1
|
Talani G, Biggio F, Mostallino MC, Batzu E, Biggio G, Sanna E. Sex-specific changes in voluntary alcohol consumption and nucleus accumbens synaptic plasticity in C57BL/6J mice exposed to neonatal maternal separation. Neuropharmacology 2024; 262:110212. [PMID: 39521040 DOI: 10.1016/j.neuropharm.2024.110212] [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: 08/04/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
The long-term influence of early-life stress on brain neurophysiology has been extensively investigated using different animal models. Among these, repeated maternal separation (RMS) in rodents is one of the most commonly adopted. In this study, we elucidated the long-lasting effects of exposure to postnatal RMS in C57BL/6J adult mice on voluntary alcohol consumption and nucleus accumbens (NAc) neurophysiology. Mice were separated from their dam for 360 min daily from postnatal day 2 (PND2) to PND17, and experiments were then performed in adult (PND60) animals. In addition, as recent evidence showed that circulating estrogens may play a protective role against stress effects on brain function, including the organization and activation of neuronal structures, we also evaluated the effect of a single injection of β-estradiol 3-benzoate (EB) at PND2, which is known to disrupt male sex differentiation, in male RMS mice. The RMS exposure was associated with an increased voluntary alcohol consumption and preference in male mice, but not in female mice or male mice treated with a single injection of EB. Patch clamp experiments conducted in NAc medium spiny neurons (MSNs) revealed that excitatory but not inhibitory synaptic transmission and long-term plasticity of glutamatergic synapses were significantly impaired in male but not in female mice exposed to the RMS protocol. This effect was again prevented in RMS male mice treated with EB. Our findings strengthen the idea of a sex-dependent influence of early-life stress on long-lasting modifications in synaptic transmission and plasticity in brain areas involved in goal-directed behavior and alcohol intake.
Collapse
Affiliation(s)
- Giuseppe Talani
- CNR Institute of Neuroscience, National Research Council, 09042, Monserrato, CA, Italy.
| | - Francesca Biggio
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042, Monserrato, CA, Italy
| | | | - Elisabetta Batzu
- CNR Institute of Neuroscience, National Research Council, 09042, Monserrato, CA, Italy
| | - Giovanni Biggio
- CNR Institute of Neuroscience, National Research Council, 09042, Monserrato, CA, Italy; Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042, Monserrato, CA, Italy
| | - Enrico Sanna
- CNR Institute of Neuroscience, National Research Council, 09042, Monserrato, CA, Italy; Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042, Monserrato, CA, Italy
| |
Collapse
|
2
|
Ma YN, Zhang CC, Sun YX, Liu X, Li XX, Wang H, Wang T, Wang XD, Su YA, Li JT, Si TM. Dorsal CA1 NECTIN3 Reduction Mediates Early-Life Stress-Induced Object Recognition Memory Deficits in Adolescent Female Mice. Neurosci Bull 2024:10.1007/s12264-024-01305-z. [PMID: 39395912 DOI: 10.1007/s12264-024-01305-z] [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: 01/10/2024] [Accepted: 04/28/2024] [Indexed: 10/14/2024] Open
Abstract
Early-life stress (ES) leads to cognitive dysfunction in female adolescents, but the underlying neural mechanisms remain elusive. Recent evidence suggests that the cell adhesion molecules NECTIN1 and NECTIN3 play a role in cognition and ES-related cognitive deficits in male rodents. In this study, we aimed to investigate whether and how nectins contribute to ES-induced cognitive dysfunction in female adolescents. Applying the well-established limited bedding and nesting material paradigm, we found that ES impairs recognition memory, suppresses prefrontal NECTIN1 and hippocampal NECTIN3 expression, and upregulates corticotropin-releasing hormone (Crh) and its receptor 1 (Crhr1) mRNA levels in the hippocampus of adolescent female mice. Genetic experiments revealed that the reduction of dorsal CA1 (dCA1) NECTIN3 mediates ES-induced object recognition memory deficits, as knocking down dCA1 NECTIN3 impaired animals' performance in the novel object recognition task, while overexpression of dCA1 NECTIN3 successfully reversed the ES-induced deficits. Notably, prefrontal NECTIN1 knockdown did not result in significant cognitive impairments. Furthermore, acute systemic administration of antalarmin, a CRHR1 antagonist, upregulated hippocampal NECTIN3 levels and rescued object and spatial memory deficits in stressed mice. Our findings underscore the critical role of dCA1 NECTIN3 in mediating ES-induced object recognition memory deficits in adolescent female mice, highlighting it as a potential therapeutic target for stress-related psychiatric disorders in women.
Collapse
Affiliation(s)
- Yu-Nu Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Chen-Chen Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ya-Xin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xiao Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xue-Xin Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Han Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ting Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
| |
Collapse
|
3
|
Cody CR, de la Villarmois EA, Fernandez AM, Lardizabal J, McKnight C, Tseng K, Brenhouse HC. Effects of early life adversity and adolescent basolateral amygdala activity on corticolimbic connectivity and anxiety behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586708. [PMID: 38853948 PMCID: PMC11160567 DOI: 10.1101/2024.03.26.586708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Early postnatal development of corticolimbic circuitry is shaped by the environment and is vulnerable to early life challenges. Prior work has shown that early life adversity (ELA) leads to hyperinnervation of glutamatergic basolateral amygdala (BLA) projections to the prefrontal cortex (PFC) in adolescence. While hyperinnervation is associated with later-life anxiety behaviors, the physiological changes underpinning corticolimbic and behavioral impacts of ELA are not understood. We tested whether postsynaptic BLA-driven PFC activity is enhanced in ELA-exposed animals, using the maternal separation (MS) model of ELA. PFC local-field potential following BLA stimulation was facilitated in MS-exposed adolescents. Since ELA increases activity of the early-developing BLA, while the PFC exhibits protracted development, we further examined impacts of glutamatergic BLA activity during early adolescence on later-life PFC innervation and heightened anxiety. In early adolescence, MS-exposed animals exhibited decreased anxiety-like behavior, and acute adolescent BLA inhibition induced behaviors that resembled those of MS animals. To examine long-lasting impacts of adolescent BLA activity on innervation, BLA-originating axonal boutons in the PFC were quantified in late adolescence after early adolescent BLA inhibition. We further tested whether late adolescent BLA-PFC changes were associated with anxious reactivity expressed as heightened acoustic startle responses. MS rearing increased BLA-PFC innervation and threat reactivity in late adolescence, however early adolescent BLA inhibition was insufficient to prevent MS effects, suggesting that earlier BLA activity or post-synaptic receptor rearrangement in the PFC drives altered innervation. Taken together, these findings highlight both pre- and postsynaptic changes in the adolescent BLA-PFC circuit following ELA.
Collapse
Affiliation(s)
- Caitlyn R Cody
- Psychology Department, Northeastern University, Boston MA 02115
| | | | | | | | - Chaney McKnight
- Psychology Department, Northeastern University, Boston MA 02115
| | - Kuei Tseng
- Department of Anatomy and Cell Biology, University of Illinois, Chicago IL 60612
| | | |
Collapse
|
4
|
Nazzari S, Grumi S, Mambretti F, Villa M, Giorda R, Bordoni M, Pansarasa O, Borgatti R, Provenzi L. Sex-dimorphic pathways in the associations between maternal trait anxiety, infant BDNF methylation, and negative emotionality. Dev Psychopathol 2024; 36:908-918. [PMID: 36855816 DOI: 10.1017/s0954579423000172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Maternal antenatal anxiety is an emerging risk factor for child emotional development. Both sex and epigenetic mechanisms, such as DNA methylation, may contribute to the embedding of maternal distress into emotional outcomes. Here, we investigated sex-dependent patterns in the association between antenatal maternal trait anxiety, methylation of the brain-derived neurotrophic factor gene (BDNF DNAm), and infant negative emotionality (NE). Mother-infant dyads (N = 276) were recruited at delivery. Maternal trait anxiety, as a marker of antenatal chronic stress exposure, was assessed soon after delivery using the Stait-Trait Anxiety Inventory (STAI-Y). Infants' BDNF DNAm at birth was assessed in 11 CpG sites in buccal cells whereas infants' NE was assessed at 3 (N = 225) and 6 months (N = 189) using the Infant Behavior Questionnaire-Revised (IBQ-R). Hierarchical linear analyses showed that higher maternal antenatal anxiety was associated with greater 6-month-olds' NE. Furthermore, maternal antenatal anxiety predicted greater infants' BDNF DNAm in five CpG sites in males but not in females. Higher methylation at these sites was associated with greater 3-to-6-month NE increase, independently of infants' sex. Maternal antenatal anxiety emerged as a risk factor for infant's NE. BDNF DNAm might mediate this effect in males. These results may inform the development of strategies to promote mothers and infants' emotional well-being.
Collapse
Affiliation(s)
- Sarah Nazzari
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Serena Grumi
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabiana Mambretti
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Marco Villa
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Roberto Giorda
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Matteo Bordoni
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Orietta Pansarasa
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Renato Borgatti
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Livio Provenzi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
5
|
Burenkova OV, Grigorenko EL. The role of epigenetic mechanisms in the long-term effects of early-life adversity and mother-infant relationship on physiology and behavior of offspring in laboratory rats and mice. Dev Psychobiol 2024; 66:e22479. [PMID: 38470450 PMCID: PMC10959231 DOI: 10.1002/dev.22479] [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: 09/15/2023] [Revised: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/13/2024]
Abstract
Maternal care during the early postnatal period of altricial mammals is a key factor in the survival and adaptation of offspring to environmental conditions. Natural variations in maternal care and experimental manipulations with maternal-child relationships modeling early-life adversity (ELA) in laboratory rats and mice have a strong long-term influence on the physiology and behavior of offspring in rats and mice. This literature review is devoted to the latest research on the role of epigenetic mechanisms in these effects of ELA and mother-infant relationship, with a focus on the regulation of hypothalamic-pituitary-adrenal axis and brain-derived neurotrophic factor. An important part of this review is dedicated to pharmacological interventions and epigenetic editing as tools for studying the causal role of epigenetic mechanisms in the development of physiological and behavioral profiles. A special section of the manuscript will discuss the translational potential of the discussed research.
Collapse
Affiliation(s)
- Olga V. Burenkova
- Department of Psychology, University of Houston, Houston, Texas, USA
- Texas Institute for Measurement, Evaluation, and Statistics, University of Houston, Houston, Texas, USA
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | - Elena L. Grigorenko
- Department of Psychology, University of Houston, Houston, Texas, USA
- Texas Institute for Measurement, Evaluation, and Statistics, University of Houston, Houston, Texas, USA
- Center for Cognitive Sciences, Sirius University of Science and Technology, Sochi, Russia
- Departments of Molecular and Human Genetics and Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Child Study Center, Yale University, New Haven, Connecticut, USA
- Research Administration, Moscow State University for Psychology and Education, Moscow, Russia
| |
Collapse
|
6
|
Li P, Yan Z. An epigenetic mechanism of social isolation stress in adolescent female mice. Neurobiol Stress 2024; 29:100601. [PMID: 38213831 PMCID: PMC10776430 DOI: 10.1016/j.ynstr.2023.100601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024] Open
Abstract
Social isolation during adolescence can increase the risk of mental disorders. Epigenetic changes induced by chronic social isolation may serve as a mechanism underlying emotional disturbances. To test this, we exposed female mice to a post-weaning 6-week social isolation (SI) stress. We found the significantly increased methylation of histone H3 at lysine 9 (H3K9), a histone mark linked to gene repression, as well as the increased H3K9 methyltransferases SUV39H1 and SETDB1, in prefrontal cortex (PFC) of SI females. To find out potential downstream genes affected by this epigenetic alteration, we examined genes linked to neuronal and synaptic functions. Activity-dependent genes, including Arc, c-Fos and Npas4, were significantly reduced in PFC of SI females, correlated with the increased H3K9me2 occupancy around Arc enhancer. Treatment of SI females with UNC0642, a selective inhibitor of H3K9 methylation, significantly attenuated the anxiety-like behavior and elevated Arc expression. These results have revealed an epigenetic mechanism and intervention avenue for anxiety induced by chronic social isolation.
Collapse
Affiliation(s)
- Pei Li
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| |
Collapse
|
7
|
De Santa F, Strimpakos G, Marchetti N, Gargari G, Torcinaro A, Arioli S, Mora D, Petrella C, Farioli-Vecchioli S. Effect of a multi-strain probiotic mixture consumption on anxiety and depression symptoms induced in adult mice by postnatal maternal separation. MICROBIOME 2024; 12:29. [PMID: 38369490 PMCID: PMC10875865 DOI: 10.1186/s40168-024-01752-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 01/04/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Intestinal microbial composition not only affects the health of the gut but also influences centrally mediated systems involved in mood, through the "gut-brain" axis, a bidirectional communication between gut microbiota and the brain. In this context, the modulation of intestinal microbiota and its metabolites through the administration of probiotics seems to represent a very promising approach in the treatment of the central nervous system alterations. Early postnatal life is a critical period during which the brain undergoes profound and essential modulations in terms of maturation and plasticity. Maternal separation (MS), i.e., the disruption of the mother-pup interaction, represents a pivotal paradigm in the study of stress-related mood disorders, by inducing persistent changes in the immune system, inflammatory processes, and emotional behavior in adult mammals. RESULTS We conducted experiments to investigate whether sustained consumption of a multi-strain probiotic formulation by adult male mice could mitigate the effects of maternal separation. Our data demonstrated that the treatment with probiotics was able to totally reverse the anxiety- and depressive-like behavior; normalize the neuro-inflammatory state, by restoring the resting state of microglia; and finally induce a proneurogenic effect. Mice subjected to maternal separation showed changes in microbiota composition compared to the control group that resulted in permissive colonization by the administered multi-strain probiotic product. As a consequence, the probiotic treatment also significantly affected the production of SCFA and in particular the level of butyrate. CONCLUSION Gut microbiota and its metabolites mediate the therapeutic action of the probiotic mix on MS-induced brain dysfunctions. Our findings extend the knowledge on the use of probiotics as a therapeutic tool in the presence of alterations of the emotional sphere that significantly impact on gut microbiota composition. Video Abstract.
Collapse
Affiliation(s)
- Francesca De Santa
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
| | - Georgios Strimpakos
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
| | - Nicole Marchetti
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
- Sciences of Nutrition, Aging, Metabolism and Gender Pathologies, Catholic University of Roma, Rome, 00100, Italy
| | - Giorgio Gargari
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Alessio Torcinaro
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
| | - Stefania Arioli
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Diego Mora
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Policlinico Umberto I, Rome, Italy
| | - Stefano Farioli-Vecchioli
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy.
| |
Collapse
|
8
|
Rocks D, Jaric I, Bellia F, Cham H, Greally JM, Suzuki M, Kundakovic M. Early-life stress and ovarian hormones alter transcriptional regulation in the nucleus accumbens resulting in sex-specific responses to cocaine. Cell Rep 2023; 42:113187. [PMID: 37777968 PMCID: PMC10753961 DOI: 10.1016/j.celrep.2023.113187] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/29/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2023] Open
Abstract
Early-life stress and ovarian hormones contribute to increased female vulnerability to cocaine addiction. Here, we reveal molecular substrates in the reward area, the nucleus accumbens, through which these female-specific factors affect immediate and conditioning responses to cocaine. We find shared involvement of X chromosome inactivation-related and estrogen signaling-related gene regulation in enhanced conditioning responses following early-life stress and during the low-estrogenic state in females. Low-estrogenic females respond to acute cocaine by opening neuronal chromatin enriched for the sites of ΔFosB, a transcription factor implicated in chronic cocaine response and addiction. Conversely, high-estrogenic females respond to cocaine by preferential chromatin closing, providing a mechanism for limiting cocaine-driven chromatin and synaptic plasticity. We find that physiological estrogen withdrawal, early-life stress, and absence of one X chromosome all nullify the protective effect of a high-estrogenic state on cocaine conditioning in females. Our findings offer a molecular framework to enable understanding of sex-specific neuronal mechanisms underlying cocaine use disorder.
Collapse
Affiliation(s)
- Devin Rocks
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Ivana Jaric
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Fabio Bellia
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Heining Cham
- Department of Psychology, Fordham University, Bronx, NY, USA
| | - John M Greally
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Masako Suzuki
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Marija Kundakovic
- Department of Biological Sciences, Fordham University, Bronx, NY, USA.
| |
Collapse
|
9
|
Bratzu J, Ciscato M, Pisanu A, Talani G, Frau R, Porcu P, Diana M, Fumagalli F, Romualdi P, Rullo L, Trezza V, Ciccocioppo R, Sanna F, Fattore L. Communal nesting differentially attenuates the impact of pre-weaning social isolation on behavior in male and female rats during adolescence and adulthood. Front Behav Neurosci 2023; 17:1257417. [PMID: 37915532 PMCID: PMC10616881 DOI: 10.3389/fnbeh.2023.1257417] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023] Open
Abstract
Introduction Early social isolation (ESI) disrupts neurodevelopmental processes, potentially leading to long-lasting emotional and cognitive changes in adulthood. Communal nesting (CN), i.e., the sharing of parental responsibilities between multiple individuals in a nest, creates a socially enriching environment known to impact social and anxiety-related behaviors. Methods This study examines the effects of (i) the CN condition and of (ii) ESI during the 3rd week of life (i.e., pre-weaning ESI) on motor, cognitive, and emotional domains during adolescence and adulthood in male and female rats reared in the two different housing conditions, as well as (iii) the potential of CN to mitigate the impact of ESI on offspring. Results We found that in a spontaneous locomotor activity test, females exhibited higher activity levels compared to males. In female groups, adolescents reared in standard housing (SH) condition spent less time in the center of the arena, suggestive of increased anxiety levels, while the CN condition increased the time spent in the center during adolescence, but not adulthood, independently from ESI. The prepulse inhibition (PPI) test showed a reduced PPI in ESI adolescent animals of both sexes and in adult males (but not in adult females), with CN restoring PPI in males, but not in adolescent females. Further, in the marble burying test SH-ESI adolescent males exhibited higher marble burying behavior than all other groups, suggestive of obsessive-compulsive traits. CN completely reversed this stress-induced effect. Interestingly, ESI and CN did not have a significant impact on burying behavior in adult animals of both sexes. Discussion Overall, our findings (i) assess the effects of ESI on locomotion, sensorimotor gating, and compulsive-like behaviors, (ii) reveal distinct vulnerabilities of males and females within these domains, and (iii) show how early-life social enrichment may successfully counteract some of the behavioral alterations induced by early-life social stress in a sex-dependent manner. This study strengthens the notion that social experiences during early-life can shape emotional and cognitive outcomes in adulthood, and points to the importance of social enrichment interventions for mitigating the negative effects of early social stress on neurodevelopment.
Collapse
Affiliation(s)
- Jessica Bratzu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Maria Ciscato
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Augusta Pisanu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Giuseppe Talani
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Marco Diana
- G.Minardi’ Cognitive Neuroscience Laboratory, CPMB Science Department, University of Sassari, Sassari, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, University of Milan, Milan, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Laura Rullo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Viviana Trezza
- Department of Science, University “Roma Tre”, Rome, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Fabrizio Sanna
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Liana Fattore
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| |
Collapse
|
10
|
Uselman TW, Jacobs RE, Bearer EL. Reconfiguration of brain-wide neural activity after early life adversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.10.557058. [PMID: 38328213 PMCID: PMC10849645 DOI: 10.1101/2023.09.10.557058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Early life adversity (ELA) predisposes individuals to both physical and mental disorders lifelong. How ELA affects brain function leading to this vulnerability is under intense investigation. Research has begun to shed light on ELA effects on localized brain regions within defined circuits. However, investigations into brain-wide neural activity that includes multiple localized regions, determines relationships of activity between regions and identifies shifts of activity in response to experiential conditions is necessary. Here, we performed longitudinal manganese-enhanced magnetic resonance imaging (MEMRI) to image the brain in normally reared or ELA-exposed adults. Images were captured in the freely moving home cage condition, and short- and long-term after naturalistic threat. Images were analyzed with new computational methods, including automated segmentation and fractional activation or difference volumes. We found that neural activity was increased after ELA compared to normal rearing in multiple brain regions, some of which are involved in defensive and/or reward circuitry. Widely distributed patterns of neural activity, "brain states", and their dynamics after threat were altered with ELA. Upon acute threat, ELA-mice retained heightened neural activity within many of these regions, and new hyperactive responses emerged in monoaminergic centers of the mid- and hindbrain. Nine days after acute threat, heightened neural activity remained within locus coeruleus and increased within posterior amygdala, ventral hippocampus, and dorso- and ventromedial hypothalamus, while reduced activity emerged within medial prefrontal cortical regions (prelimbic, infralimbic, anterior cingulate). These results reveal that functional imbalances arise between multiple brain-systems which are dependent upon context and cumulative experiences after ELA.
Collapse
Affiliation(s)
- Taylor W Uselman
- University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Russell E Jacobs
- Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033
- California Institute of Technology, Pasadena, CA 91125
| | - Elaine L Bearer
- University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- California Institute of Technology, Pasadena, CA 91125
| |
Collapse
|
11
|
Yuan M, Yang B, Rothschild G, Mann JJ, Sanford LD, Tang X, Huang C, Wang C, Zhang W. Epigenetic regulation in major depression and other stress-related disorders: molecular mechanisms, clinical relevance and therapeutic potential. Signal Transduct Target Ther 2023; 8:309. [PMID: 37644009 PMCID: PMC10465587 DOI: 10.1038/s41392-023-01519-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/14/2023] [Accepted: 05/31/2023] [Indexed: 08/31/2023] Open
Abstract
Major depressive disorder (MDD) is a chronic, generally episodic and debilitating disease that affects an estimated 300 million people worldwide, but its pathogenesis is poorly understood. The heritability estimate of MDD is 30-40%, suggesting that genetics alone do not account for most of the risk of major depression. Another factor known to associate with MDD involves environmental stressors such as childhood adversity and recent life stress. Recent studies have emerged to show that the biological impact of environmental factors in MDD and other stress-related disorders is mediated by a variety of epigenetic modifications. These epigenetic modification alterations contribute to abnormal neuroendocrine responses, neuroplasticity impairment, neurotransmission and neuroglia dysfunction, which are involved in the pathophysiology of MDD. Furthermore, epigenetic marks have been associated with the diagnosis and treatment of MDD. The evaluation of epigenetic modifications holds promise for further understanding of the heterogeneous etiology and complex phenotypes of MDD, and may identify new therapeutic targets. Here, we review preclinical and clinical epigenetic findings, including DNA methylation, histone modification, noncoding RNA, RNA modification, and chromatin remodeling factor in MDD. In addition, we elaborate on the contribution of these epigenetic mechanisms to the pathological trait variability in depression and discuss how such mechanisms can be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Biao Yang
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gerson Rothschild
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Radiology, Columbia University, New York, NY, 10032, USA
| | - Larry D Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuang Wang
- Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Medical Big Data Center, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
12
|
Shin S, Lee S. The impact of environmental factors during maternal separation on the behaviors of adolescent C57BL/6 mice. Front Mol Neurosci 2023; 16:1147951. [PMID: 37293540 PMCID: PMC10244624 DOI: 10.3389/fnmol.2023.1147951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Neonatal maternal separation is a widely used method to construct an early-life stress model in rodents. In this method, pups are separated from their mothers for several hours every day during the first 2 weeks of life, which results in adverse early-life events. It is a known fact that maternal separation can exert a significant impact on the behavior and psychological health, such as anxiety and depression, in adolescent offspring. However, environmental conditions during maternal separation can differ such as the presence of other animals or by placing pups in a different dam. To investigate the differential effects of various conditions of maternal separation on the behavior of adolescent mice, we created the following groups: (1) iMS group: pups were moved to an isolated room with no other adult mice in a nearby cage, (2) eDam group: the pups randomly exchanged their dams, (3) OF group: pups were shifted to another cage with the bedding material containing maternal odor (olfactory stimulation), and (4) MS group: pups were shifted to another vivarium. From postnatal day (PND) 2-20 (i.e., 19 consecutive days), pups were separated from the dam daily for 4 h and exposed to various environments (MS, iMS, eDam, and OF) or were left undisturbed [control (CON) group]. A series of behavioral assessments were conducted to evaluate locomotion, anxiety, recognition, learning, and memory in adolescent offspring. The results showed that neonatal maternal separation led to impaired recognition memory, motor coordination, and motor skill learning across all groups. However, the iMS group exhibited anxiety-like behavior in the elevated plus maze test and enhanced the extinction of fear memory in the auditory fear conditioning test. The OF and eDam groups displayed partially recovered short-term working memory in the Y-maze test but exhibited opposite exploratory behaviors. The OF group spent more time in the center, while the eDam group spent less time. These findings demonstrated that exposure to different environmental conditions during maternal separation causes behavioral alterations in adolescent offspring, providing a potential explanation for the variation in behavioral phenotypes observed in the early-life stress models.
Collapse
|
13
|
Warhaftig G, Almeida D, Turecki G. Early life adversity across different cell- types in the brain. Neurosci Biobehav Rev 2023; 148:105113. [PMID: 36863603 DOI: 10.1016/j.neubiorev.2023.105113] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/13/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023]
Abstract
Early life adversity (ELA)- which includes physical, psychological, emotional, and sexual abuse is one of the most common predictors to diverse psychopathologies later in adulthood. As ELA has a lasting impact on the brain at a developmental stage, recent findings from the field highlighted the specific contributions of different cell types to ELA and their association with long lasting consequences. In this review we will gather recent findings describing morphological, transcriptional and epigenetic alterations within neurons, glia and perineuronal nets and their associated cellular subpopulation. The findings reviewed and summarized here highlight important mechanisms underlying ELA and point to therapeutic approaches for ELA and related psychopathologies later in life.
Collapse
Affiliation(s)
- Gal Warhaftig
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada
| | - Daniel Almeida
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal QC H3A 1A1, Canada.
| |
Collapse
|
14
|
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: 6] [Impact Index Per Article: 6.0] [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
|
15
|
Zühlsdorff K, López-Cruz L, Dutcher EG, Jones JA, Pama C, Sawiak S, Khan S, Milton AL, Robbins TW, Bullmore ET, Dalley JW. Sex-dependent effects of early life stress on reinforcement learning and limbic cortico-striatal functional connectivity. Neurobiol Stress 2023; 22:100507. [PMID: 36505960 PMCID: PMC9731893 DOI: 10.1016/j.ynstr.2022.100507] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Major depressive disorder (MDD) is a stress-related condition hypothesized to involve aberrant reinforcement learning (RL) with positive and negative stimuli. The present study investigated whether repeated early maternal separation (REMS) stress, a procedure widely recognized to cause depression-like behaviour, affects how subjects learn from positive and negative feedback. The REMS procedure was implemented by separating male and female rats from their dam for 6 h each day from post-natal day 5-19. Control rat offspring were left undisturbed during this period. Rats were tested as adults for behavioral flexibility and feedback sensitivity on a probabilistic reversal learning task. A computational approach based on RL theory was used to derive latent behavioral variables related to reward learning and flexibility. To assess underlying brain substrates, a seed-based functional MRI connectivity analysis was applied both before and after an additional adulthood stressor in control and REMS rats. Female but not male rats exposed to REMS stress showed increased response 'stickiness' (repeated responses regardless of reward outcome). Following repeated adulthood stress, reduced functional connectivity from the basolateral amygdala (BLA) to the dorsolateral striatum (DLS), cingulate cortex (Cg), and anterior insula (AI) cortex was observed in females. By contrast, control male rats exposed to the second stressor showed impaired learning from negative feedback (i.e., non-reward) and reduced functional connectivity from the BLA to the DLS and AI compared to maternally separated males. RL in male rats exposed to REMS was unaffected. The fMRI data further revealed that connectivity between the mOFC and other prefrontal cortical and subcortical structures was positively correlated with response 'stickiness'. These findings reveal differences in how females and males respond to early life adversity and subsequent stress. These effects may be mediated by functional divergence in resting-state connectivity between the basolateral amygdala and fronto-striatal brain regions.
Collapse
Affiliation(s)
- Katharina Zühlsdorff
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, CB2 3EB, UK
| | - Laura López-Cruz
- Faculty of Science, Technology, Engineering & Mathematics, The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK
| | - Ethan G. Dutcher
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, CB2 3EB, UK
| | - Jolyon A. Jones
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, CB2 3EB, UK
| | - Claudia Pama
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, CB2 3EB, UK
| | - Stephen Sawiak
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, CB2 3EB, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Box 65, Cambridge, CB2 0QQ, UK
| | - Shahid Khan
- GlaxoSmithKline Research & Development, Stevenage, UK
| | - Amy L. Milton
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, CB2 3EB, UK
| | - Trevor W. Robbins
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, CB2 3EB, UK
| | - Edward T. Bullmore
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, CB2 3EB, UK
- Department of Psychiatry, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Cambridge, CB2 0SZ, UK
| | - Jeffrey W. Dalley
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, CB2 3EB, UK
- Department of Psychiatry, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Cambridge, CB2 0SZ, UK
| |
Collapse
|
16
|
Płaza O, Gałecki P, Orzechowska A, Gałecka M, Sobolewska-Nowak J, Szulc A. Pharmacogenetics and Schizophrenia-Can Genomics Improve the Treatment with Second-Generation Antipsychotics? Biomedicines 2022; 10:biomedicines10123165. [PMID: 36551925 PMCID: PMC9775397 DOI: 10.3390/biomedicines10123165] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/19/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia (SCZ) is a complex psychiatric disorder of multifactorial origin, in which both genetic and environmental factors have an impact on its onset, course, and outcome. Large variability in response and tolerability of medication among individuals makes it difficult to predict the efficacy of a chosen therapeutic method and create universal and precise guidelines for treatment. Pharmacogenetic research allows for the identification of genetic polymorphisms associated with response to a chosen antipsychotic, thus allowing for a more effective and personal approach to treatment. This review focuses on three frequently prescribed second-generation antipsychotics (SGAs), risperidone, olanzapine, and aripiprazole, and aims to analyze the current state and future perspectives in research dedicated to identifying genetic factors associated with antipsychotic response. Multiple alleles of genes involved in pharmacokinetics (particularly isoenzymes of cytochrome P450), as well as variants of genes involved in dopamine, serotonin, and glutamate neurotransmission, have already been identified as ones of significant impact on antipsychotic response. It must, however, be noted that although currently obtained results are promising, trials with bigger study groups and unified protocols are crucial for standardizing methods and determining objective antipsychotic response status.
Collapse
Affiliation(s)
- Olga Płaza
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Partyzantów 2/4, 05-800 Pruszków, Poland
- Correspondence:
| | - Piotr Gałecki
- Department of Adult Psychiatry, Medical University of Łódź, Aleksandrowska 159, 91-229 Łódź, Poland
| | - Agata Orzechowska
- Department of Adult Psychiatry, Medical University of Łódź, Aleksandrowska 159, 91-229 Łódź, Poland
| | - Małgorzata Gałecka
- Department of Psychotherapy, Medical University of Łódź, Aleksandrowska 159, 91-229 Łódź, Poland
| | - Justyna Sobolewska-Nowak
- Department of Adult Psychiatry, Medical University of Łódź, Aleksandrowska 159, 91-229 Łódź, Poland
| | - Agata Szulc
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Partyzantów 2/4, 05-800 Pruszków, Poland
| |
Collapse
|
17
|
Petković A, Chaudhury D. Encore: Behavioural animal models of stress, depression and mood disorders. Front Behav Neurosci 2022; 16:931964. [PMID: 36004305 PMCID: PMC9395206 DOI: 10.3389/fnbeh.2022.931964] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Animal studies over the past two decades have led to extensive advances in our understanding of pathogenesis of depressive and mood disorders. Among these, rodent behavioural models proved to be of highest informative value. Here, we present a comprehensive overview of the most popular behavioural models with respect to physiological, circuit, and molecular biological correlates. Behavioural stress paradigms and behavioural tests are assessed in terms of outcomes, strengths, weaknesses, and translational value, especially in the domain of pharmacological studies.
Collapse
Affiliation(s)
| | - Dipesh Chaudhury
- Laboratory of Neural Systems and Behaviour, Department of Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
18
|
Takahashi A. The role of social isolation stress in escalated aggression in rodent models. Neurosci Res 2022:S0168-0102(22)00212-7. [PMID: 35917930 DOI: 10.1016/j.neures.2022.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/26/2022]
Abstract
Anti-social behavior and violence are major public health concerns. Globally, violence contributes to more than 1.6 million deaths each year. Previous studies have reported that social rejection or neglect exacerbates aggression. In rodent models, social isolation stress is used to demonstrate the adverse effects of social deprivation on physiological, endocrinological, immunological, and behavioral parameters, including aggressive behavior. This review summarizes recent rodent studies on the effect of social isolation stress during different developmental periods on aggressive behavior and the underlying neural mechanisms. Social isolation during adulthood affects the levels of neurosteroids and neuropeptides and increases aggressive behavior. These changes are ethologically relevant for the adaptation to changes in local environmental conditions in the natural habitats. Chronic deprivation of social interaction after weaning, especially during the juvenile to adolescent periods, leads to the disruption of the development of appropriate social behavior and the maladaptive escalation of aggressive behavior. The understanding of neurobiological mechanisms underlying social isolation-induced escalated aggression will aid in the development of therapeutic interventions for escalated aggression.
Collapse
Affiliation(s)
- Aki Takahashi
- Laboratory of Behavioral Neurobiology, Faculty of Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
19
|
Reemst K, Ruigrok SR, Bleker L, Naninck EFG, Ernst T, Kotah JM, Lucassen PJ, Roseboom TJ, Pollux BJA, de Rooij SR, Korosi A. Sex-dependence and comorbidities of the early-life adversity induced mental and metabolic disease risks: Where are we at? Neurosci Biobehav Rev 2022; 138:104627. [PMID: 35339483 DOI: 10.1016/j.neubiorev.2022.104627] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/15/2022] [Accepted: 03/13/2022] [Indexed: 01/02/2023]
Abstract
Early-life adversity (ELA) is a major risk factor for developing later-life mental and metabolic disorders. However, if and to what extent ELA contributes to the comorbidity and sex-dependent prevalence/presentation of these disorders remains unclear. We here comprehensively review and integrate human and rodent ELA (pre- and postnatal) studies examining mental or metabolic health in both sexes and discuss the role of the placenta and maternal milk, key in transferring maternal effects to the offspring. We conclude that ELA impacts mental and metabolic health with sex-specific presentations that depend on timing of exposure, and that human and rodent studies largely converge in their findings. ELA is more often reported to impact cognitive and externalizing domains in males, internalizing behaviors in both sexes and concerning the metabolic dimension, adiposity in females and insulin sensitivity in males. Thus, ELA seems to be involved in the origin of the comorbidity and sex-specific prevalence/presentation of some of the most common disorders in our society. Therefore, ELA-induced disease states deserve specific preventive and intervention strategies.
Collapse
Affiliation(s)
- Kitty Reemst
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands
| | - Silvie R Ruigrok
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands
| | - Laura Bleker
- Amsterdam University Medical Center, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam, The Netherlands
| | - Eva F G Naninck
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands
| | - Tiffany Ernst
- Wageningen University, Department of Animal Sciences, Experimental Zoology &Evolutionary Biology Group, Wageningen, The Netherlands
| | - Janssen M Kotah
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands
| | - Paul J Lucassen
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands; Centre for Urban Mental Health, University of Amsterdam, The Netherlands
| | - Tessa J Roseboom
- Amsterdam University Medical Center, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam, The Netherlands
| | - Bart J A Pollux
- Wageningen University, Department of Animal Sciences, Experimental Zoology &Evolutionary Biology Group, Wageningen, The Netherlands
| | - Susanne R de Rooij
- Amsterdam University Medical Center, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam, The Netherlands
| | - Aniko Korosi
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Levis SC, Baram TZ, Mahler SV. Neurodevelopmental origins of substance use disorders: Evidence from animal models of early-life adversity and addiction. Eur J Neurosci 2022; 55:2170-2195. [PMID: 33825217 PMCID: PMC8494863 DOI: 10.1111/ejn.15223] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 01/06/2023]
Abstract
Addiction is a chronic relapsing disorder with devastating personal, societal, and economic consequences. In humans, early-life adversity (ELA) such as trauma, neglect, and resource scarcity are linked with increased risk of later-life addiction, but the brain mechanisms underlying this link are still poorly understood. Here, we focus on data from rodent models of ELA and addiction, in which causal effects of ELA on later-life responses to drugs and the neurodevelopmental mechanisms by which ELA increases vulnerability to addiction can be determined. We first summarize evidence for a link between ELA and addiction in humans, then describe how ELA is commonly modeled in rodents. Since addiction is a heterogeneous disease with many individually varying behavioral aspects that may be impacted by ELA, we next discuss common rodent assays of addiction-like behaviors. We then summarize the specific addiction-relevant behavioral phenotypes caused by ELA in male and female rodents and discuss some of the underlying changes in brain reward and stress circuits that are likely responsible. By better understanding the behavioral and neural mechanisms by which ELA promotes addiction vulnerability, we hope to facilitate development of new approaches for preventing or treating addiction in those with a history of ELA.
Collapse
Affiliation(s)
- Sophia C. Levis
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA
| | - Tallie Z. Baram
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA
- Department of Pediatrics, University of California Irvine, Irvine, CA
| | - Stephen V. Mahler
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA
| |
Collapse
|
21
|
SWI/SNF chromatin remodeler complex within the reward pathway is required for behavioral adaptations to stress. Nat Commun 2022; 13:1807. [PMID: 35379786 PMCID: PMC8980038 DOI: 10.1038/s41467-022-29380-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 02/22/2022] [Indexed: 01/01/2023] Open
Abstract
Enduring behavioral changes upon stress exposure involve changes in gene expression sustained by epigenetic modifications in brain circuits, including the mesocorticolimbic pathway. Brahma (BRM) and Brahma Related Gene 1 (BRG1) are ATPase subunits of the SWI/SNF complexes involved in chromatin remodeling, a process essential to enduring plastic changes in gene expression. Here, we show that in mice, social defeat induces changes in BRG1 nuclear distribution. The inactivation of the Brg1/Smarca4 gene within dopamine-innervated regions or the constitutive inactivation of the Brm/Smarca2 gene leads to resilience to repeated social defeat and decreases the behavioral responses to cocaine without impacting midbrain dopamine neurons activity. Within striatal medium spiny neurons, Brg1 gene inactivation reduces the expression of stress- and cocaine-induced immediate early genes, increases levels of heterochromatin and at a global scale decreases chromatin accessibility. Altogether these data demonstrate the pivotal function of SWI/SNF complexes in behavioral and transcriptional adaptations to salient environmental challenges. Repeated exposure to social stressors in rodents results in behavioural changes. Here the authors show that behavioural adaptations to stress are associated with nuclear organization changes through SWI/SNF chromatin remodeler in specific neuronal populations of the mesolimbic system.
Collapse
|
22
|
Corticosterone induces discrete epigenetic signatures in the dorsal and ventral hippocampus that depend upon sex and genotype: focus on methylated Nr3c1 gene. Transl Psychiatry 2022; 12:109. [PMID: 35296634 PMCID: PMC8927334 DOI: 10.1038/s41398-022-01864-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023] Open
Abstract
The genomic effects of circulating glucocorticoids are particularly relevant in cortico-limbic structures, which express a high concentration of steroid hormone receptors. To date, no studies have investigated genomic differences in hippocampal subregions, namely the dorsal (dHPC) and ventral (vHPC) hippocampus, in preclinical models treated with exogenous glucocorticoids. Chronic oral corticosterone (CORT) in mouse is a pharmacological approach that disrupts the activity of the hypothalamic-pituitary-adrenal axis, increases affective behavior, and induces genomic changes after stress in the HPC of wildtype (WT) mice and mice heterozygous for the gene coding for brain-derived neurotrophic factor Val66Met (hMet), a variant associated with genetic susceptibility to stress. Using RNA-sequencing, we investigated the genomic signatures of oral CORT in the dHPC and vHPC of WT and hMet male and female mice, and examined sex and genotype differences in response to oral CORT. Males under CORT showed lower glycemia and increased anxiety- and depression-like behavior compared to females that showed instead opposite affective behavior in response to CORT. Rank-rank-hypergeometric overlap (RRHO) was used to identify genes from a continuous gradient of significancy that were concordant across groups. RRHO showed that CORT-induced differentially expressed genes (DEGs) in WT mice and hMet mice converged in the dHPC of males and females, while in the vHPC, DEGs converged in males and diverged in females. The vHPC showed a higher number of DEGs compared to the dHPC and exhibited sex differences related to glucocorticoid receptor (GR)-binding genes and epigenetic modifiers. Methyl-DNA-immunoprecipitation in the vHPC revealed differential methylation of the exons 1C and 1F of the GR gene (Nr3c1) in hMet females. Together, we report behavioral and endocrinological sex differences in response to CORT, as well as epigenetic signatures that i) differ in the dHPC and vHPC,ii) are distinct in males and females, and iii) implicate differential methylation of Nr3c1 selectively in hMet females.
Collapse
|
23
|
Waters RC, Worth HM, Vasquez B, Gould E. Inhibition of adult neurogenesis reduces avoidance behavior in male, but not female, mice subjected to early life adversity. Neurobiol Stress 2022; 17:100436. [PMID: 35146080 PMCID: PMC8819473 DOI: 10.1016/j.ynstr.2022.100436] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/08/2022] [Accepted: 01/24/2022] [Indexed: 12/20/2022] Open
Abstract
Early life adversity (ELA) increases the risk of developing neuropsychiatric illnesses such as anxiety disorders. However, the mechanisms connecting these negative early life experiences to illness later in life remain unclear. In rodents, plasticity mechanisms, specifically adult neurogenesis in the ventral hippocampus, have been shown to be altered by ELA and important for buffering against detrimental stress-induced outcomes. The current study sought to explore whether adult neurogenesis contributes to ELA-induced changes in avoidance behavior. Using the GFAP-TK transgenic model, which allows for the inhibition of adult neurogenesis, and CD1 littermate controls, we subjected mice to an ELA paradigm of maternal separation and early weaning (MSEW) or control rearing. We found that mice with intact adult neurogenesis showed no behavioral changes in response to MSEW. After reducing adult neurogenesis, however, male mice previously subjected to MSEW had an unexpected decrease in avoidance behavior. This finding was not observed in female mice, suggesting that a sex difference exists in the role of adult-born neurons in buffering against ELA-induced changes in behavior. Taken together with the existing literature on ELA and avoidance behavior, this work suggests that strain differences exist in susceptibility to ELA and that adult-born neurons may play a role in regulating adaptive behavior.
Collapse
|
24
|
Sex-specific behavioral and structural alterations caused by early-life stress in C57BL/6 and BTBR mice. Behav Brain Res 2021; 414:113489. [PMID: 34303728 DOI: 10.1016/j.bbr.2021.113489] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/06/2021] [Accepted: 07/21/2021] [Indexed: 11/21/2022]
Abstract
Lately, the development of various mental illnesses, such as depression, personality disorders, and autism spectrum disorders, is often associated with traumatic events in childhood. Nonetheless, the mechanism giving rise to this predisposition is still unknown. Because the development of a disease often depends on a combination of a genetic background and environment, we decided to evaluate the effect of early-life stress on BTBR mice, which have behavioral, neuroanatomical, and physiological features of autism spectrum disorders. As early-life stress, we used prolonged separation of pups from their mothers in the first 2 weeks of life (3 h once a day). We assessed effects of the early-life stress on juvenile (postnatal day 23) and adolescent (postnatal days 37-38) male and female mice of strains C57BL/6 (B6) and BTBR. We found that in both strains, the early-life stress did not lead to changes in the level of social behavior, which is an important characteristic of autism-related behavior. Nonetheless, the early-life stress resulted in increased locomotor activity in juvenile BTBR mice. In adolescent mice, the stress early in life caused a low level of anxiety in B6 males and BTBR females and increased exploratory activity in adolescent BTBR males and females. In addition, adolescent B6 male and female mice with a history of the early-life stress tended to have a thinner motor cortex as assessed by magnetic resonance imaging. As compared to B6 mice, BTBR mice showed reduced levels of social behavior and exploratory activity but their level of locomotor activity was higher. BTBR mice had smaller whole-brain, cortical, and dorsal hippocampal volumes; decreased motor cortex thickness; and increased ventral-hippocampus volume as compared to B6 mice, and these parameters correlated with the level of exploratory behavior of BTBR mice. Overall, the effects of early postnatal stress are sex- and strain-dependent.
Collapse
|
25
|
Tsotsokou G, Nikolakopoulou M, Kouvelas ED, Mitsacos A. Neonatal maternal separation affects metabotropic glutamate receptor 5 expression and anxiety-related behavior of adult rats. Eur J Neurosci 2021; 54:4550-4564. [PMID: 34137089 DOI: 10.1111/ejn.15358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 01/15/2023]
Abstract
Exposure to early life stress leads to long-term neurochemical and behavioral alterations. Stress-induced psychiatric disorders, such as depression, have recently been linked to dysregulation of glutamate signaling, mainly via its postsynaptic receptors. The role of metabotropic glutamate receptor 5 (mGluR5) in stress-induced psychopathology has been the target of several studies in humans. In rodents, blockade of mGluR5 produces antidepressant-like actions, whereas mice lacking mGluR5 exhibit altered anxiety-like behaviors and learning. In this study, we used well-known rodent models of early life stress based on mother-infant separation during the first 3 weeks of life in order to examine the effects of neonatal maternal separation on mGluR5 expression and on anxiety-related behavior in adulthood. We observed that brief (15 min) neonatal maternal separation, but not prolonged (3 h), induced increases in mGluR5 mRNA and protein expression levels in medial prefrontal cortex and mGluR5 protein levels in dorsal, but not ventral, hippocampus of adult rat brain. Behavioral testing using the open-field and the elevated-plus maze tasks showed that brief maternal separations resulted in increased exploratory and decreased anxiety-related behavior, whereas prolonged maternal separations resulted in increased anxiety-related behavior in adulthood. The data indicate that the long-lasting effects of neonatal mother-offspring separation on anxiety-like behavior and mGluR5 expression depend on the duration of maternal separation and suggest that the increased mGluR5 receptors in medial prefrontal cortex and hippocampus of adult rats exposed to brief neonatal maternal separations may underlie their heightened ability to cope with stress.
Collapse
Affiliation(s)
- Giota Tsotsokou
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| | - Maria Nikolakopoulou
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| | - Elias D Kouvelas
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| | - Ada Mitsacos
- School of Health Sciences, Department of Medicine, Laboratory of Physiology, University Campus, University of Patras, Patras, Greece
| |
Collapse
|
26
|
Kooiker CL, Birnie MT, Baram TZ. The Paraventricular Thalamus: A Potential Sensor and Integrator of Emotionally Salient Early-Life Experiences. Front Behav Neurosci 2021; 15:673162. [PMID: 34079442 PMCID: PMC8166219 DOI: 10.3389/fnbeh.2021.673162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/20/2021] [Indexed: 02/03/2023] Open
Abstract
Early-life experiences influence a broad spectrum of behaviors throughout the lifespan that contribute to resilience or vulnerability to mental health disorders. Yet, how emotionally salient experiences early in life are encoded, stored, and processed and the mechanisms by which they influence future behaviors remain poorly understood. The paraventricular nucleus of the thalamus (PVT) is a key structure in modulating positive and negative experiences and behaviors in adults. However, little is known of the PVT's role in encoding and integrating emotionally salient experiences that occur during neonatal, infancy, and childhood periods. In this review, we (1) describe the functions and connections of the PVT and its regulation of behavior, (2) introduce novel technical approaches to elucidating the role of the PVT in mediating enduring changes in adult behaviors resulting from early-life experiences, and (3) conclude that PVT neurons of neonatal rodents are engaged by both positive and negative emotionally salient experiences, and their activation may enduringly govern future behavior-modulating PVT activity during emotionally salient contexts.
Collapse
Affiliation(s)
- Cassandra L. Kooiker
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA, United States
| | - Matthew T. Birnie
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| | - Tallie Z. Baram
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
27
|
Henderson HJM, Etem G, Bjorni M, Belnap MA, Rosellini B, Halladay LR. Sex-dependent and ontogenetic effects of low dose ethanol on social behavioral deficits induced by mouse maternal separation. Behav Brain Res 2021; 406:113241. [PMID: 33727047 DOI: 10.1016/j.bbr.2021.113241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/06/2021] [Accepted: 03/09/2021] [Indexed: 10/21/2022]
Abstract
Early life stress can induce lifelong emotional and social behavioral deficits that may in some cases be alleviated by drugs or alcohol. A model for early life stress, rodent maternal separation, recapitulates these behavioral sequelae, which are not limited to potentiated anxiety-like behavior, attenuated social motivation, and altered reward-seeking. Here we employed mouse maternal separation with early weaning (MSEW), consisting of pup-dam separation lasting 4-8 hours on postnatal days (PD) 2-16, with early weaning on PD 17. Prior MSEW studies have limited subjects by age or sex, so we more comprehensively investigated MSEW effects in both sexes, during adolescence and adulthood. We found universal effects of MSEW to include lifelong enhancement of anxiety-like and despair behavior, as well as deficits in social motivation. We also observed some sex-dependent effects of MSEW, namely that female MSEW mice exhibited social habituation to a greater degree than their male counterparts. Low dose ethanol administration had no major effects on the social behavior of non-stressed mice. But interestingly, MSEW-induced social habituation was counteracted by low dose ethanol in adolescent female mice, and potentiated in adolescent male mice. These effects were absent in adult animals, suggesting that ethanol may exert differential effects on the developing brain in such a manner to produce age-, sex-, and stress-dependent effects upon social behavior. Together, results indicate that MSEW reliably produces long-lasting impairments in emotional and social behaviors in both sexes and across the lifespan, but may exert more salient social behavioral effects on female animals.
Collapse
Affiliation(s)
- Hannah J M Henderson
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Gabrielle Etem
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Max Bjorni
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Malia A Belnap
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Bryce Rosellini
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Lindsay R Halladay
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA.
| |
Collapse
|
28
|
Levis SC, Mahler SV, Baram TZ. The Developmental Origins of Opioid Use Disorder and Its Comorbidities. Front Hum Neurosci 2021; 15:601905. [PMID: 33643011 PMCID: PMC7904686 DOI: 10.3389/fnhum.2021.601905] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/20/2021] [Indexed: 12/12/2022] Open
Abstract
Opioid use disorder (OUD) rarely presents as a unitary psychiatric condition, and the comorbid symptoms likely depend upon the diverse risk factors and mechanisms by which OUD can arise. These factors are heterogeneous and include genetic predisposition, exposure to prescription opioids, and environmental risks. Crucially, one key environmental risk factor for OUD is early life adversity (ELA). OUD and other substance use disorders are widely considered to derive in part from abnormal reward circuit function, which is likely also implicated in comorbid mental illnesses such as depression, bipolar disorder, and schizophrenia. ELA may disrupt reward circuit development and function in a manner predisposing to these disorders. Here, we describe new findings addressing the effects of ELA on reward circuitry that lead to OUD and comorbid disorders, potentially via shared neural mechanisms. We discuss some of these OUD-related problems in both humans and animals. We also highlight the increasingly apparent, crucial contribution of biological sex in mediating the range of ELA-induced disruptions of reward circuitry which may confer risk for the development of OUD and comorbid neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sophia C. Levis
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - Stephen V. Mahler
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - Tallie Z. Baram
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
29
|
Alteba S, Portugalov A, Hillard CJ, Akirav I. Inhibition of Fatty Acid Amide Hydrolase (FAAH) During Adolescence and Exposure to Early Life Stress may Exacerbate Depression-like Behaviors in Male and Female Rats. Neuroscience 2021; 455:89-106. [PMID: 33359656 DOI: 10.1016/j.neuroscience.2020.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 01/06/2023]
Abstract
Early-life stress (ELS) is associated with later onset of depression. Early cannabis use may be a risk factor that interacts with environmental factors to increase the risk of psychopathologies. We aimed to examine the long-term effects of ELS on depression- and anxiety-like behavior, and examine whether chronic fatty acid amide hydrolase (FAAH) inhibition during mid-adolescence could ameliorate or exacerbate ELS effects on behavior. Male and female rats were exposed to ELS during post-natal days (P) 7-14, injected with the FAAH inhibitor URB597 (0.4 mg/kg, i.p.) or vehicle for 2 weeks during mid-adolescence (P30-45) or late-adolescence (P45-60). Rats were tested in adulthood for behavior and alterations in CB1 receptors (CB1r) and glucocorticoid receptors (GRs) in the brains' stress circuit. ELS produced decreased social preference, impaired social recognition, increased learned helplessness and anxiety-like behavior. Administering URB597 during mid-adolescence did not prevent the deleterious long-term effects of ELS on behavior in males and females. When URB597 was administered during late-adolescence, it ameliorated ELS-induced depression- and anxiety-like behavior. Moreover, in males, ELS and URB597 decreased CB1r levels in the prefrontal cortex (PFC) and CA1 and GRs in the PFC and basolateral amygdala (BLA). In females, ELS and URB decreased CB1r in the BLA and GRs in the CA1 and BLA. The findings suggest that mid-adolescence, as opposed to late-adolescence, may not be a potential developmental period for chronic treatment with FAAH inhibitors and that sex-dependent alterations in CB1r and GRs expression in the BLA-PFC-CA1 circuit may contribute to the depressive behavioral phenotype.
Collapse
Affiliation(s)
- Shirley Alteba
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa 3498838, Israel
| | - Anna Portugalov
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa 3498838, Israel
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee 53226, USA
| | - Irit Akirav
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
30
|
Sprowles JLN, Vorhees CV, Williams MT. Impact of preweaning stress on long-term neurobehavioral outcomes in Sprague-Dawley rats: Differential effects of barren cage rearing, pup isolation, and the combination. Neurotoxicol Teratol 2021; 84:106956. [PMID: 33524508 DOI: 10.1016/j.ntt.2021.106956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/06/2021] [Accepted: 01/17/2021] [Indexed: 12/23/2022]
Abstract
Two developmental stressors were compared in preweaning rats exposed to either one stressor or both. Stressors were barren cage rearing or maternal separation (pup isolation). 40 gravid Sprague-Dawley CD/IGS rats were randomly assigned to two cage conditions: standard (Std) cage or barren cage (Bar), 20 litters/condition throughout gestation and lactation. After delivery, litters were randomly culled to 4 males and 4 females. The second stressor was maternal separation: Two male/female pairs per litter were isolated from their dam 4 h/day (Iso) and two pairs were not (Norm). Hence, there were 4 conditions: Std-Norm, Std-Iso, Bar-Norm, and Bar-Iso. One pair/litter/stress condition received the following: elevated zero-maze (EZM), open-field, swim channel, Cincinnati water maze, conditioned fear, and open-field with methamphetamine challenge. The second pair/litter/condition received the light-dark test, swim channel, Morris water maze, forced swim, and EZM with diazepam challenge. Barren rearing reduced EZM time-in-open, whereas isolation rearing reduced open-field activity in males and increased it in females. Effects on straight channel swimming were minor. In the Cincinnati water maze test of egocentric learning, isolation rearing increased errors whereas barren cage housing reduced errors in combination with normal rearing. Barren cage with maternal separation (pup isolation) increased Cincinnati water maze escape latency but not errors. Barren cage housing reduced hyperactivity in response to methamphetamine. Isolation rearing increased time in open in the EZM after diazepam challenge. Trends were seen in the Morris water maze. These suggested that barren cage and isolation rearing in combination reduced latency on acquisition on days 1 and 2 in males, whereas females had increased latency on days 2 and 3. Combined exposure to two developmental stressors did not induce additive or synergistic effects, however the data show that these stressors had long-term effects with some evidence that the combination of both caused effects when either stressor alone did not, but synergism was not observed.
Collapse
Affiliation(s)
- Jenna L N Sprowles
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.
| |
Collapse
|
31
|
Li M, Fu X, Xie W, Guo W, Li B, Cui R, Yang W. Effect of Early Life Stress on the Epigenetic Profiles in Depression. Front Cell Dev Biol 2020; 8:867. [PMID: 33117794 PMCID: PMC7575685 DOI: 10.3389/fcell.2020.00867] [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: 05/14/2020] [Accepted: 08/11/2020] [Indexed: 12/23/2022] Open
Abstract
Depression is one of the most common mental disorders and has caused an overwhelming burden on world health. Abundant studies have suggested that early life stress may grant depressive-like phenotypes in adults. Childhood adversities that occurred in the developmental period amplified stress events in adulthood. Epigenetic-environment interaction helps to explain the role of early life stress on adulthood depression. Early life stress shaped the epigenetic profiles of the HPA axis, monoamine, and neuropeptides. In the context of early adversities increasing the risk of depression, early life stress decreased the activity of the glucocorticoid receptors, halted the circulation and production of serotonin, and reduced the molecules involved in modulating the neurogenesis and neuroplasticity. Generally, DNA methylation, histone modifications, and the regulation of non-coding RNAs programmed the epigenetic profiles to react to early life stress. However, genetic precondition, subtypes of early life stress, the timing of epigenetic status evaluated, demographic characteristics in humans, and strain traits in animals favored epigenetic outcomes. More research is needed to investigate the direct evidence for how early life stress-induced epigenetic changes contribute to the vulnerability of depression.
Collapse
Affiliation(s)
- Ming Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xiying Fu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wei Xie
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wanxu Guo
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
32
|
Alteba S, Mizrachi Zer-Aviv T, Tenenhaus A, Ben David G, Adelman J, Hillard CJ, Doron R, Akirav I. Antidepressant-like effects of URB597 and JZL184 in male and female rats exposed to early life stress. Eur Neuropsychopharmacol 2020; 39:70-86. [PMID: 32891517 DOI: 10.1016/j.euroneuro.2020.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/21/2020] [Accepted: 08/17/2020] [Indexed: 12/27/2022]
Abstract
Early life stress (ELS) may increase predisposition to depression. Despite extensive research, there is still a lack of knowledge of how to optimally treat depression. We aimed to establish a role for the endocannabinoid (ECB) system within the hippocampal-nucleus accumbens (NAc) network as a possible effective target in combating the pathophysiological development of depression-like behavior and neuronal alterations that are precipitated by ELS. Male and female rats were exposed to ELS during post-natal days (P) 7-14, injected with the fatty acid amide hydrolase (FAAH) inhibitor URB597 or the monoacylglycerol lipase (MAGL) inhibitor JZL184 for 2 weeks during late-adolescence (P45-60). Rats were tested starting at P90 for depressive- and anxiety-like behaviors as well as social preference and recognition; alterations in FAAH and MAGL activity; the expression of brain-derived neurotrophic factor (BDNF); and plasticity in the hippocampal-NAc pathway. FAAH and MAGL inhibitors during late-adolescence prevented: (i) the long-term effects of ELS on depression- and anxiety-like behavior and the impairment in social behavior and neuronal plasticity in males and females; (ii) ELS-induced alterations in MAGL activity in males' hippocampus and females' hippocampus and NAc; and (iii) ELS-induced alterations in BDNF in males' hippocampus and NAc and females' hippocampus. Significant correlations were observed between alterations in MAGL and BDNF levels and the behavioral phenotype. The findings suggest that alterations in MAGL activity and BDNF expression in the hippocampal-NAc network contribute to the depressive-like behavioral phenotype in ELS males and females. Moreover, the study suggests FAAH and MAGL inhibitors as potential intervention drugs for depression.
Collapse
Affiliation(s)
- Shirley Alteba
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa, 3498838, Israel
| | - Tomer Mizrachi Zer-Aviv
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa, 3498838, Israel
| | - Adi Tenenhaus
- Department of Education and Psychology, The Open University of Israel, Israel
| | - Gilad Ben David
- Department of Education and Psychology, The Open University of Israel, Israel
| | - Jacob Adelman
- Department of Pharmacology and Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee 53226, USA
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee 53226, USA
| | - Ravid Doron
- Department of Education and Psychology, The Open University of Israel, Israel
| | - Irit Akirav
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel; The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
33
|
Braun K, Bock J, Wainstock T, Matas E, Gaisler-Salomon I, Fegert J, Ziegenhain U, Segal M. Experience-induced transgenerational (re-)programming of neuronal structure and functions: Impact of stress prior and during pregnancy. Neurosci Biobehav Rev 2020; 117:281-296. [DOI: 10.1016/j.neubiorev.2017.05.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022]
|
34
|
Xia Z, Qi W, Xiaofeng G, Jiguang K, Hongfei H, Yuchen Z, Yihan Z, Yan W, Nannan L, Yiwei L, Hongsheng B, Xiaobai L. AMBMP activates WNT pathway and alleviates stress-induced behaviors in maternal separation and chronic stress models. Eur J Pharmacol 2020; 881:173192. [DOI: 10.1016/j.ejphar.2020.173192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
|
35
|
Wang D, Levine JLS, Avila-Quintero V, Bloch M, Kaffman A. Systematic review and meta-analysis: effects of maternal separation on anxiety-like behavior in rodents. Transl Psychiatry 2020; 10:174. [PMID: 32483128 PMCID: PMC7264128 DOI: 10.1038/s41398-020-0856-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 05/06/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
The mechanisms by which childhood maltreatment increases anxiety is unclear, but a propensity for increased defensive behavior in rodent models of early life stress (ELS) suggests that work in rodents may clarify important mechanistic details about this association. A key challenge in studying the effects of ELS on defensive behavior in rodents is the plethora of inconsistent results. This is particularly prominent with the maternal separation (MS) literature, one of the most commonly used ELS models in rodents. To address this issue we conducted a systematic review and meta-analysis, examining the effects of MS on exploratory-defensive behavior in mice and rats using the open field test (OFT) and the elevated plus maze (EPM). This search yielded a total of 49 studies, 24 assessing the effect of MS on behavior in the EPM, 11 tested behavior in the OFT, and 14 studies provided data on both tasks. MS was associated with increased defensive behavior in rats (EPM: Hedge's g = -0.48, p = 0.02; OFT: Hedge's g = -0.33, p = 0.05), effect sizes that are consistent with the anxiogenic effect of early adversity reported in humans. In contrast, MS did not alter exploratory behavior in mice (EPM: Hedge's g = -0.04, p = 0.75; OFT: Hedge's g = -0.03, p = 0.8). There was a considerable amount of heterogeneity between studies likely related to the lack of standardization of the MS protocol. Together, these findings suggest important differences in the ability of MS to alter circuits that regulate defensive behaviors in mice and rats.
Collapse
Affiliation(s)
- Daniel Wang
- grid.47100.320000000419368710Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
| | - Jessica L. S. Levine
- grid.47100.320000000419368710Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, CT 06519 USA
| | - Victor Avila-Quintero
- grid.47100.320000000419368710Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, CT 06519 USA
| | - Michael Bloch
- grid.47100.320000000419368710Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA ,grid.47100.320000000419368710Child Study Center, Yale University School of Medicine, 230 South Frontage Road, New Haven, CT 06519 USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA.
| |
Collapse
|
36
|
Ponzoni L, Braida D, Carboni L, Moretti M, Viani P, Clementi F, Zoli M, Gotti C, Sala M. Persistent cognitive and affective alterations at late withdrawal stages after long-term intermittent exposure to tobacco smoke or electronic cigarette vapour: Behavioural changes and their neurochemical correlates. Pharmacol Res 2020; 158:104941. [PMID: 32450347 DOI: 10.1016/j.phrs.2020.104941] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/19/2022]
Abstract
Smoking cessation induces a withdrawal syndrome associated with anxiety, depression, and impaired neurocognitive functions, but much less is known about the withdrawal of e-cigarettes (e-CIG). We investigated in Balb/c mice the behavioural and neurochemical effects of withdrawal for up to 90 days after seven weeks' intermittent exposure to e-CIG vapour or cigarette smoke (CIG). The withdrawal of e-CIG and CIG induced early behavioural alterations such as spatial memory deficits (spatial object recognition task), increased anxiety (elevated plus maze test) and compulsive-like behaviour (marble burying test) that persisted for 60-90 days. Notably, attention-related (virtual object recognition task) and depression-like behaviours (tail suspension and sucrose preference tests) appeared only 15-30 days after withdrawal and persisted for as long as up to 90 days. At hippocampal level, the withdrawal-induced changes in the levels of AMPA receptor GluA1 and GluA2/3 subunits, PSD 95 protein, corticotropin-releasing factor (Crf) and Crf receptor 1 (CrfR1) mRNA were biphasic: AMPA receptor subunit and PSD95 protein levels initially remained unchanged and decreased after 60-90 days, whereas Crf/CrfR1 mRNA levels initially increased and then markedly decreased after 60 days. These late reductions correlated with the behavioural impairments, particularly the appearance of depression-like behaviours. Our findings show that major behavioural and neurochemical alterations persist or even first appear late after the withdrawal of chronic CIG smoke or e-CIG vapour exposure, and underline importance of conducting similar studies of humans, including e-CIG vapers.
Collapse
Affiliation(s)
- Luisa Ponzoni
- CNR, Institute of Neuroscience, Milan, Italy; Fondazione Zardi-Gori, Milan, Italy; Dept. of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Daniela Braida
- Dept. of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Milena Moretti
- CNR, Institute of Neuroscience, Milan, Italy; Dept. of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Paola Viani
- Dept. of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Francesco Clementi
- CNR, Institute of Neuroscience, Milan, Italy; Dept. of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Michele Zoli
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology (CfNN), University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Cecilia Gotti
- CNR, Institute of Neuroscience, Milan, Italy; Dept. of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy.
| | - Mariaelvina Sala
- CNR, Institute of Neuroscience, Milan, Italy; Dept. of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
37
|
Maternal Separation Early in Life Alters the Expression of Genes Npas4 and Nr1d1 in Adult Female Mice: Correlation with Social Behavior. Behav Neurol 2020; 2020:7830469. [PMID: 32190129 PMCID: PMC7072106 DOI: 10.1155/2020/7830469] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/05/2020] [Accepted: 02/15/2020] [Indexed: 12/22/2022] Open
Abstract
Early-life stress affects neuronal plasticity of the brain regions participating in the implementation of social behavior. Our previous studies have shown that brief and prolonged separation of pups from their mothers leads to enhanced social behavior in adult female mice. The goal of the present study was to characterize the expression of genes (which are engaged in synaptic plasticity) Egr1, Npas4, Arc, and Homer1 in the prefrontal cortex and dorsal hippocampus of adult female mice with a history of early-life stress. In addition, we evaluated the expression of stress-related genes: glucocorticoid and mineralocorticoid receptors (Nr3c1 and Nr3c2) and Nr1d1, which encodes a transcription factor (also known as REVERBα) modulating sociability and anxiety-related behavior. C57Bl/6 mice were exposed to either maternal separation (MS, 3 h once a day) or handling (HD, 15 min once a day) on postnatal days 2 through 14. In adulthood, the behavior of female mice was analyzed by some behavioral tests, and on the day after the testing of social behavior, we measured the gene expression. We found increased Npas4 expression only in the prefrontal cortex and higher Nr1d1 expression in both the prefrontal cortex and dorsal hippocampus of adult female mice with a history of MS. The expression of the studied genes did not change in HD female mice. The expression of stress-related genes Nr3c1 and Nr3c2 was unaltered in both groups. We propose that the upregulation of Npas4 and Nr1d1 in females with a history of early-life stress and the corresponding enhancement of social behavior may be regarded as an adaptation mechanism reversing possible aberrations caused by early-life stress.
Collapse
|
38
|
Prenatal maternal stress is associated with increased sensitivity to neuropathic pain and sex-specific changes in supraspinal mRNA expression of epigenetic- and stress-related genes in adulthood. Behav Brain Res 2020; 380:112396. [DOI: 10.1016/j.bbr.2019.112396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
|
39
|
Abstract
Depression is one of the most common psychiatric disorders affecting public health. Studies over the past years suggest that the methylations of some specific genes such as BDNF, SLC6A4, and NR3C1 play an important role in the development of depression. Recently, epigenetic evidences suggest that the expression levels of DNA methyltransferases differ in several brain areas including the prefrontal cortex, hippocampus, amygdala, and nucleus accumbens in depression patients and animal models, but the potential link between the expression levels of DNA methylatransferases and the methylations of specific genes needs further investigation to clarify the pathogenesis of depression.
Collapse
Affiliation(s)
- Zhenghao Duan
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jie Lu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
40
|
Agarwal P, Palin N, Walker SL, Glasper ER. Sex-dependent effects of paternal deprivation and chronic variable stress on novel object recognition in adult California mice (Peromyscus californicus). Horm Behav 2020; 117:104610. [PMID: 31669457 DOI: 10.1016/j.yhbeh.2019.104610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022]
Abstract
Early-life stress exposure can confer vulnerability for development of psychiatric illnesses and impaired cognition in adulthood. It is well-known that early-life stress can dysregulate the hypothalamic-pituitary-adrenal (HPA) axis in a sex-dependent manner. Specifically, uniparental rodent models of prolonged disrupted mother-offspring relationships (e.g., maternal separation) have demonstrated greater alterations in stress responsivity in adult males, compared to females. Also, chronic early-life stressors (e.g., limited bedding model) impair cognitive function in males more than females. However, the sex-dependent effects of early-life stress and later-life chronic HPA axis activation on cognition have not been well-characterized. Here, we utilized the biparental California mouse (Peromyscus californicus) to model the early-life adversity of paternal deprivation (PD). Fathers either remained in the nest (biparental care) or were permanently removed (PD) on postnatal day (PND) 1. Adult offspring were exposed to daily handling (control) or chronic variable stress (CVS; three stressors for seven days). Twenty-four hours after the final stressor, the novel object recognition (NOR) task commenced, followed by serum collection for corticosterone (CORT) analysis. Independent of sex or rearing, CVS increased CORT. Exploration during acquisition for the NOR task was increased as a result of CVS and PD. During NOR testing, non-stressed females exhibited greater difference scores (i.e., increased recognition memory), compared to non-stressed males. However, the addition of CVS diminished difference scores in females - an effect not observed in CVS-exposed males. Overall, these data suggest that neonatal paternal experience, sex, and chronic stress contribute to exploratory behavior, cognition, and stress hormone concentrations in a biparental species.
Collapse
Affiliation(s)
- P Agarwal
- Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | - N Palin
- Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | - S L Walker
- Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742, USA
| | - E R Glasper
- Department of Psychology, University of Maryland, College Park, MD 20742, USA; Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
41
|
Torres-Berrío A, Issler O, Parise EM, Nestler EJ. Unraveling the epigenetic landscape of depression: focus on early life stress
. DIALOGUES IN CLINICAL NEUROSCIENCE 2019; 21:341-357. [PMID: 31949402 PMCID: PMC6952747 DOI: 10.31887/dcns.2019.21.4/enestler] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Depression is a devastating psychiatric disorder caused by a combination of genetic predisposition and life events, mainly exposure to stress. Early life stress (ELS) in particular is known to "scar" the brain, leading to an increased susceptibility to developing depression later in life via epigenetic mechanisms. Epigenetic processes lead to changes in gene expression that are not due to changes in DNA sequence, but achieved via modulation of chromatin modifications, DNA methylation, and noncoding RNAs. Here we review common epigenetic mechanisms including the enzymes that take part in reading, writing, and erasing specific epigenetic marks. We then describe recent developments in understanding how ELS leads to changes in the epigenome that are manifested in increased susceptibility to depression-like abnormalities in animal models. We conclude with highlighting the need for future studies that will potentially enable the utilisation of the understanding of epigenetic changes linked to ELS for the development of much-needed novel therapeutic strategies and biomarker discovery.
.
Collapse
Affiliation(s)
- Angélica Torres-Berrío
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Orna Issler
- Author affiliations: Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Eric M Parise
- Author affiliations: Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Eric J Nestler
- Author affiliations: Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| |
Collapse
|
42
|
Glastad KM, Graham RJ, Ju L, Roessler J, Brady CM, Berger SL. Epigenetic Regulator CoREST Controls Social Behavior in Ants. Mol Cell 2019; 77:338-351.e6. [PMID: 31732456 DOI: 10.1016/j.molcel.2019.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/13/2019] [Accepted: 10/11/2019] [Indexed: 11/25/2022]
Abstract
Ants acquire distinct morphological and behavioral phenotypes arising from a common genome, underscoring the importance of epigenetic regulation. In Camponotus floridanus, "Major" workers defend the colony, but can be epigenetically reprogrammed to forage for food analogously to "Minor" workers. Here, we utilize reprogramming to investigate natural behavioral specification. Reprogramming of Majors upregulates Minor-biased genes and downregulates Major-biased genes, engaging molecular pathways fundamental to foraging behavior. We discover the neuronal corepressor for element-1-silencing transcription factor (CoREST) is upregulated upon reprogramming and required for the epigenetic switch to foraging. Genome-wide profiling during reprogramming reveals CoREST represses expression of enzymes that degrade juvenile hormone (JH), a hormone elevated upon reprogramming. High CoREST, low JH-degrader expression, and high JH levels are mirrored in natural Minors, revealing parallel mechanisms of natural and reprogrammed foraging. These results unveil chromatin regulation via CoREST as central to programming of ant social behavior, with potential far-reaching implications for behavioral epigenetics.
Collapse
Affiliation(s)
- Karl M Glastad
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Riley J Graham
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Linyang Ju
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julian Roessler
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cristina M Brady
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shelley L Berger
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
43
|
Abstract
The developmental period constitutes a critical window of sensitivity to stress. Indeed, early-life adversity increases the risk to develop psychiatric diseases, but also gastrointestinal disorders such as the irritable bowel syndrome at adulthood. In the past decade, there has been huge interest in the gut-brain axis, especially as regards stress-related emotional behaviours. Animal models of early-life adversity, in particular, maternal separation (MS) in rodents, demonstrate lasting deleterious effects on both the gut and the brain. Here, we review the effects of MS on both systems with a focus on stress-related behaviours. In addition, we discuss more recent findings showing the impact of gut-directed interventions, including nutrition with pre- and probiotics, illustrating the role played by gut microbiota in mediating the long-term effects of MS. Overall, preclinical studies suggest that nutritional approaches with pro- and prebiotics may constitute safe and efficient strategies to attenuate the effects of early-life stress on the gut-brain axis. Further research is required to understand the complex mechanisms underlying gut-brain interaction dysfunctions after early-life stress as well as to determine the beneficial impact of gut-directed strategies in a context of early-life adversity in human subjects.
Collapse
|
44
|
Reshetnikov VV, Ryabushkina YA, Bondar NP. Impact of mothers’ experience and early‐life stress on aggression and cognition in adult male mice. Dev Psychobiol 2019; 62:36-49. [DOI: 10.1002/dev.21887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/26/2019] [Accepted: 05/22/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Vasiliy V. Reshetnikov
- Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences (SB RAS) Novosibirsk Russia
| | - Yulia A. Ryabushkina
- Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences (SB RAS) Novosibirsk Russia
- Novosibirsk State University Novosibirsk Russia
| | - Natalia P. Bondar
- Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences (SB RAS) Novosibirsk Russia
- Novosibirsk State University Novosibirsk Russia
| |
Collapse
|
45
|
Rodiño-Janeiro BK, Pardo-Camacho C, Santos J, Martínez C. Mucosal RNA and protein expression as the next frontier in IBS: abnormal function despite morphologically intact small intestinal mucosa. Am J Physiol Gastrointest Liver Physiol 2019; 316:G701-G719. [PMID: 30767681 DOI: 10.1152/ajpgi.00186.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Irritable bowel syndrome (IBS) is one of the commonest gastrointestinal disorders. Although long-time considered a pure functional disorder, intense research in past years has rendered a very complex and varied array of observations indicating the presence of structural and molecular abnormalities underlying characteristic motor and sensitive changes and clinical manifestations. Analysis of gene and protein expression in the intestinal mucosa has shed light on the molecular mechanisms implicated in IBS physiopathology. This analysis uncovers constitutive and inductive genetic and epigenetic marks in the small and large intestine that highlight the role of epithelial barrier, immune activation, and mucosal processing of foods and toxins and several new molecular pathways in the origin of IBS. The incorporation of innovative high-throughput techniques into IBS research is beginning to provide new insights into highly structured and interconnected molecular mechanisms modulating gene and protein expression at tissue level. Integration and correlation of these molecular mechanisms with clinical and environmental data applying systems biology/medicine and data mining tools emerge as crucial steps that will allow us to get meaningful and more definitive comprehension of IBS-detailed development and show the real mechanisms and causality of the disease and the way to identify more specific diagnostic biomarkers and effective treatments.
Collapse
Affiliation(s)
- Bruno Kotska Rodiño-Janeiro
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca , Barcelona , Spain.,Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina) , Barcelona , Spain
| | - Cristina Pardo-Camacho
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca , Barcelona , Spain.,Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina) , Barcelona , Spain
| | - Javier Santos
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca , Barcelona , Spain.,Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina) , Barcelona , Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas , Madrid , Spain
| | - Cristina Martínez
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca , Barcelona , Spain.,Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina) , Barcelona , Spain
| |
Collapse
|
46
|
Bludau A, Royer M, Meister G, Neumann ID, Menon R. Epigenetic Regulation of the Social Brain. Trends Neurosci 2019; 42:471-484. [PMID: 31103351 DOI: 10.1016/j.tins.2019.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022]
Abstract
Social behavior, a highly adaptive and crucial component of mammalian life, is regulated by particularly sensitive regulatory brain mechanisms. Substantial evidence implicates classical epigenetic mechanisms including histone modifications, DNA methylation, and nucleosome remodeling as well as nonclassical mechanisms mediated by noncoding RNA in the regulation of social behavior. These mechanisms collectively form the 'epigenetic network' that orchestrates genomic integration of salient and transient social experiences. Consequently, its dysregulation has been linked to behavioral deficits and psychopathologies. This review focuses on the role of the epigenetic network in regulating the enduring effects of social experiences during early-life, adolescence, and adulthood. We discuss research in animal models, primarily rodents, and associations between dysregulation of epigenetic mechanisms and human psychopathologies, specifically autism spectrum disorder (ASD) and schizophrenia.
Collapse
Affiliation(s)
- Anna Bludau
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Melanie Royer
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany; Biochemistry Center Regensburg (BZR), Laboratory of RNA Biology, University of Regensburg, Regensburg, Germany
| | - Gunter Meister
- Biochemistry Center Regensburg (BZR), Laboratory of RNA Biology, University of Regensburg, Regensburg, Germany
| | - Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Rohit Menon
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
47
|
Risbrough VB, Glynn LM, Davis EP, Sandman CA, Obenaus A, Stern HS, Keator DB, Yassa MA, Baram TZ, Baker DG. Does Anhedonia Presage Increased Risk of Posttraumatic Stress Disorder? : Adolescent Anhedonia and Posttraumatic Disorders. Curr Top Behav Neurosci 2019; 38:249-265. [PMID: 29796839 PMCID: PMC9167566 DOI: 10.1007/7854_2018_51] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Anhedonia, the reduced ability to experience pleasure, is a dimensional entity linked to multiple neuropsychiatric disorders, where it is associated with diminished treatment response, reduced global function, and increased suicidality. It has been suggested that anhedonia and the related disruption in reward processing may be critical precursors to development of psychiatric symptoms later in life. Here, we examine cross-species evidence supporting the hypothesis that early life experiences modulate development of reward processing, which if disrupted, result in anhedonia. Importantly, we find that anhedonia may confer risk for later neuropsychiatric disorders, especially posttraumatic stress disorder (PTSD). Whereas childhood trauma has long been associated with increased anhedonia and increased subsequent risk for trauma-related disorders in adulthood, here we focus on an additional novel, emerging direct contributor to anhedonia in rodents and humans: fragmented, chaotic environmental signals ("FRAG") during critical periods of development. In rodents, recent data suggest that adolescent anhedonia may derive from aberrant pleasure/reward circuit maturation. In humans, recent longitudinal studies support that FRAG is associated with increased anhedonia in adolescence. Both human and rodent FRAG exposure also leads to aberrant hippocampal function. Prospective studies are underway to examine if anhedonia is also a marker of PTSD risk. These preliminary cross-species studies provide a critical construct for future examination of the etiology of trauma-related symptoms in adults and for and development of prophylactic and therapeutic interventions. In addition, longitudinal studies of reward circuit development with and without FRAG will be critical to test the mechanistic hypothesis that early life FRAG modifies reward circuitry with subsequent consequences for adolescent-emergent anhedonia and contributes to risk and resilience to trauma and stress in adulthood.
Collapse
Affiliation(s)
- Victoria B Risbrough
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.
- Center of Excellence for Stress and Mental Health, San Diego Veterans Administration, La Jolla, CA, USA.
| | - Laura M Glynn
- Department of Psychology, Chapman University, Orange, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Elysia P Davis
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Department of Psychology, University of Denver, Denver, CO, USA
| | - Curt A Sandman
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Hal S Stern
- Department of Statistics, University of California, Irvine, CA, USA
| | - David B Keator
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Department of Computer Science, University of California, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, CA, USA
| | - Michael A Yassa
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California, Irvine, CA, USA
| | - Dewleen G Baker
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Center of Excellence for Stress and Mental Health, San Diego Veterans Administration, La Jolla, CA, USA
| |
Collapse
|
48
|
Jaric I, Rocks D, Cham H, Herchek A, Kundakovic M. Sex and Estrous Cycle Effects on Anxiety- and Depression-Related Phenotypes in a Two-Hit Developmental Stress Model. Front Mol Neurosci 2019; 12:74. [PMID: 31031589 PMCID: PMC6470284 DOI: 10.3389/fnmol.2019.00074] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/11/2019] [Indexed: 12/13/2022] Open
Abstract
Stress during sensitive developmental periods can adversely affect physical and psychological development and contribute to later-life mental disorders. In particular, adverse experiences during childhood dramatically increase the risk for the development of depression and anxiety disorders. Although women of reproductive age are twice as likely to develop anxiety and depression than men of the corresponding age, little is known about sex-specific factors that promote or protect against the development of psychopathology. To examine potential developmental mechanisms driving sex disparity in risk for anxiety and depression, we established a two-hit developmental stress model including maternal separation in early life followed by social isolation in adolescence. Our study shows complex interactions between early-life and adolescent stress, between stress and sex, and between stress and female estrogen status in shaping behavioral phenotypes of adult animals. In general, increased locomotor activity and body weight reduction were the only two phenotypes where two stressors showed synergistic activity. In terms of anxiety- and depression-related phenotypes, single exposure to early-life stress had the most significant impact and was female-specific. We show that early-life stress disrupts the protective role of estrogen in females, and promotes female vulnerability to anxiety- and depression-related phenotypes associated with the low-estrogenic state. We found plausible transcriptional and epigenetic alterations in psychiatric risk genes, Nr3c1 and Cacna1c, that likely contributed to the stress-induced behavioral effects. In addition, two general transcriptional regulators, Egr1 and Dnmt1, were found to be dysregulated in maternally-separated females and in animals exposed to both stressors, respectively, providing insights into possible transcriptional mechanisms that underlie behavioral phenotypes. Our findings provide a novel insight into developmental risk factors and biological mechanisms driving sex differences in depression and anxiety disorders, facilitating the search for more effective, sex-specific treatments for these disorders.
Collapse
Affiliation(s)
- Ivana Jaric
- Department of Biological Sciences, Fordham University, Bronx, NY, United States
| | - Devin Rocks
- Department of Biological Sciences, Fordham University, Bronx, NY, United States
| | - Heining Cham
- Department of Psychology, Fordham University, Bronx, NY, United States
| | - Alice Herchek
- Department of Biological Sciences, Fordham University, Bronx, NY, United States
| | - Marija Kundakovic
- Department of Biological Sciences, Fordham University, Bronx, NY, United States
| |
Collapse
|
49
|
Effects of lipopolysaccharide administration and maternal deprivation on anxiety and depressive symptoms in male and female Wistar rats: Neurobehavioral and biochemical assessments. Behav Brain Res 2019; 362:46-55. [DOI: 10.1016/j.bbr.2019.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/30/2018] [Accepted: 01/07/2019] [Indexed: 01/06/2023]
|
50
|
Umeoka EHL, Robinson EJ, Turimella SL, van Campen JS, Motta-Teixeira LC, Sarabdjitsingh RA, Garcia-Cairasco N, Braun K, de Graan PN, Joëls M. Hyperthermia-induced seizures followed by repetitive stress are associated with age-dependent changes in specific aspects of the mouse stress system. J Neuroendocrinol 2019; 31:e12697. [PMID: 30773738 DOI: 10.1111/jne.12697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/04/2019] [Accepted: 02/13/2019] [Indexed: 11/28/2022]
Abstract
Stress is among the most frequently self-reported factors provoking epileptic seizures in children and adults. It is still unclear, however, why some people display stress-sensitive seizures and others do not. Recently, we showed that young epilepsy patients with stress-sensitive seizures exhibit a dysregulated hypothalamic-pituitary-adrenal (HPA)-axis. Most likely, this dysregulation gradually develops, and is triggered by stressors occurring early in life (early-life stress [ELS]). ELS may be particularly impactful when overlapping with the period of epileptogenesis. To examine this in a controlled and prospective manner, the present study investigated the effect of repetitive variable stressors or control treatment between postnatal day (PND) 12 and 24 in male mice exposed on PND10 to hyperthermia (HT)-induced prolonged seizures (control: normothermia). A number of peripheral and central indices of HPA-axis activity were evaluated at pre-adolescent and young adult age (ie, at PND25 and 90, respectively). At PND25 but not at PND90, body weight gain and absolute as well as relative (to body weight) thymus weight were reduced by ELS (vs control), whereas relative adrenal weight was enhanced, confirming the effectiveness of the stress treatment. Basal and stress-induced corticosterone levels were unaffected, though, by ELS at both ages. HT by itself did not affect any of these peripheral markers of HPA-axis activity, nor did it interact with ELS. However, centrally we did observe age-specific interaction effects of HT and ELS with regard to hippocampal glucocorticoid receptor mRNA expression, neurogenesis with the immature neurone marker doublecortin and the number of hilar (ectopic) granule cells using Prox1 staining. This lends some support to the notion that exposure to repetitive stress after HT-induced seizures may dysregulate central components of the stress system in an age-dependent manner. Such dysregulation could be one of the mechanisms conferring higher vulnerability of individuals with epilepsy to develop seizures in the face of stress.
Collapse
Affiliation(s)
- Eduardo H L Umeoka
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Edward J Robinson
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Sada Lakshmi Turimella
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Jolien S van Campen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lívia C Motta-Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - R Angela Sarabdjitsingh
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Norberto Garcia-Cairasco
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kees Braun
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pierre N de Graan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Marian Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|