prolyl isomerase 5) had both animal and human variants associated with resilience, supporting the idea of shared biological pathways. Further, for OPRM1, OXTR (oxytocin receptor), CRHR1 (corticotropin-releasing hormone receptor 1), COMT (catechol-O-methyltransferase), BDNF (brain-derived neurotrophic factor), APOE (apolipoprotein E), and SLC6A4 (solute carrier family 6 member 4), the same allele was associated with resilience across divergent resilience definitions, which suggests these genes may therefore provide a starting point for further research examining commonality in resilience pathways.
Collapse
Affiliation(s)
- Stephanie Cahill
- Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Faculty of Humanities, Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
| | - Tarani Chandola
- Faculty of Humanities, Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
- Methods Hub, Department of Sociology, Faculty of Social Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Reinmar Hager
- Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
8
|
Kositsyn YMHB, Volgin AD, de Abreu MS, Demin KA, Zabegalov KN, Maslov GO, Petersen EV, Kolesnikova TO, Strekalova T, Kalueff AV. Towards translational modeling of behavioral despair and its treatment in zebrafish. Behav Brain Res 2022; 430:113906. [PMID: 35489477 DOI: 10.1016/j.bbr.2022.113906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/03/2022] [Accepted: 04/24/2022] [Indexed: 11/26/2022]
Abstract
Depression is a widespread and severely debilitating neuropsychiatric disorder whose key clinical symptoms include low mood, anhedonia and despair (the inability or unwillingness to overcome stressors). Experimental animal models are widely used to improve our mechanistic understanding of depression pathogenesis, and to develop novel antidepressant therapies. In rodents, various experimental models of 'behavioral despair' have already been developed and rigorously validated. Complementing rodent studies, the zebrafish (Danio rerio) is emerging as a powerful model organism to assess pathobiological mechanisms of depression and other related affective disorders. Here, we critically discuss the developing potential and important translational implications of zebrafish models for studying despair and its mechanisms, and the utility of such aquatic models for antidepressant drug screening.
Collapse
Affiliation(s)
- Yuriy M H B Kositsyn
- School of Pharmacy, Southwest University, Chongqing, China; Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia; Sirius University of Science and Technology, Sochi, Russia
| | - Andrew D Volgin
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia; Sirius University of Science and Technology, Sochi, Russia
| | - Murilo S de Abreu
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia; Bioscience Institute, University of Passo Fundo, Passo Fundo, RS, Brazil; Sirius University of Science and Technology, Sochi, Russia.
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medcial Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Granov Russian Scientific Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Sirius University of Science and Technology, Sochi, Russia
| | | | - Gleb O Maslov
- Ural Federal University, Ekaterinburg, Russia; Sirius University of Science and Technology, Sochi, Russia
| | | | | | - Tatiana Strekalova
- University of Maastricht, Maastricht, Netherlands; Sirius University of Science and Technology, Sochi, Russia
| | - Allan V Kalueff
- Ural Federal University, Ekaterinburg, Russia; University of Maastricht, Maastricht, Netherlands; Sirius University of Science and Technology, Sochi, Russia.
| |
Collapse
|
9
|
Gao Y, Rodríguez LV. The Effect of Chronic Psychological Stress on Lower Urinary Tract Function: An Animal Model Perspective. Front Physiol 2022; 13:818993. [PMID: 35388285 PMCID: PMC8978557 DOI: 10.3389/fphys.2022.818993] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/18/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic psychological stress can affect urinary function and exacerbate lower urinary tract (LUT) dysfunction (LUTD), particularly in patients with overactive bladder (OAB) or interstitial cystitis–bladder pain syndrome (IC/BPS). An increasing amount of evidence has highlighted the close relationship between chronic stress and LUTD, while the exact mechanisms underlying it remain unknown. The application of stress-related animal models has provided powerful tools to explore the effect of chronic stress on LUT function. We systematically reviewed recent findings and identified stress-related animal models. Among them, the most widely used was water avoidance stress (WAS), followed by social stress, early life stress (ELS), repeated variable stress (RVS), chronic variable stress (CVS), intermittent restraint stress (IRS), and others. Different types of chronic stress condition the induction of relatively distinguished changes at multiple levels of the micturition pathway. The voiding phenotypes, underlying mechanisms, and possible treatments of stress-induced LUTD were discussed together. The advantages and disadvantages of each stress-related animal model were also summarized to determine the better choice. Through the present review, we hope to expand the current knowledge of the pathophysiological basis of stress-induced LUTD and inspire robust therapies with better outcomes.
Collapse
Affiliation(s)
- Yunliang Gao
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Larissa V. Rodríguez
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- *Correspondence: Larissa V. Rodríguez,
| |
Collapse
|
10
|
Zhang J, Kaye AP, Wang J, Girgenti MJ. Transcriptomics of the depressed and PTSD brain. Neurobiol Stress 2021; 15:100408. [PMID: 34703849 PMCID: PMC8524242 DOI: 10.1016/j.ynstr.2021.100408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 12/13/2022] Open
Abstract
Stress is the response of an organism to demands for change, yet excessive or chronic stress contributes to nearly all psychiatric disorders. The advent of high-throughput transcriptomic methods such as single cell RNA sequencing poses new opportunities to understand the neurobiology of stress, yet substantial barriers to understanding stress remain. Stress adaptation is an organismal survival mechanism conserved across all organisms, yet there is an infinity of potential stressful experiences. Unraveling shared and separate transcriptional programs for adapting to stressful experience remains a challenge, despite methodological and analytic advances. Here we review the state of the field focusing on the technologies used to study the transcriptome for the stress neurobiologist, and also attempt to identify central questions about the heterogeneity of stress for those applying transcriptomic approaches. We further explore how postmortem transcriptome studies aided by preclinical animal models are converging on common molecular pathways for adaptation to aversive experience. Finally, we discuss approaches to integrate large genomic datasets with human neuroimaging and other datasets.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Computer Science, University of California- Irvine, Irvine, CA, USA
| | - Alfred P. Kaye
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Jiawei Wang
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Matthew J. Girgenti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- National Center for PTSD, U.S. Department of Veterans Affairs, USA
| |
Collapse
|
11
|
Modulation of behavioral and neurochemical responses of adult zebrafish by fluoxetine, eicosapentaenoic acid and lipopolysaccharide in the prolonged chronic unpredictable stress model. Sci Rep 2021; 11:14289. [PMID: 34253753 PMCID: PMC8275758 DOI: 10.1038/s41598-021-92422-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Long-term recurrent stress is a common cause of neuropsychiatric disorders. Animal models are widely used to study the pathogenesis of stress-related psychiatric disorders. The zebrafish (Danio rerio) is emerging as a powerful tool to study chronic stress and its mechanisms. Here, we developed a prolonged 11-week chronic unpredictable stress (PCUS) model in zebrafish to more fully mimic chronic stress in human populations. We also examined behavioral and neurochemical alterations in zebrafish, and attempted to modulate these states by 3-week treatment with an antidepressant fluoxetine, a neuroprotective omega-3 polyunsaturated fatty acid eicosapentaenoic acid (EPA), a pro-inflammatory endotoxin lipopolysaccharide (LPS), and their combinations. Overall, PCUS induced severe anxiety and elevated norepinephrine levels, whereas fluoxetine (alone or combined with other agents) corrected most of these behavioral deficits. While EPA and LPS alone had little effects on the zebrafish PCUS-induced anxiety behavior, both fluoxetine (alone or in combination) and EPA restored norepinephrine levels, whereas LPS + EPA increased dopamine levels. As these data support the validity of PCUS as an effective tool to study stress-related pathologies in zebrafish, further research is needed into the ability of various conventional and novel treatments to modulate behavioral and neurochemical biomarkers of chronic stress in this model organism.
Collapse
|
12
|
Häusl AS, Brix LM, Hartmann J, Pöhlmann ML, Lopez JP, Menegaz D, Brivio E, Engelhardt C, Roeh S, Bajaj T, Rudolph L, Stoffel R, Hafner K, Goss HM, Reul JMHM, Deussing JM, Eder M, Ressler KJ, Gassen NC, Chen A, Schmidt MV. The co-chaperone Fkbp5 shapes the acute stress response in the paraventricular nucleus of the hypothalamus of male mice. Mol Psychiatry 2021; 26:3060-3076. [PMID: 33649453 PMCID: PMC8505251 DOI: 10.1038/s41380-021-01044-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 01/31/2023]
Abstract
Disturbed activation or regulation of the stress response through the hypothalamic-pituitary-adrenal (HPA) axis is a fundamental component of multiple stress-related diseases, including psychiatric, metabolic, and immune disorders. The FK506 binding protein 51 (FKBP5) is a negative regulator of the glucocorticoid receptor (GR), the main driver of HPA axis regulation, and FKBP5 polymorphisms have been repeatedly linked to stress-related disorders in humans. However, the specific role of Fkbp5 in the paraventricular nucleus of the hypothalamus (PVN) in shaping HPA axis (re)activity remains to be elucidated. We here demonstrate that the deletion of Fkbp5 in Sim1+ neurons dampens the acute stress response and increases GR sensitivity. In contrast, Fkbp5 overexpression in the PVN results in a chronic HPA axis over-activation, and a PVN-specific rescue of Fkbp5 expression in full Fkbp5 KO mice normalizes the HPA axis phenotype. Single-cell RNA sequencing revealed the cell-type-specific expression pattern of Fkbp5 in the PVN and showed that Fkbp5 expression is specifically upregulated in Crh+ neurons after stress. Finally, Crh-specific Fkbp5 overexpression alters Crh neuron activity, but only partially recapitulates the PVN-specific Fkbp5 overexpression phenotype. Together, the data establish the central and cell-type-specific importance of Fkbp5 in the PVN in shaping HPA axis regulation and the acute stress response.
Collapse
Affiliation(s)
- Alexander S Häusl
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Lea M Brix
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Jakob Hartmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Max L Pöhlmann
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Juan-Pablo Lopez
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Danusa Menegaz
- Electrophysiology Core Unit, Max Planck Institute of Psychiatry, Munich, Germany
| | - Elena Brivio
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Clara Engelhardt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Simone Roeh
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Thomas Bajaj
- Department of Psychiatry and Psychotherapy, Bonn Clinical Center, University of Bonn, Bonn, Germany
| | - Lisa Rudolph
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Rainer Stoffel
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Hannah M Goss
- Neuro-Epigenetics Research Group, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Johannes M H M Reul
- Neuro-Epigenetics Research Group, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Jan M Deussing
- Research Group Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Matthias Eder
- Electrophysiology Core Unit, Max Planck Institute of Psychiatry, Munich, Germany
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Nils C Gassen
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Bonn Clinical Center, University of Bonn, Bonn, Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
13
|
Creutzberg KC, Sanson A, Viola TW, Marchisella F, Begni V, Grassi-Oliveira R, Riva MA. Long-lasting effects of prenatal stress on HPA axis and inflammation: A systematic review and multilevel meta-analysis in rodent studies. Neurosci Biobehav Rev 2021; 127:270-283. [PMID: 33951412 DOI: 10.1016/j.neubiorev.2021.04.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/25/2021] [Accepted: 04/27/2021] [Indexed: 12/22/2022]
Abstract
Exposure to prenatal stress (PNS) can lead to long-lasting neurobiological and behavioral consequences for the offspring, which may enhance the susceptibility for mental disorders. The hypothalamus-pituitary-adrenal (HPA) axis and the immune system are two major factors involved in the stress response. Here, we performed a systematic review and meta-analysis of rodent studies that investigated the effects of PNS exposure on the HPA axis and inflammatory cytokines in adult offspring. Our analysis shows that animals exposed to PNS display a consistent increase in peripheral corticosterone (CORT) levels and central corticotrophin-releasing hormone (CRH), while decreased levels of its receptor 2 (CRHR2). Meta-regression revealed that sex and duration of PNS protocol are covariates that moderate these results. There was no significant effect of PNS in glucocorticoid receptor (GR), CRH receptor 1 (CRHR1), pro- and anti-inflammatory cytokines. Our findings suggest that PNS exposure elicits long-lasting effects on the HPA axis function, providing an important tool to investigate in preclinical settings key pathological aspects related to early-life stress exposure. Furthermore, researchers should be aware of the mixed outcomes of PNS on inflammatory markers in the adult brain.
Collapse
Affiliation(s)
- Kerstin Camile Creutzberg
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | - Alice Sanson
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | - Thiago Wendt Viola
- School of Medicine, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul, Avenida Ipiranga 6681, Building 12A, 90619-900, Porto Alegre, RS, Brazil.
| | - Francesca Marchisella
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | - Rodrigo Grassi-Oliveira
- School of Medicine, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul, Avenida Ipiranga 6681, Building 12A, 90619-900, Porto Alegre, RS, Brazil.
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133, Milan, Italy; Biological Psychiatry Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
14
|
Omics Insights into Animal Resilience and Stress Factors. Animals (Basel) 2020; 11:ani11010047. [PMID: 33383711 PMCID: PMC7824193 DOI: 10.3390/ani11010047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/26/2022] Open
Abstract
Resilience is conceived as a dynamic developmental process involving the achievement of positive adaptation within the context of significant adversity. Resilience is not a unique ability but rather a set of capacities of a system put in place to absorb a disturbance and to reorganize while trying to retain the same function, structure, and identity. This review describes the characteristics and the molecular mechanisms of resilience to understand the core elements of resilience and its indicators. The objectives of this review are: (1) to define some of the leading environmental stressors and clarify the mechanism of vulnerability or resilience outcomes; (2) to clarify some of the prominent epigenetic modulations mediating resilience or vulnerability as a stress response; (3) to highlight the neural mechanisms related to stress resilience since the central nervous system is a highly dynamic structure characterized by an everlasting plasticity feature, which therefore has the opportunity to modify resilience. The review aims to introduce the reader to the concept of resilience seen as an ability acquired in life and not only inherited from birth.
Collapse
|
15
|
Miretti S, Lecchi C, Ceciliani F, Baratta M. MicroRNAs as Biomarkers for Animal Health and Welfare in Livestock. Front Vet Sci 2020; 7:578193. [PMID: 33392281 PMCID: PMC7775535 DOI: 10.3389/fvets.2020.578193] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/30/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small and highly conserved non-coding RNA molecules that orchestrate a wide range of biological processes through the post-transcriptional regulation of gene expression. An intriguing aspect in identifying these molecules as biomarkers is derived from their role in cell-to-cell communication, their active secretion from cells into the extracellular environment, their high stability in body fluids, and their ease of collection. All these features confer on miRNAs the potential to become a non-invasive tool to score animal welfare. There is growing interest in the importance of miRNAs as biomarkers for assessing the welfare of livestock during metabolic, environmental, and management stress, particularly in ruminants, pigs, and poultry. This review provides an overview of the current knowledge regarding the potential use of tissue and/or circulating miRNAs as biomarkers for the assessment of the health and welfare status in these livestock species.
Collapse
Affiliation(s)
- Silvia Miretti
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Cristina Lecchi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Fabrizio Ceciliani
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Mario Baratta
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| |
Collapse
|
16
|
Understanding complex dynamics of behavioral, neurochemical and transcriptomic changes induced by prolonged chronic unpredictable stress in zebrafish. Sci Rep 2020; 10:19981. [PMID: 33203921 PMCID: PMC7673038 DOI: 10.1038/s41598-020-75855-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Stress-related neuropsychiatric disorders are widespread, debilitating and often treatment-resistant illnesses that represent an urgent unmet biomedical problem. Animal models of these disorders are widely used to study stress pathogenesis. A more recent and historically less utilized model organism, the zebrafish (Danio rerio), is a valuable tool in stress neuroscience research. Utilizing the 5-week chronic unpredictable stress (CUS) model, here we examined brain transcriptomic profiles and complex dynamic behavioral stress responses, as well as neurochemical alterations in adult zebrafish and their correction by chronic antidepressant, fluoxetine, treatment. Overall, CUS induced complex neurochemical and behavioral alterations in zebrafish, including stable anxiety-like behaviors and serotonin metabolism deficits. Chronic fluoxetine (0.1 mg/L for 11 days) rescued most of the observed behavioral and neurochemical responses. Finally, whole-genome brain transcriptomic analyses revealed altered expression of various CNS genes (partially rescued by chronic fluoxetine), including inflammation-, ubiquitin- and arrestin-related genes. Collectively, this supports zebrafish as a valuable translational tool to study stress-related pathogenesis, whose anxiety and serotonergic deficits parallel rodent and clinical studies, and genomic analyses implicate neuroinflammation, structural neuronal remodeling and arrestin/ubiquitin pathways in both stress pathogenesis and its potential therapy.
Collapse
|
17
|
Jiménez Vásquez FDJ, Guerrero DM, Osornio MR, Rubio Osornio MDC, Suárez SO, Retana-Márquez S. Decreased serotonin content and release in the ventral hippocampus of prenatally stressed male rats in response to forced swim test. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
18
|
Kõiv K, Vares M, Kroon C, Metelitsa M, Tiitsaar K, Laugus K, Jaako K, Harro J. Effect of chronic variable stress on sensitization to amphetamine in high and low sucrose-consuming rats. J Psychopharmacol 2019; 33:1512-1523. [PMID: 31208275 DOI: 10.1177/0269881119856000] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Individual vulnerability to stress manifests in the interaction of innate properties and environment. There is a growing interest in the individual variability in vulnerability to stress and how it contributes to the development of psychiatric disorders. Intake of palatable substances is often measured in animal models. We have previously demonstrated that the consumption of sucrose solution is a stable trait in rats. AIMS The present study aimed to compare the sensitivity of rats with high vs low liquid sucrose consumption to chronic variable stress and the stress effect on behavioural sensitization to amphetamine. METHODS Male Wistar rats were subjected to a chronic stress regimen and subsequent repeated treatment with amphetamine (1 mg/kg, intraperitoneally). Fifty-kHz ultrasonic vocalizations, locomotor activity and stereotypies were measured. RESULTS In no-stress baseline conditions, the behavioural response to acute amphetamine was similar in rats with high vs low sucrose consumption. Prior chronic stress potentiated the effect of amphetamine only in rats with high sucrose consumption. Behavioural sensitization to repeated administration of amphetamine was observed in non-stressed rats with lower sucrose preference, but not in the respective stressed group that had increased monoamine turnover in the nucleus accumbens. In contrast, in rats with high sucrose preference the amphetamine sensitization effect was prevalent in stressed rats, but not in non-stressed animals. INTERPRETATION Chronic stress can change the psychostimulant effect but this depends on the inherent reward sensitivity of the animal. Trait-wise, sucrose intake reflects vulnerability to chronic stress and may interact with the development of addiction.
Collapse
Affiliation(s)
- Kadri Kõiv
- Department of Psychology, University of Tartu, Tartu, Estonia
| | - Marten Vares
- Department of Psychology, University of Tartu, Tartu, Estonia
| | - Cristina Kroon
- Department of Psychology, University of Tartu, Tartu, Estonia
| | - Mait Metelitsa
- Department of Psychology, University of Tartu, Tartu, Estonia
| | - Kai Tiitsaar
- Department of Psychology, University of Tartu, Tartu, Estonia
| | - Karita Laugus
- Department of Psychology, University of Tartu, Tartu, Estonia
| | - Külli Jaako
- Department of Pharmacology, University of Tartu, Tartu, Estonia
| | - Jaanus Harro
- Department of Psychology, University of Tartu, Tartu, Estonia
| |
Collapse
|
19
|
Firmino EMS, Kuntze LB, Lagatta DC, Dias DPM, Resstel LBM. Effect of chronic stress on cardiovascular and ventilatory responses activated by both chemoreflex and baroreflex in rats. ACTA ACUST UNITED AC 2019; 222:jeb.204883. [PMID: 31558591 DOI: 10.1242/jeb.204883] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/20/2019] [Indexed: 01/08/2023]
Abstract
Chronic stress results in physiological and somatic changes. It has been recognized as a risk factor for several types of cardiovascular dysfunction and changes in autonomic mechanisms, such as baroreflex and chemoreflex activity. However, the effects of different types of chronic stress on these mechanisms are still poorly understood. Therefore, in the present study, we investigated, in adult male rats, the effect of repeated restraint stress (RRS) or chronic variable stress (CVS) on baroreflex, chemoreflex and heart rate variability in a protocol of 14 days of stress sessions. Exposure to RRS and CVS indicated no changes in the basal level of either arterial pressure or heart rate. However, RRS and CVS were able to attenuate sympathovagal modulation and spontaneous baroreflex gain. Additionally, only RRS was able to increase the power of the low-frequency band of the systolic blood pressure spectrum, as well as the slope of linear regression of baroreflex bradycardic and tachycardic responses induced by vasoactive compounds. Additionally, our study is one of the first to show that exposure to RRS and CVS decreases the magnitude of the pressor response and potentiates respiratory responses to chemoreflex activation, which can trigger cardiovascular and respiratory pathologies. Furthermore, the basal respiratory parameters, such as minute ventilation and tidal volume, were significantly decreased by both protocols of chronic stress. However, only CVS increased the basal respiratory frequency. In this way, the findings of the present study demonstrate the impact of chronic stress in terms of not only depressive-like behavior but also alterations of the autonomic baroreflex responses and cardiocirculatory variability (systolic blood pressure and pulse interval).Our results provide evidence that chronic stress promotes autonomic dysregulation, and impairment of baroreflex, chemoreflex and heart rate variability.
Collapse
Affiliation(s)
- Egidi Mayara Silva Firmino
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14090-090, Brazil
| | - Luciana Bärg Kuntze
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14090-090, Brazil
| | - Davi Campos Lagatta
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14090-090, Brazil
| | | | - Leonardo Barbosa Moraes Resstel
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14090-090, Brazil
| |
Collapse
|
20
|
Abstract
BACKGROUND Stress-related mental disorders are the most prevalent and cost-intensive disorders of our time. On the other hand, the maintenance of mental health despite stressors, i. e. resilience, is a frequent phenomenon. Research on psychological resilience and its underlying mechanisms offers innovative possibilities for health promotion. It requires a consistent understanding of resilience and adequate methods of operationalization. OBJECTIVES Modern concepts of the definition, operationalization and assessment of resilience as well as its implications for study designs in resilience research. MATERIAL AND METHODS Analysis and discussion of current works and expert recommendations for the design of resilience research. RESULTS Resilience research is undergoing a period of transition. Based on a new understanding of resilience as a dynamic and modifiable process, new approaches for operationalization and assessment were proposed. These include, for example, a transdiagnostic approach and the identification of resilience mechanisms, the consideration of stressor exposure in measuring the construct, and longitudinal cohort studies. CONCLUSIONS In the upcoming decades, further profitable findings from current prospective longitudinal studies can be expected. One challenge for future resilience research consists in the continuous dissemination and implementation of the approaches described.
Collapse
|
21
|
Rabelo-da-Ponte FD, Pessoa Gomes JM, Torres NL, Barbosa JIC, de Andrade GM, Macedo D, Ceppi B. Behavioral, affective, and cognitive alterations induced by individual and combined environmental stressors in rats. BRAZILIAN JOURNAL OF PSYCHIATRY 2019; 41:289-296. [PMID: 30892378 PMCID: PMC6804305 DOI: 10.1590/1516-4446-2018-0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 08/01/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To evaluate whether exposing rats to individual or combined environmental stressors triggers endophenotypes related to mood and anxiety disorders, and whether this effect depends on the nature of the behavior (i.e., innate or learned). METHODS We conducted a three-phase experimental protocol. In phase I (baseline), animals subjected to mixed schedule of reinforcement were trained to press a lever with a fixed interval of 1 minute and a limited hold of 3 seconds. On the last day of phase I, an open-field test was performed and the animals were divided into four experimental groups (n=8/group). In phase II (repeated stress), each group was exposed to either hot air blast (HAB), paradoxical sleep deprivation (PSD) or both (HAB+PSD group) on alternate days over a 10-day period. Control group animals were not exposed to stressors. In phase III (post-stress evaluation), behavior was analyzed on the first (short-term effects), third (mid-term effects), and fifth (long-term effects) days after repeated stress. RESULTS The PSD group presented operant hyperactivity, the HAB group presented spontaneous hypoactivity and anxiety, and the HAB+PSD group presented spontaneous hyperactivity, operant hypoactivity, impulsivity, loss of interest, and cognitive impairment. CONCLUSION A combination of environmental stressors (HAB and PSD) may induce endophenotypes related to bipolar disorder.
Collapse
Affiliation(s)
| | | | - Nathércia Lima Torres
- Departamento de Psicologia, Universidade Federal do Ceará (UFC), Fortaleza, CE, Brazil
| | | | - Geanne Matos de Andrade
- Laboratório de Neurociências e Comportamento, Departamento de Fisiologia e Farmacologia, UFC, Fortaleza, CE, Brazil
| | - Danielle Macedo
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Núcleo de Pesquisa e Desenvolvimento de Medicamentos, UFC, Fortaleza, CE, Brazil.,Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Ribeirão Preto, SP, Brazil
| | - Bruno Ceppi
- Laboratório de Neurociências e Comportamento, Departamento de Fisiologia e Farmacologia, UFC, Fortaleza, CE, Brazil
| |
Collapse
|
22
|
Snijders C, Pries LK, Sgammeglia N, Al Jowf G, Youssef NA, de Nijs L, Guloksuz S, Rutten BPF. Resilience Against Traumatic Stress: Current Developments and Future Directions. Front Psychiatry 2018; 9:676. [PMID: 30631285 PMCID: PMC6315131 DOI: 10.3389/fpsyt.2018.00676] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 11/23/2018] [Indexed: 12/22/2022] Open
Abstract
Given the high prevalence of stress-related mental disorders, their impact on person, family, and society and the paucity of treatment options for most of these disorders, there is currently a pressing need for innovative approaches to deal with these issues and enhance well-being. One approach which has received increasing attention over the last decade is to shift our scientific and clinical focus from risk factors for psychopathology to factors promoting resilience and mental well-being. In order to summarize and evaluate the current state of scientific affairs on the biological basis of resilience, we provide an overview of the literature on animal and human studies of resilience. Because resilience can only truly be operationalized through longitudinal data collection and analyses, we focus primarily on longitudinal studies. This review shows that the concept of resilience is currently being operationalized, measured and even defined in widely variable manners, both within animal and human studies. We further provide an overview of existing and new strategies that could help promote resilience and which are proposed to be implemented more often in clinical situations. Finally, we summarize the challenges the field is facing and provide recommendations for future research.
Collapse
Affiliation(s)
- Clara Snijders
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
| | - Lotta-Katrin Pries
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
| | - Noemi Sgammeglia
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
| | - Ghazi Al Jowf
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
- College of Applied Medical Sciences, Department of Public Health, King Faisal University, Al-Ahsa, Saudi Arabia
- European Graduate School of Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Nagy A. Youssef
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Office of Academic Affairs, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Laurence de Nijs
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
| | - Sinan Guloksuz
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Bart P. F. Rutten
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
23
|
Borsook D, Youssef AM, Simons L, Elman I, Eccleston C. When pain gets stuck: the evolution of pain chronification and treatment resistance. Pain 2018; 159:2421-2436. [PMID: 30234696 PMCID: PMC6240430 DOI: 10.1097/j.pain.0000000000001401] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
It is well-recognized that, despite similar pain characteristics, some people with chronic pain recover, whereas others do not. In this review, we discuss possible contributions and interactions of biological, social, and psychological perturbations that underlie the evolution of treatment-resistant chronic pain. Behavior and brain are intimately implicated in the production and maintenance of perception. Our understandings of potential mechanisms that produce or exacerbate persistent pain remain relatively unclear. We provide an overview of these interactions and how differences in relative contribution of dimensions such as stress, age, genetics, environment, and immune responsivity may produce different risk profiles for disease development, pain severity, and chronicity. We propose the concept of "stickiness" as a soubriquet for capturing the multiple influences on the persistence of pain and pain behavior, and their stubborn resistance to therapeutic intervention. We then focus on the neurobiology of reward and aversion to address how alterations in synaptic complexity, neural networks, and systems (eg, opioidergic and dopaminergic) may contribute to pain stickiness. Finally, we propose an integration of the neurobiological with what is known about environmental and social demands on pain behavior and explore treatment approaches based on the nature of the individual's vulnerability to or protection from allostatic load.
Collapse
Affiliation(s)
- David Borsook
- Center for Pain and the Brain, Boston Children’s (BCH), McLean and Massachusetts Hospitals (MGH), Boston MA
- Departments of Anesthesia (BCH), Psychiatry (MGH, McLean) and Radiology (MGH)
| | - Andrew M Youssef
- Center for Pain and the Brain, Boston Children’s (BCH), McLean and Massachusetts Hospitals (MGH), Boston MA
| | - Laura Simons
- Department of Anesthesia, Stanford University, Palo Alto, CA
| | | | - Christopher Eccleston
- Centre for Pain Research, University of Bath, UK
- Department of Clinical and Health Psychology, Ghent University, Belgium
| |
Collapse
|
24
|
Burrage E, Marshall KL, Santanam N, Chantler PD. Cerebrovascular dysfunction with stress and depression. Brain Circ 2018; 4:43-53. [PMID: 30276336 PMCID: PMC6126243 DOI: 10.4103/bc.bc_6_18] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 12/11/2022] Open
Abstract
Maintenance of adequate tissue perfusion through a dense network of cerebral microvessels is critical for the perseveration of normal brain function. Regulation of the cerebral blood flow has to ensure adequate delivery of nutrients and oxygen with moment-to-moment adjustments to avoid both hypo- and hyper-perfusion of the brain tissue. Even mild impairments of cerebral blood flow regulation can have significant implications on brain function. Evidence suggests that chronic stress and depression elicits multifaceted functional impairments to the cerebral microcirculation, which plays a critical role in brain health and the pathogenesis of stress-related cognitive impairment and cerebrovascular events. Identifying the functional and structural changes to the brain that are induced by stress is crucial for achieving a realistic understanding of how related illnesses, which are highly disabling and with a large economic cost, can be managed or reversed. This overview discusses the stress-induced alterations in neurovascular coupling with specific attention to cerebrovascular regulation (endothelial dependent and independent vasomotor function, microvessel density). The pathophysiological consequences of cerebral microvascular dysfunction with stress and depression are explored.
Collapse
Affiliation(s)
- Emily Burrage
- Department of Neuroscience, West Virginia University Rockefeller Neuroscience Institute, Morgantown, WV, USA
| | - Kent L. Marshall
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Nalini Santanam
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Paul D. Chantler
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| |
Collapse
|
25
|
Biggio F, Talani G, Locci V, Pisu M, Boero G, Ciarlo B, Grayson D, Serra M. Low doses of prenatal ethanol exposure and maternal separation alter HPA axis function and ethanol consumption in adult male rats. Neuropharmacology 2018; 131:271-281. [DOI: 10.1016/j.neuropharm.2017.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/26/2017] [Accepted: 12/03/2017] [Indexed: 11/25/2022]
|
26
|
Maternal separation induces long-term effects on monoamines and brain-derived neurotrophic factor levels on the frontal cortex, amygdala, and hippocampus: differential effects after a stress challenge. Behav Pharmacol 2017; 28:545-557. [DOI: 10.1097/fbp.0000000000000324] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Yin X, Guven N, Dietis N. Stress-based animal models of depression: Do we actually know what we are doing? Brain Res 2016; 1652:30-42. [DOI: 10.1016/j.brainres.2016.09.027] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/03/2016] [Accepted: 09/19/2016] [Indexed: 01/10/2023]
|
28
|
Sex, boldness and stress experience affect convict cichlid, Amatitlania nigrofasciata, open field behaviour. Anim Behav 2015. [DOI: 10.1016/j.anbehav.2015.05.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
Golbidi S, Frisbee JC, Laher I. Chronic stress impacts the cardiovascular system: animal models and clinical outcomes. Am J Physiol Heart Circ Physiol 2015; 308:H1476-98. [DOI: 10.1152/ajpheart.00859.2014] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 04/03/2015] [Indexed: 01/01/2023]
Abstract
Psychological stresses are associated with cardiovascular diseases to the extent that cardiovascular diseases are among the most important group of psychosomatic diseases. The longstanding association between stress and cardiovascular disease exists despite a large ambiguity about the underlying mechanisms. An array of possibilities have been proposed including overactivity of the autonomic nervous system and humoral changes, which then converge on endothelial dysfunction that initiates unwanted cardiovascular consequences. We review some of the features of the two most important stress-activated systems, i.e., the humoral and nervous systems, and focus on alterations in endothelial function that could ensue as a result of these changes. Cardiac and hematologic consequences of stress are also addressed briefly. It is likely that activation of the inflammatory cascade in association with oxidative imbalance represents key pathophysiological components of stress-induced cardiovascular changes. We also review some of the commonly used animal models of stress and discuss the cardiovascular outcomes reported in these models of stress. The unique ability of animals for adaptation under stressful conditions lessens the extrapolation of laboratory findings to conditions of human stress. An animal model of unpredictable chronic stress, which applies various stress modules in a random fashion, might be a useful solution to this predicament. The use of stress markers as indicators of stress intensity is also discussed in various models of animal stress and in clinical studies.
Collapse
Affiliation(s)
- Saeid Golbidi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, Canada; and
| | - Jefferson C. Frisbee
- Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center, Morgantown, West Virginia
| | - Ismail Laher
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, Canada; and
| |
Collapse
|
30
|
Christianson JP, Flyer-Adams JG, Drugan RC, Amat J, Daut RA, Foilb AR, Watkins LR, Maier SF. Learned stressor resistance requires extracellular signal-regulated kinase in the prefrontal cortex. Front Behav Neurosci 2014; 8:348. [PMID: 25324750 PMCID: PMC4183187 DOI: 10.3389/fnbeh.2014.00348] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/18/2014] [Indexed: 12/16/2022] Open
Abstract
Behaviorally controllable stressors confer protection from the neurochemical and behavioral consequences of future uncontrollable stressors, a phenomenon termed “behavioral immunization”. Recent data implicate protein synthesis within the ventromedial prefrontal cortex (mPFC) as critical to behavioral immunization. Adult, male Sprague-Dawley rats were exposed to a series of controllable tailshocks and 1 week later to uncontrollable tailshocks, followed 24 h later by social exploration and shuttlebox escape tests. To test the involvement of N-methyl-D-aspartate receptors (NMDARs) and the extracellular signal-regulated kinase (ERK) cascade in behavioral immunization, either D-AP5 or the MEK inhibitor U0126 was injected to the prelimbic (PL) or infralimbic (IL) mPFC prior to controllable stress exposure. Phosphorylated ERK and P70S6K, regulators of transcription and translation, were quantified by Western blot or immunohistochemistry after controllable or uncontrollable tailshocks. Prior controllable stress prevented the social exploration and shuttlebox performance deficits caused by the later uncontrollable stressor, and this effect was blocked by injections of D-AP5 into mPFC. A significant increase in phosphorylated ERK1 and ERK2, but not P70S6K, occurred within the PL and IL in rats exposed to controllable stress, but not to uncontrollable stress. However, U0126 only prevented behavioral immunization when injected to the PL. We provide evidence that NMDAR and ERK dependent signaling within the PL region is required for behavioral immunization, a learned form of stressor resistance.
Collapse
Affiliation(s)
- John P Christianson
- Center for Neuroscience, Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA ; Department of Psychology, Boston College Chestnut Hill, MA, USA
| | - Johanna G Flyer-Adams
- Center for Neuroscience, Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Robert C Drugan
- Department of Psychology, University of New Hampshire Durham, NH, USA
| | - Jose Amat
- Center for Neuroscience, Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Rachel A Daut
- Center for Neuroscience, Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Allison R Foilb
- Department of Psychology, Boston College Chestnut Hill, MA, USA
| | - Linda R Watkins
- Center for Neuroscience, Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Steven F Maier
- Center for Neuroscience, Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| |
Collapse
|
31
|
Boleij H, Willems J, Leijten M, Klooster JV, Lesscher H, Kirchhoff S, Lavrijsen M, Arndt SS, Ohl F. Chronic social stress does not affect behavioural habituation in male CD1 mice. Behav Brain Res 2014; 273:34-44. [DOI: 10.1016/j.bbr.2014.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/07/2014] [Accepted: 07/09/2014] [Indexed: 12/29/2022]
|
32
|
Abstract
The well-replicated observation that many people maintain mental health despite exposure to severe psychological or physical adversity has ignited interest in the mechanisms that protect against stress-related mental illness. Focusing on resilience rather than pathophysiology in many ways represents a paradigm shift in clinical-psychological and psychiatric research that has great potential for the development of new prevention and treatment strategies. More recently, research into resilience also arrived in the neurobiological community, posing nontrivial questions about ecological validity and translatability. Drawing on concepts and findings from transdiagnostic psychiatry, emotion research, and behavioral and cognitive neuroscience, we propose a unified theoretical framework for the neuroscientific study of general resilience mechanisms. The framework is applicable to both animal and human research and supports the design and interpretation of translational studies. The theory emphasizes the causal role of stimulus appraisal (evaluation) processes in the generation of emotional responses, including responses to potential stressors. On this basis, it posits that a positive (non-negative) appraisal style is the key mechanism that protects against the detrimental effects of stress and mediates the effects of other known resilience factors. Appraisal style is shaped by three classes of cognitive processes--positive situation classification, reappraisal, and interference inhibition--that can be investigated at the neural level. Prospects for the future development of resilience research are discussed.
Collapse
|
33
|
Biggio F, Pisu MG, Garau A, Boero G, Locci V, Mostallino MC, Olla P, Utzeri C, Serra M. Maternal separation attenuates the effect of adolescent social isolation on HPA axis responsiveness in adult rats. Eur Neuropsychopharmacol 2014; 24:1152-61. [PMID: 24745548 DOI: 10.1016/j.euroneuro.2014.03.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 03/21/2014] [Accepted: 03/27/2014] [Indexed: 12/21/2022]
Abstract
Adverse early life experiences that occur during childhood and adolescence can have negative impacts on behavior later in life. The main goal of our work was to assess how the association between stressful experiences during neonatal and adolescent periods may influence stress responsiveness and brain plasticity in adult rats. Stressful experiences included maternal separation and social isolation at weaning. Three hours of separation from the pups (3-14 PND) significantly increased frequencies of maternal arched-back nursing and licking-grooming across the first two weeks postpartum. Separation also induced a long-lasting increase in dams blood levels of corticosterone. Maternal separation did not modify brain and plasma allopregnanolone and corticosterone levels in adult offspring, but they demonstrate partial recovery from the reduction induced by social isolation during adolescence. Moreover, the enhancement of corticosterone and allopregnanolone levels induced by foot shock stress in socially isolated animals that were subjected to maternal separation was markedly reduced with respect to that observed in animals that were just socially isolated. All experimental groups showed a significant reduction of BDNF and Arc protein expression in the hippocampus. However, the reduction of BDNF observed in animals that were maternally separated and subjected to social isolation was less significantly pronounced than in animals that were just socially isolated. The results sustained the mismatch hypothesis stating that aversive experiences early in life trigger adaptive processes, thereby rendering an individual to be better adapted to aversive challenges later in life.
Collapse
Affiliation(s)
- F Biggio
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari 09100, Italy
| | - M G Pisu
- C.N.R., Institute of Neuroscience, Cagliari 09100, Italy
| | - A Garau
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari 09100, Italy
| | - G Boero
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari 09100, Italy
| | - V Locci
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari 09100, Italy
| | - M C Mostallino
- Nanobiotechnology Laboratory, University of Cagliari, Cagliari 09100, Italy; C.N.R., Institute of Neuroscience, Cagliari 09100, Italy
| | - P Olla
- Nanobiotechnology Laboratory, University of Cagliari, Cagliari 09100, Italy
| | - C Utzeri
- Nanobiotechnology Laboratory, University of Cagliari, Cagliari 09100, Italy
| | - M Serra
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari 09100, Italy; Center of Excellence for Neurobiology of Dependence, University of Cagliari, Cagliari 09100, Italy.
| |
Collapse
|
34
|
Labermaier C, Kohl C, Hartmann J, Devigny C, Altmann A, Weber P, Arloth J, Quast C, Wagner KV, Scharf SH, Czibere L, Widner-Andrä R, Brenndörfer J, Landgraf R, Hausch F, Jones KA, Müller MB, Uhr M, Holsboer F, Binder EB, Schmidt MV. A polymorphism in the Crhr1 gene determines stress vulnerability in male mice. Endocrinology 2014; 155:2500-10. [PMID: 24773341 DOI: 10.1210/en.2013-1986] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chronic stress is a risk factor for psychiatric disorders but does not necessarily lead to uniform long-term effects on mental health, suggesting modulating factors such as genetic predispositions. Here we address the question whether natural genetic variations in the mouse CRH receptor 1 (Crhr1) locus modulate the effects of adolescent chronic social stress (ACSS) on long-term stress hormone dysregulation in outbred CD1 mice, which allows a better understanding of the currently reported genes × environment interactions of early trauma and CRHR1 in humans. We identified 2 main haplotype variants in the mouse Crhr1 locus that modulate the long-term effects of ACSS on basal hypothalamic-pituitary-adrenal axis activity. This effect is likely mediated by higher levels of CRHR1, because Crhr1 mRNA expression and CRHR1 binding were enhanced in risk haplotype carriers. Furthermore, a CRHR1 receptor antagonist normalized these long-term effects. Deep sequencing of the Crhr1 locus in CD1 mice revealed a large number of linked single-nucleotide polymorphisms with some located in important regulatory regions, similar to the location of human CRHR1 variants implicated in modulating gene × stress exposure interactions. Our data support that the described gene × stress exposure interaction in this animal model is based on naturally occurring genetic variations in the Crhr1 gene associated with enhanced CRHR1-mediated signaling. Our results suggest that patients with a specific genetic predisposition in the CRHR1 gene together with an exposure to chronic stress may benefit from a treatment selectively antagonizing CRHR1 hyperactivity.
Collapse
Affiliation(s)
- Christiana Labermaier
- Max Planck Institute of Psychiatry (C.L., C.K., J.H., C.D., A.A, P.W., J.A., C.Q., K.V.W., S.H.S., L.C., R.W-A., J.B., R.L., F.H., M.U., F.H., E.B.B., M.V.S.), 80804 Munich, Germany; Lundbeck Research USA (K.A.J.), Paramus, New Jersey 07652; and Johannes Gutenberg University Medical Center (M.B.M.), 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Barth L, Sütterlin R, Nenniger M, Vogt KE. Functional differentiation of stem cell-derived neurons from different murine backgrounds. Front Cell Neurosci 2014; 8:49. [PMID: 24600351 PMCID: PMC3929830 DOI: 10.3389/fncel.2014.00049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/03/2014] [Indexed: 01/15/2023] Open
Abstract
Murine stem cell-derived neurons have been used to study a wide variety of neuropsychiatric diseases with a hereditary component, ranging from autism to Alzheimer's. While a significant amount of data on their molecular biology has been generated, there is little data on the physiology of these cultures. Different mouse strains show clear differences in behavioral and other neurobiologically relevant readouts. We have studied the physiology of early differentiation and network formation in neuronal cultures derived from three different mouse embryonic stem cell lines. We have found largely overlapping patterns with some significant differences in the timing of the functional milestones. Neurons from R1 showed the fastest development of intrinsic excitability, while E14Tg2a and J1 were slower. This was also reflected in an earlier appearance of synaptic activity in R1 cultures, while E14Tg2a and J1 were delayed by up to 2 days. In conclusion, stem cells from all backgrounds could be successfully differentiated into functioning neural networks with similar developmental patterns. Differences in the timing of specific milestones, suggest that control cell lines and time-points should be carefully chosen when investigating genetic alterations that lead to subtle deficits in neuronal function.
Collapse
Affiliation(s)
- Lydia Barth
- Department of Neurobiology/Pharmacology, Biozentrum, University of Basel Basel, Switzerland
| | - Rosmarie Sütterlin
- Department of Neurobiology/Pharmacology, Biozentrum, University of Basel Basel, Switzerland
| | - Markus Nenniger
- Department of Neurobiology/Pharmacology, Biozentrum, University of Basel Basel, Switzerland
| | - Kaspar E Vogt
- Department of Neurobiology/Pharmacology, Biozentrum, University of Basel Basel, Switzerland
| |
Collapse
|
36
|
Abstract
Exposure to an uncontrollable stressor elicits a constellation of physiological and behavioral sequel in laboratory rats that often reflect aspects of anxiety and other emotional disruptions. We review evidence suggesting that plasticity within the serotonergic dorsal raphe nucleus (DRN) is critical to the expression of uncontrollable stressor-induced anxiety. Specifically, after uncontrollable stressor exposure subsequent anxiogenic stimuli evoke greater 5-HT release in DRN terminal regions including the amygdala and striatum; and pharmacological blockade of postsynaptic 5-HT(2C) receptors in these regions prevents expression of stressor-induced anxiety. Importantly, the controllability of stress, the presence of safety signals, and a history of exercise mitigate the expression of stressor-induced anxiety. These stress-protective factors appear to involve distinct neural substrates; with stressor controllability requiring the medial prefrontal cortex, safety signals the insular cortex and exercise affecting the 5-HT system directly. Knowledge of the distinct yet converging mechanisms underlying these stress-protective factors could provide insight into novel strategies for the treatment and prevention of stress-related psychiatric disorders.
Collapse
|
37
|
Epigenetics and the regulation of stress vulnerability and resilience. Neuroscience 2013; 264:157-70. [PMID: 24333971 DOI: 10.1016/j.neuroscience.2013.12.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/29/2013] [Accepted: 12/03/2013] [Indexed: 12/13/2022]
Abstract
The human brain has a remarkable capacity to adapt to and learn from a wide range of variations in the environment. However, environmental challenges can also precipitate psychiatric disorders in susceptible individuals. Why any given experience should induce one brain to adapt while another is edged toward psychopathology remains poorly understood. Like all aspects of psychological function, both nature (genetics) and nurture (life experience) sculpt the brain's response to stressful stimuli. Here we review how these two influences intersect at the epigenetic regulation of neuronal gene transcription, and we discuss how the regulation of genomic DNA methylation near key stress-response genes may influence psychological susceptibility or resilience to environmental stressors. Our goal is to offer a perspective on the epigenetics of stress responses that works to bridge the gap between the study of this molecular process in animal models and its potential usefulness for understanding stress vulnerabilities in humans.
Collapse
|
38
|
Daskalakis NP, Yehuda R, Diamond DM. Animal models in translational studies of PTSD. Psychoneuroendocrinology 2013; 38:1895-911. [PMID: 23845512 DOI: 10.1016/j.psyneuen.2013.06.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 06/05/2013] [Accepted: 06/05/2013] [Indexed: 01/29/2023]
Abstract
Understanding the neurobiological mechanisms of post-traumatic stress disorder (PTSD) is of vital importance for developing biomarkers and more effective pharmacotherapy for this disorder. The design of bidirectional translational studies addressing all facets of PTSD is needed. Animal models of PTSD are needed not only to capture the complexity of PTSD behavioral characteristics, but also to address experimentally the influence of variety of factors which might determine an individual's vulnerability or resilience to trauma, e.g., genetic predisposition, early-life experience and social support. The current review covers recent translational approaches to bridge the gap between human and animal PTSD research and to create a framework for discovery of biomarkers and novel therapeutics.
Collapse
Affiliation(s)
- Nikolaos P Daskalakis
- Traumatic Stress Studies Division & Laboratory of Molecular Neuropsychiatry, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA; Mental Health Care Center, PTSD Clinical Research Program & Laboratory of Clinical Neuroendocrinology and Neurochemistry, James J. Peters Veterans Affairs Medical Center, Bronx, USA
| | | | | |
Collapse
|
39
|
Peric T, Comin A, Corazzin M, Montillo M, Cappa A, Campanile G, Prandi A. Short communication: Hair cortisol concentrations in Holstein-Friesian and crossbreed F1 heifers. J Dairy Sci 2013; 96:3023-7. [DOI: 10.3168/jds.2012-6151] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 01/25/2013] [Indexed: 12/18/2022]
|
40
|
Zovkic IB, Meadows JP, Kaas GA, Sweatt JD. Interindividual Variability in Stress Susceptibility: A Role for Epigenetic Mechanisms in PTSD. Front Psychiatry 2013; 4:60. [PMID: 23805109 PMCID: PMC3693073 DOI: 10.3389/fpsyt.2013.00060] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/11/2013] [Indexed: 12/13/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric condition characterized by intrusive and persistent memories of a psychologically traumatic event that leads to significant functional and social impairment in affected individuals. The molecular bases underlying persistent outcomes of a transient traumatic event have remained elusive for many years, but recent studies in rodents have implicated epigenetic modifications of chromatin structure and DNA methylation as fundamental mechanisms for the induction and stabilization of fear memory. In addition to mediating adaptations to traumatic events that ultimately cause PTSD, epigenetic mechanisms are also involved in establishing individual differences in PTSD risk and resilience by mediating long-lasting effects of genes and early environment on adult function and behavior. In this review, we discuss the current evidence for epigenetic regulation of PTSD in human studies and in animal models and comment on ways in which these models can be expanded. In addition, we identify key outstanding questions in the study of epigenetic mechanisms of PTSD in the context of rapidly evolving technologies that are constantly updating and adjusting our understanding of epigenetic modifications and their functional roles. Finally, we discuss the potential application of epigenetic approaches in identifying markers of risk and resilience that can be utilized to promote early intervention and develop therapeutic strategies to combat PTSD after symptom onset.
Collapse
Affiliation(s)
- Iva B Zovkic
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham , Birmingham, AL , USA
| | | | | | | |
Collapse
|