1
|
Gómez-Ilescas A, Silveira PP. Early adversity and the comorbidity between metabolic disease and psychopathology. J Physiol 2025. [PMID: 40349327 DOI: 10.1113/jp285927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/01/2025] [Indexed: 05/14/2025] Open
Abstract
Although the co-existence of metabolic and psychiatric disorders in the same individual (comorbidity) is very prevalent, the mechanisms by which these disorders co-occur are poorly understood, but a history of early-life adversity is a common developmental risk factor. Exposure to adverse environments during critical periods of development (e.g. fetal life and infancy) modifies the metabolism and the function of the brain persistently, influencing behaviours that contribute to both metabolic and mental health disarrangements over the life course. We will review molecular and clinical evidence supporting the notion that early adversity is an important risk factor for the comorbidity between metabolic and psychiatric conditions. We will also discuss the possible mechanisms involved: neurometabolic programming, epigenetic alterations and the cumulative effects of altered inflammatory and oxidative pathways linked to early adversity.
Collapse
Affiliation(s)
| | - Patricia Pelufo Silveira
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, QC, Canada
- Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
2
|
Mikulovic S, Lenschow C. Neural control of sex differences in affiliative and prosocial behaviors. Neurosci Biobehav Rev 2025; 171:106039. [PMID: 39914700 DOI: 10.1016/j.neubiorev.2025.106039] [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] [Received: 06/12/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
Social interactions are vital for various taxa and species. Prosocial and affiliative dynamics within a group and between individuals are not only pleasurable and rewarding, but also appear to actively contribute to well-being, cognitive performance, and disease prevention. Moreover, disturbances in acting or being prosocial can represent a major burden for an individual and their affective partners. These disruptions are evident across a spectrum of neuropsychiatric conditions, including depression and autism spectrum disorders. Importantly, interactive patterns of prosocial and affiliative behavior can vary with sex. The fact that genders are differentially affected by neuropsychiatric disorders associated with social impairment underscores the high importance of this research in uncovering the underlying neural correlates and mechanisms. This review focuses on elucidating sex-related differences in prosocial and affiliative behaviors and their potential association with sexually different neural correlates. Specifically, we aim to shed light on the complex interplay between sex, behavior, and neurobiology in affiliative and prosocial interaction patterns.
Collapse
Affiliation(s)
- Sanja Mikulovic
- Leibniz Institute for Neurobiology, Brennecke Straße, Magdeburg, Germany.
| | - Constanze Lenschow
- Otto-von-Guericke University Magdeburg, Institute of Biology (House 91), Leipziger Straße 44, Magdeburg 39120, Germany.
| |
Collapse
|
3
|
Naoi M, Wu Y, Maruyama W, Shamoto-Nagai M. Phytochemicals Modulate Biosynthesis and Function of Serotonin, Dopamine, and Norepinephrine for Treatment of Monoamine Neurotransmission-Related Psychiatric Diseases. Int J Mol Sci 2025; 26:2916. [PMID: 40243512 PMCID: PMC11988947 DOI: 10.3390/ijms26072916] [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] [Received: 01/12/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Serotonin (5-HT), dopamine (DA), and norepinephrine (NE) are key monoamine neurotransmitters regulating behaviors, mood, and cognition. 5-HT affects early brain development, and its dysfunction induces brain vulnerability to stress, raising the risk of depression, anxiety, and autism in adulthood. These neurotransmitters are synthesized from tryptophan and tyrosine via hydroxylation and decarboxylation, and are metabolized by monoamine oxidase (MAO). This review aims to summarize the current findings on the role of dietary phytochemicals in modulating monoamine neurotransmitter biosynthesis, metabolism, and function, with an emphasis on their potential therapeutic applications in neuropsychiatric disorders. Phytochemicals exert antioxidant, neurotrophic, and neurohormonal activities, regulate gene expression, and induce epigenetic modifications. Phytoestrogens activate the estrogen receptors or estrogen-responsive elements of the promoter of target genes, enhance transcription of tryptophan hydroxylase and tyrosine hydroxylase, while inhibiting that of MAO. These compounds also influence the interaction between genetic and environmental factors, potentially reversing dysregulated neurotransmission and the brain architecture associated with neuropsychiatric conditions. Despite promising preclinical findings, clinical applications of phytochemicals remain challenging. Advances in nanotechnology and targeted delivery systems offer potential solutions to enhance clinical efficacy. This review discusses mechanisms, challenges, and strategies, underscoring the need for further research to advance phytochemical-based interventions for neuropsychiatric diseases.
Collapse
Affiliation(s)
- Makoto Naoi
- Department of Health and Nutritional Sciences, Faculty of Health Sciences, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin 320-195, Aichi, Japan; (Y.W.); (W.M.); (M.S.-N.)
| | | | | | | |
Collapse
|
4
|
Veríssimo LF, Alves FHF, Estrada VB, da Costa Marques LA, Andrade KC, Bonancea AM, Okano NT, Corrêa FMDA, Pelosi GG. Cardiovascular effects of early maternal separation and escitalopram treatment in rats with depressive-like behaviour. Auton Neurosci 2024; 256:103223. [PMID: 39616948 DOI: 10.1016/j.autneu.2024.103223] [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] [Received: 07/17/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024]
Abstract
Depression and cardiovascular diseases are two of the world's major health problems. Escitalopram (ESC) is widely used because of its safety in relation to other drugs in that class; however, it can affect the cardiovascular system. The present study evaluated the cardiovascular parameters of depressive-like male rats and the cardiovascular effects of ESC treatment on that condition. The EMS protocol consisted of separating the litter from the dam for 3 h over 13 days. Animals were anesthetized with tribromoethanol (250 mg/kg, intraperitoneally) and the catheters were inserted into the femoral and into the femoral vein. Depressive-like rats showed an increase in the pressor response to phenylephrine (Emax:depressive = 50.36 ± 2.997 mmHg; non-depressive = 39.51 ± 3.328 mmHg; p < 0.05) and a reduction in the EC50 (depressive = 0.6203 ± 0.03005 μg/kg; non-depressive = 0.7320 ± 0.03519 μg/kg; p < 0.05) with no change in the other cardiovascular parameters. After treatment with ESC, a reduction of intrinsic heart rate was observed in the depressive-like rats (control: 342 ± 6 bpm; ESC: 316 ± 5 bpm; p < 0.05). In addition, ESC treatment increased the bradycardic (control: -97.81 ± 8.3 bpm; ESC: -137.1 ± 12.31 bpm; p = 0.0236; t = 2.502) during the baroreflex response, caused by an increase in cardiac parasympathetic modulation in the heart, in depressive-like rats (p < 0.001). The findings suggest that depressive-like rats showed cardiovascular changes, and that ESC treatment was able to reverse these changes, suggesting that ESC has a good safety profile for depressive patients with cardiovascular disease due to increased parasympathetic modulation.
Collapse
Affiliation(s)
- Luiz Fernando Veríssimo
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| | | | - Viviane Batista Estrada
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| | | | - Karoliny Coelho Andrade
- Department of Health Sciences Faculty of Medicine Federal University of Lavras (UFLA), Lavras, Minas Gerais, Brazil
| | - Amanda Monteiro Bonancea
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| | - Natália Tavares Okano
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| | | | - Gislaine Garcia Pelosi
- Department of Physiological Sciences, Center of Biological Sciences, State University of Londrina, Paraná, Brazil
| |
Collapse
|
5
|
Ramkumar R, Edge-Partington M, Terstege DJ, Adigun K, Ren Y, Khan NS, Rouhi N, Jamani NF, Tsutsui M, Epp JR, Sargin D. Long-Term Impact of Early-Life Stress on Serotonin Connectivity. Biol Psychiatry 2024; 96:287-299. [PMID: 38316332 DOI: 10.1016/j.biopsych.2024.01.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Chronic childhood stress is a prominent risk factor for developing affective disorders, yet mechanisms underlying this association remain unclear. Maintenance of optimal serotonin (5-HT) levels during early postnatal development is critical for the maturation of brain circuits. Understanding the long-lasting effects of early-life stress (ELS) on serotonin-modulated brain connectivity is crucial to develop treatments for affective disorders arising from childhood stress. METHODS Using a mouse model of chronic developmental stress, we determined the long-lasting consequences of ELS on 5-HT circuits and behavior in females and males. Using FosTRAP mice, we cross-correlated regional c-Fos density to determine brain-wide functional connectivity of the raphe nucleus. We next performed in vivo fiber photometry to establish ELS-induced deficits in 5-HT dynamics and optogenetics to stimulate 5-HT release to improve behavior. RESULTS Adult female and male mice exposed to ELS showed heightened anxiety-like behavior. ELS further enhanced susceptibility to acute stress by disrupting the brain-wide functional connectivity of the raphe nucleus and the activity of 5-HT neuron population, in conjunction with increased orbitofrontal cortex (OFC) activity and disrupted 5-HT release in medial OFC. Optogenetic stimulation of 5-HT terminals in the medial OFC elicited an anxiolytic effect in ELS mice in a sex-dependent manner. CONCLUSIONS These findings suggest a significant disruption in 5-HT-modulated brain connectivity in response to ELS, with implications for sex-dependent vulnerability. The anxiolytic effect of the raphe-medial OFC circuit stimulation has potential implications for developing targeted stimulation-based treatments for affective disorders that arise from early life adversities.
Collapse
Affiliation(s)
- Raksha Ramkumar
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Moriah Edge-Partington
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Dylan J Terstege
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kabirat Adigun
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yi Ren
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nazmus S Khan
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Nahid Rouhi
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Naila F Jamani
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mio Tsutsui
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan R Epp
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derya Sargin
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
6
|
Lauby SC, Lapp HE, Salazar M, Semyrenko S, Chauhan D, Margolis AE, Champagne FA. Postnatal maternal care moderates the effects of prenatal bisphenol exposure on offspring neurodevelopmental, behavioral, and transcriptomic outcomes. PLoS One 2024; 19:e0305256. [PMID: 38861567 PMCID: PMC11166292 DOI: 10.1371/journal.pone.0305256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Bisphenols (BP), including BPA and "BPA-free" structural analogs, are commonly used plasticizers that are present in many plastics and are known endocrine disrupting chemicals. Prenatal exposure to BPA has been associated with negative neurodevelopmental and behavioral outcomes in children and in rodent models. Prenatal BPA exposure has also been shown to impair postnatal maternal care provisioning, which can also affect offspring neurodevelopment and behavior. However, there is limited knowledge regarding the biological effects of prenatal exposure to bisphenols other than BPA and the interplay between prenatal bisphenol exposure and postnatal maternal care on adult behavior. The purpose of the current study was to determine the interactive impact of prenatal bisphenol exposure and postnatal maternal care on neurodevelopment and behavior in rats. Our findings suggest that the effects of prenatal bisphenol exposure on eye-opening, adult attentional set shifting and anxiety-like behavior in the open field are dependent on maternal care in the first five days of life. Interestingly, maternal care might also attenuate the effects of prenatal bisphenol exposure on eye opening and adult attentional set shifting. Finally, transcriptomic profiles in male and female medial prefrontal cortex and amygdala suggest that the interactive effects of prenatal bisphenol exposure and postnatal maternal care converge on estrogen receptor signaling and are involved in biological processes related to gene expression and protein translation and synthesis. Overall, these findings indicate that postnatal maternal care plays a critical role in the expression of the effects of prenatal bisphenol exposure on neurodevelopment and adult behavior. Understanding the underlying biological mechanisms involved might allow us to identify potential avenues to mitigate the adverse effects of prenatal bisphenol exposure and improve health and well-being in human populations.
Collapse
Affiliation(s)
- Samantha C. Lauby
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
- Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin, Austin, Texas, United States of America
| | - Hannah E. Lapp
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
| | - Melissa Salazar
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
| | - Sofiia Semyrenko
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
| | - Danyal Chauhan
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
| | - Amy E. Margolis
- Department of Psychiatry, Columbia University Irving Medical Center, New York City, New York, United States of America
| | - Frances A. Champagne
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
- Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
7
|
Bennett SN, Chang AB, Rogers FD, Jones P, Peña CJ. Thyroid hormones mediate the impact of early-life stress on ventral tegmental area gene expression and behavior. Horm Behav 2024; 159:105472. [PMID: 38141539 PMCID: PMC10922504 DOI: 10.1016/j.yhbeh.2023.105472] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/17/2023] [Accepted: 12/12/2023] [Indexed: 12/25/2023]
Abstract
Proper thyroid function is essential to the developing brain, including dopamine neuron differentiation, growth, and maintenance. Stress across the lifespan impacts thyroid hormone signaling and anxiety disorders and depression have been associated with thyroid dysfunction (both hypo- and hyper-active). However, less is known about how stress during postnatal development impacts thyroid function and related brain development. Our previous work in mice demonstrated that early-life stress (ELS) transiently impinged on expression of a transcription factor in dopamine neurons, Otx2, shown to be regulated by thyroid hormones. We hypothesized that thyroid hormone signaling may link experience of ELS with transcriptional dysregulation within the dopaminergic midbrain, and ultimately behavior. Here, we find that ELS transiently increases thyroid-stimulating hormone levels (inversely related to thyroid signaling) in both male and female mice at P21, an effect which recovers by adolescence. We next tested whether transient treatment of ELS mice with synthetic thyroid hormone (levothyroxine, LT4) could ameliorate the impact of ELS on sensitivity to future stress, and on expression of genes related to dopamine neuron development and maintenance, thyroid signaling, and plasticity within the ventral tegmental area. Among male mice, but not females, juvenile LT4 treatment prevented hypersensitivity to adult stress. We also found that rescuing developmental deficits in thyroid hormone signaling after ELS restored levels of some genes altered directly by ELS, and prevented alterations in expression of other genes sensitive to the second hit of adult stress. These findings suggest that thyroid signaling mediates the deleterious impact of ELS on VTA development, and that temporary treatment of hypothyroidism after ELS may be sufficient to prevent future stress hypersensitivity.
Collapse
Affiliation(s)
| | - Austin B Chang
- Princeton Neuroscience Institute, Princeton University, USA
| | - Forrest D Rogers
- Princeton Neuroscience Institute, Princeton University, USA; Department of Molecular Biology, Princeton University, USA
| | - Parker Jones
- Princeton Neuroscience Institute, Princeton University, USA
| | | |
Collapse
|
8
|
Nicolaides NC, Kanaka-Gantenbein C, Pervanidou P. Developmental Neuroendocrinology of Early-Life Stress: Impact on Child Development and Behavior. Curr Neuropharmacol 2024; 22:461-474. [PMID: 37563814 PMCID: PMC10845081 DOI: 10.2174/1570159x21666230810162344] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/22/2023] [Accepted: 03/15/2023] [Indexed: 08/12/2023] Open
Abstract
Our internal balance, or homeostasis, is threatened or perceived as threatened by stressful stimuli, the stressors. The stress system is a highly conserved system that adjusts homeostasis to the resting state. Through the concurrent activation of the hypothalamic-pituitary-adrenal axis and the locus coeruleus/norepinephrine-autonomic nervous systems, the stress system provides the appropriate physical and behavioral responses, collectively termed as "stress response", to restore homeostasis. If the stress response is prolonged, excessive or even inadequate, several acute or chronic stress-related pathologic conditions may develop in childhood, adolescence and adult life. On the other hand, earlylife exposure to stressors has been recognized as a major contributing factor underlying the pathogenesis of non-communicable disorders, including neurodevelopmental disorders. Accumulating evidence suggests that early-life stress has been associated with an increased risk for attention deficit hyperactivity disorder and autism spectrum disorder in the offspring, although findings are still controversial. Nevertheless, at the molecular level, early-life stressors alter the chemical structure of cytosines located in the regulatory regions of genes, mostly through the addition of methyl groups. These epigenetic modifications result in the suppression of gene expression without changing the DNA sequence. In addition to DNA methylation, several lines of evidence support the role of non-coding RNAs in the evolving field of epigenetics. In this review article, we present the anatomical and functional components of the stress system, discuss the proper, in terms of quality and quantity, stress response, and provide an update on the impact of early-life stress on child development and behavior.
Collapse
Affiliation(s)
- Nicolas C. Nicolaides
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens, School of Medicine, ‘Aghia Sophia’ Children's Hospital, Athens, 11527, Greece
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
- School of Medicine, University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Christina Kanaka-Gantenbein
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens, School of Medicine, ‘Aghia Sophia’ Children's Hospital, Athens, 11527, Greece
| | - Panagiota Pervanidou
- Unit of Developmental and Behavioral Pediatrics, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children's Hospital, Athens, Greece
| |
Collapse
|
9
|
Maayan L, Maayan M. Inflammatory mediation of the relationship between early adversity and major depressive disorder: A systematic review. J Psychiatr Res 2024; 169:364-377. [PMID: 38154266 DOI: 10.1016/j.jpsychires.2023.11.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/30/2023] [Accepted: 11/15/2023] [Indexed: 12/30/2023]
Abstract
Early adverse experience is related to psychiatric illness that occurs decades later. The mechanisms underlying this phenomenon have not been fully identified. There is a translational and clinical literature linking early adversity with Major Depressive Disorder (MDD) and inflammation. We reviewed articles that examine whether inflammation mediates this relationship. METHODS Literature review of PUB MED, CINAHL and APA Psycinfo articles that explicitly examine inflammation as a mediator between early adversity and depression using ((((((((((adversity) OR (trauma)) OR (maltreatment)) OR (child abuse)) AND (inflammation)) OR (inflammatory cytokines)) OR (crp)) OR (il-6)) OR (tnf)) AND (mediates)) AND (depression))))))))) as key words. RESULTS 2842 articles were initially identified. 1338 non-human studies were excluded and 512 more were filtered out as reviews. The remaining 992 titles and, when necessary, abstracts and manuscripts were reviewed and 956 were removed as being of other non-related phenomena. Four additional studies were added by hand searching the references of remaining studies. Out of these 40, 15 explicitly examined inflammation as a mediator of the relationship between early adversity and later depression. Approximately half (8/15) showed evidence that inflammation mediated the relationship between early adversity and depression. Sensitivity analyses showed that studies taking place in clinical populations, in youth and those that used the Adverse Childhood Events Scale to measure adversity, and IL-6 and TNF-α (as opposed to CRP) to measure inflammation were most likely to show mediation. CONCLUSIONS There is evidence to support the model of inflammation mediating the relationship between early adversity and depression. Certain measures in clinical populations appear more likely to support this model. Further study with more standardized, robust methods will help to answer this question more definitively and may elucidate a subtype of depression related to early adversity by alterations in immune function.
Collapse
Affiliation(s)
- Lawrence Maayan
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.
| | - Michal Maayan
- Skidmore College, 815 N. Broadway, Saratoga Springs, NY, 12866, USA
| |
Collapse
|
10
|
Gundacker A, Glat M, Wais J, Stoehrmann P, Pollak A, Pollak DD. Early-life iron deficiency persistently disrupts affective behaviour in mice. Ann Med 2023; 55:1265-1277. [PMID: 37096819 PMCID: PMC10132221 DOI: 10.1080/07853890.2023.2191003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/09/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND/OBJECTIVE Iron deficiency (ID) is the most common nutrient deficiency, affecting two billion people worldwide, including about 30% of pregnant women. During gestation, the brain is particularly vulnerable to environmental insults, which can irrevocably impair critical developmental processes. Consequently, detrimental consequences of early-life ID for offspring brain structure and function have been described. Although early life ID has been associated with an increased long-term risk for several neuropsychiatric disorders, the effect on depressive disorders has remained unresolved. MATERIALS AND METHODS A mouse model of moderate foetal and neonatal ID was established by keeping pregnant dams on an iron-deficient diet throughout gestation until postnatal day 10. The ensuing significant decrease of iron content in the offspring brain, as well as the impact on maternal behaviour and offspring vocalization was determined in the first postnatal week. The consequences of early-life ID for depression- and anxiety-like behaviour in adulthood were revealed employing dedicated behavioural assays. miRNA sequencing of hippocampal tissue of offspring revealed specific miRNAs signatures accompanying the behavioural deficits of foetal and neonatal ID in the adult brain. RESULTS Mothers receiving iron-deficient food during pregnancy and lactation exhibited significantly less licking and grooming behaviour, while active pup retrieval and pup ultrasonic vocalizations were unaltered. Adult offspring with a history of foetal and neonatal ID showed an increase in depression- and anxiety-like behaviour, paralleled by a deranged miRNA expression profile in the hippocampus, specifically levels of miR200a and miR200b. CONCLUSION ID during the foetal and neonatal periods has life-long consequences for affective behaviour in mice and leaves a specific and persistent mark on the expression of miRNAs in the brain. Foetal and neonatal ID needs to be further considered as risk factor for the development of depression and anxiety disorders later in life.Key MessagesMarginal reduction of gestational alimentary iron intake decreases brain iron content of the juvenile offspring.Early-life ID is associated with increased depression- and anxiety-like behaviour in adulthood.Reduction of maternal alimentary iron intake during pregnancy is reflected in an alteration of miRNA signatures in the adult offspring brain.
Collapse
Affiliation(s)
- Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Micaela Glat
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jonathan Wais
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Peter Stoehrmann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Arnold Pollak
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Daniela D. Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
11
|
Lauby SC, Lapp HE, Salazar M, Semyrenko S, Chauhan D, Margolis AE, Champagne FA. Postnatal maternal care moderates the effects of prenatal bisphenol exposure on offspring neurodevelopmental, behavioral, and transcriptomic outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558481. [PMID: 37786706 PMCID: PMC10541647 DOI: 10.1101/2023.09.19.558481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Bisphenols (BPs), including BPA and "BPA-free" structural analogs, are commonly used plasticizers that are present in many plastics and are known endocrine disrupting chemicals. Prenatal exposure to BPA has been associated with negative neurodevelopmental and behavioral outcomes in children and rodent models. Prenatal BPA exposure has also been shown to impair postnatal maternal care provisioning, which can also affect offspring neurodevelopment and behavior. However, there is limited knowledge regarding the biological effects of prenatal exposure to bisphenols other than BPA and the interplay between prenatal BP exposure and postnatal maternal care on adult behavior. The purpose of the current study was to determine the interactive impact of prenatal BP exposure and postnatal maternal care on neurodevelopment and behavior. Our findings suggest that the effects of prenatal BP exposure on eye-opening, adult attentional set shifting and anxiety-like behavior in the open field are dependent on maternal care in the first five days of life. Interestingly, maternal care might also attenuate the effects of prenatal BP exposure on eye opening and adult attentional set shifting. Finally, transcriptomic profiles in male and female medial prefrontal cortex and amygdala suggest that the interactive effects of prenatal BP exposure and postnatal maternal care converge on estrogen receptor signaling and are involved in biological processes related to gene expression and protein translation and synthesis. Overall, these findings indicate that postnatal maternal care plays a critical role in the expression of the effects of prenatal BP exposure on neurodevelopment and adult behavior. Understanding the underlying biological mechanisms involved might allow us to identify potential avenues to mitigate the adverse effects of prenatal BP exposure and improve health and well-being in human populations.
Collapse
Affiliation(s)
- Samantha C Lauby
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
- Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin
| | - Hannah E Lapp
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
| | - Melissa Salazar
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
| | - Sofiia Semyrenko
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
| | - Danyal Chauhan
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
| | - Amy E Margolis
- Department of Psychiatry, Columbia University Irving Medical Center
| | - Frances A Champagne
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
- Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin
| |
Collapse
|
12
|
Sur D, Agranyoni O, Kirby M, Cohen N, Bagaev A, Karandasheva K, Shmerkin E, Gorobets D, Savita BK, Avneri R, Divon MS, Lax E, Michaelevski I, Pinhasov A. Nurture outpaces nature: fostering with an attentive mother alters social dominance in a mouse model of stress sensitivity. Mol Psychiatry 2023; 28:3816-3828. [PMID: 37845494 DOI: 10.1038/s41380-023-02273-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023]
Abstract
Maternal care is critical for epigenetic programming during postnatal brain development. Stress is recognized as a critical factor that may affect maternal behavior, yet owing to high heterogeneity in stress response, its impact varies among individuals. We aimed here to understand the connection between inborn stress vulnerability, maternal care, and early epigenetic programming using mouse populations that exhibit opposite poles of the behavioral spectrum (social dominance [Dom] and submissiveness [Sub]) and differential response to stress. In contrast to stress-resilient Dom dams, stress-vulnerable Sub dams exhibit significantly lower maternal attachment, serum oxytocin, and colonic Lactobacillus reuteri populations. Sub offspring showed a reduced hippocampal expression of key methylation genes at postnatal day (PND) 7 and a lack of developmentally-dependent increase in 5-methylcytosine (5-mC) at PND 21. In addition, Sub pups exhibit significant hypermethylation of gene promoters connected with glutamatergic synapses and behavioral responses. We were able to reverse the submissive endophenotype through cross-fostering Sub pups with Dom dams (Sub/D). Thus, Sub/D pups exhibited elevated hippocampal expression of DNMT3A at PND 7 and increased 5-mC levels at PND 21. Furthermore, adult Sub/D offspring exhibited increased sociability, social dominance, and hippocampal glutamate and monoamine levels resembling the neurochemical profile of Dom mice. We postulate that maternal inborn stress vulnerability governs epigenetic patterning sculpted by maternal care and intestinal microbiome diversity during early developmental stages and shapes the array of gene expression patterns that may dictate neuronal architecture with a long-lasting impact on stress sensitivity and the social behavior of offspring.
Collapse
Affiliation(s)
- Debpali Sur
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Oryan Agranyoni
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Michael Kirby
- Dr. Miriam and Sheldon G. Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Naamah Cohen
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Anastasia Bagaev
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Kristina Karandasheva
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Elena Shmerkin
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Denis Gorobets
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Brajesh Kumar Savita
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Raphael Avneri
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Mali-Salmon Divon
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
- Dr. Miriam and Sheldon G. Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Elad Lax
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Izhak Michaelevski
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel.
- Dr. Miriam and Sheldon G. Adelson School of Medicine, Ariel University, Ariel, Israel.
| |
Collapse
|
13
|
Bennett SN, Chang AB, Rogers FD, Jones P, Peña CJ. Thyroid hormones mediate the impact of early-life stress on ventral tegmental area gene expression and behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554785. [PMID: 37662236 PMCID: PMC10473690 DOI: 10.1101/2023.08.25.554785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Proper thyroid function is essential to the developing brain, including dopamine neuron differentiation, growth, and maintenance. Stress across the lifespan impacts thyroid hormone signaling and anxiety disorders and depression have been associated with thyroid dysfunction (both hypo- and hyper-active). However, less is known about how stress during postnatal development impacts thyroid function and related brain development. Our previous work in mice demonstrated that early-life stress (ELS) transiently impinged on expression of a transcription factor in dopamine neurons shown to be regulated by thyroid hormones. We hypothesized that thyroid hormone signaling may link experience of ELS with transcriptional dysregulation within the dopaminergic midbrain, and ultimately behavior. Here, we find that ELS transiently increases thyroid-stimulating hormone levels (inversely related to thyroid signaling) in both male and female mice at P21, an effect which recovers by adolescence. We next tested whether transient treatment of ELS mice with synthetic thyroid hormone (levothyroxine, LT4) could ameliorate the impact of ELS on sensitivity to future stress, and on expression of genes related to dopamine neuron development and maintenance, thyroid signaling, and plasticity within the ventral tegmental area. Among male mice, but not females, juvenile LT4 treatment prevented hypersensitivity to adult stress. We also found that rescuing developmental deficits in thyroid hormone signaling after ELS restored levels of some genes altered directly by ELS, and prevented alterations in expression of other genes sensitive to the second hit of adult stress. These findings suggest that thyroid signaling mediates the deleterious impact of ELS on VTA development, and that temporary treatment of hypothyroidism after ELS may be sufficient to prevent future stress hypersensitivity.
Collapse
|
14
|
Raymann S, Schalbetter SM, Schaer R, Bernhardt AC, Mueller FS, Meyer U, Weber-Stadlbauer U. Late prenatal immune activation in mice induces transgenerational effects via the maternal and paternal lineages. Cereb Cortex 2023; 33:2273-2286. [PMID: 36857721 DOI: 10.1093/cercor/bhac207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 11/14/2022] Open
Abstract
Prenatal exposure to infectious or noninfectious immune activation is an environmental risk factor for neurodevelopmental disorders and mental illnesses. Recent research using animal models suggests that maternal immune activation (MIA) during early to middle stages of pregnancy can induce transgenerational effects on brain and behavior, likely via inducing stable epigenetic modifications across generations. Using a mouse model of viral-like MIA, which is based on gestational treatment with poly(I:C), the present study explored whether transgenerational effects can also emerge when MIA occurs in late pregnancy. Our findings demonstrate that the direct descendants born to poly(I:C)-treated mothers display deficits in temporal order memory, which are similarly present in second- and third-generation offspring. These transgenerational effects were mediated via both the maternal and paternal lineages and were accompanied by transient changes in maternal care. In addition to the cognitive effects, late prenatal immune activation induced generation-spanning effects on the prefrontal expression of gamma-aminobutyric acid (GABA)ergic genes, including parvalbumin and distinct alpha-subunits of the GABAA receptor. Together, our results suggest that MIA in late pregnancy has the potential to affect cognitive functions and prefrontal gene expression patterns in multiple generations, highlighting its role in shaping disease risk across generations.
Collapse
Affiliation(s)
- Stephanie Raymann
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Sina M Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Ron Schaer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Alexandra C Bernhardt
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
15
|
Nolvi S, Merz EC, Kataja EL, Parsons CE. Prenatal Stress and the Developing Brain: Postnatal Environments Promoting Resilience. Biol Psychiatry 2022; 93:942-952. [PMID: 36870895 DOI: 10.1016/j.biopsych.2022.11.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 12/25/2022]
Abstract
Heightened maternal stress during pregnancy is associated with atypical brain development and an elevated risk for psychopathology in offspring. Supportive environments during early postnatal life may promote brain development and reverse atypical developmental trajectories induced by prenatal stress. We reviewed studies focused on the role of key early environmental factors in moderating associations between prenatal stress exposure and infant brain and neurocognitive outcomes. Specifically, we focused on the associations between parental caregiving quality, environmental enrichment, social support, and socioeconomic status with infant brain and neurocognitive outcomes. We examined the evidence that these factors may moderate the effects of prenatal stress on the developing brain. Complementing findings from translational models, human research suggests that high-quality early postnatal environments are associated with indices of infant neurodevelopment that have also been associated with prenatal stress, such as hippocampal volume and frontolimbic connectivity. Human studies also suggest that maternal sensitivity and higher socioeconomic status may attenuate the effects of prenatal stress on established neurocognitive and neuroendocrine mediators of risk for psychopathology, such as hypothalamic-pituitary-adrenal axis functioning. Biological pathways that may underlie the effects of positive early environments on the infant brain, including the epigenome, oxytocin, and inflammation, are also discussed. Future research in humans should examine resilience-promoting processes in relation to infant brain development using large sample sizes and longitudinal designs. The findings from this review could be incorporated into clinical models of risk and resilience during the perinatal period and used to design more effective early programs that reduce risk for psychopathology.
Collapse
Affiliation(s)
- Saara Nolvi
- Department of Psychology and Speech-Language Pathology, Turku Institute for Advanced Studies, University of Turku, Turku, Finland; Department of Clinical Medicine, FinnBrain Birth Cohort Study, Center for Population Health Research, University of Turku, Turku, Finland.
| | - Emily C Merz
- Department of Psychology, Colorado State University, Fort Collins, Colorado
| | - Eeva-Leena Kataja
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Center for Population Health Research, University of Turku, Turku, Finland
| | - Christine E Parsons
- Department of Clinical Medicine, Interacting Minds Center, Aarhus University, Aarhus, Denmark
| |
Collapse
|
16
|
Granata L, Gluck A, Parakoyi A, Brenhouse H. One week of maternal separation induces more frequent, but less predictable, maternal caregiving behaviors. Int J Dev Neurosci 2022; 82:806-814. [PMID: 36181242 PMCID: PMC10164341 DOI: 10.1002/jdn.10230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/09/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
Rodent models of early life adversity disrupt typical interactions between dams and offspring, impacting pup development over the lifespan. Predictability of caregiver interactions is a critical feature of the environment, and unpredictability is associated with behavioral and cognitive deficits in offspring. In the maternal separation (MS) paradigm, dams are not able to engage with pups while they are separated, and maternal care is impacted even after pups and dams are reunited. Using a 3.5-h daily MS protocol in rats, the present study sought to compare diurnal patterns of maternal behavior, specifically predictability and fragmentation of care, between MS- and control-reared dams. Three observation periods were assessed (1430, 2330, and 0830) between postnatal days 8-9. Frequencies and durations of maternal behaviors were measured, including pup-directed licking/grooming, arched-back and passive nursing, and carrying pups, as well as non-pup-directed self-grooming, rearing, burrowing, nest-building, and eating. The frequency of nest entries was interpreted as a measure of fragmentation, and entropy rate was calculated from transitional probability matrices to measure predictability of maternal behavioral sequences. After dam-pup reunion, MS dams engaged in more bouts of nursing and licking/grooming and more nest entries, and behavioral sequences were less predictable than control dams. MS-induced enhancement of care is a replication of previous research, but unpredictability and fragmented care during MS is a novel finding, as these measures have not been previously reported for MS.
Collapse
Affiliation(s)
- Lauren Granata
- Psychology Department, Northeastern University, Boston, MA, USA
| | - Ayalah Gluck
- Psychology Department, Northeastern University, Boston, MA, USA
| | | | | |
Collapse
|
17
|
Lapp HE, Margolis AE, Champagne FA. Impact of a bisphenol A, F, and S mixture and maternal care on the brain transcriptome of rat dams and pups. Neurotoxicology 2022; 93:22-36. [PMID: 36041667 PMCID: PMC9985957 DOI: 10.1016/j.neuro.2022.08.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 01/19/2023]
Abstract
Products containing BPA structural analog replacements have increased in response to growing public concern over adverse effects of BPA. Although humans are regularly exposed to a mixture of bisphenols, few studies have examined effects of prenatal exposure to BPA alternatives or bisphenol mixtures. In the present study, we investigate the effect of exposure to an environmentally-relevant, low-dose (150 ug/kg body weight per day) mixture of BPA, BPS, and BPF during gestation on the brain transcriptome in Long-Evans pups and dams using Tag RNA-sequencing. We also examined the association between dam licking and grooming, which also has enduring effects on pup neural development, and the transcriptomes. Associations between licking and grooming and the transcriptome were region-specific, with the hypothalamus having the greatest number of differentially expressed genes associated with licking and grooming in both dams and pups. Prenatal bisphenol exposure also had region-specific effects on gene expression and pup gene expression was affected more robustly than dam gene expression. In dams, the prelimbic cortex had the greatest number of differentially expressed genes associated with prenatal bisphenol exposure. Prenatal bisphenol exposure changed the expression of over 2000 genes in pups, with the majority being from the pup amygdala. We used Gene Set Enrichment Analysis (GSEA) to asses enrichment of gene ontology biological processes for each region. Top GSEA terms were diverse and varied by brain region and included processes known to have strong associations with steroid hormone regulation, cilium-related terms, metabolic/biosynthetic process terms, and immune terms. Finally, hypothesis-driven analysis of genes related to estrogen response, parental behavior, and epigenetic regulation of gene expression revealed region-specific expression associated with licking and grooming and bisphenol exposure that were distinct in dams and pups. These data highlight the effects of bisphenols on multiple physiological process that are highly dependent on timing of exposure (prenatal vs. adulthood) and brain region, and reiterate the contributions of multiple environmental and experiential factors in shaping the brain.
Collapse
Affiliation(s)
- H E Lapp
- Department of Psychology, University of Texas at Austin, 108 E. Dean Keaton St, Austin, TX 78712, USA.
| | - A E Margolis
- Department of Psychiatry, Columbia University Irving Medical Center, 1051 Riverside Drive, New York, NY 10032, USA
| | - F A Champagne
- Department of Psychology, University of Texas at Austin, 108 E. Dean Keaton St, Austin, TX 78712, USA
| |
Collapse
|
18
|
Li Q, Zhao W, Kendrick KM. Affective touch in the context of development, oxytocin signaling, and autism. Front Psychol 2022; 13:967791. [PMID: 36506943 PMCID: PMC9728590 DOI: 10.3389/fpsyg.2022.967791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022] Open
Abstract
Touch represents one of our most important senses throughout life and particularly in the context of our social and emotional experiences. In this review, we draw on research on touch processing from both animal models and humans. Firstly, we briefly describe the cutaneous touch receptors and neural processing of both affective and discriminative touch. We then outline how our sense of touch develops and summarize increasing evidence demonstrating how essential early tactile stimulation is for the development of brain and behavior, with a particular focus on effects of tactile stimulation in infant animals and pediatric massage and Kangaroo care in human infants. Next, the potential mechanisms whereby early tactile stimulation influences both brain and behavioral development are discussed, focusing on its ability to promote neural plasticity changes and brain interhemispheric communication, development of social behavior and bonding, and reward sensitivity through modulation of growth factor, oxytocin, and opioid signaling. Finally, we consider the implications of evidence for atypical responses to touch in neurodevelopmental disorders such as autism spectrum disorder and discuss existing evidence and future priorities for establishing potential beneficial effects of interventions using massage or pharmacological treatments targeting oxytocin or other neurochemical systems.
Collapse
Affiliation(s)
- Qin Li
- School of Foreign Language, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Ministry of Education, Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Weihua Zhao
- Ministry of Education, Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Keith M. Kendrick
- Ministry of Education, Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
19
|
Maternal Obesity and Gut Microbiota Are Associated with Fetal Brain Development. Nutrients 2022; 14:nu14214515. [PMID: 36364776 PMCID: PMC9654759 DOI: 10.3390/nu14214515] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/28/2022] Open
Abstract
Obesity in pregnancy induces metabolic syndrome, low-grade inflammation, altered endocrine factors, placental function, and the maternal gut microbiome. All these factors impact fetal growth and development, including brain development. The lipid metabolic transporters of the maternal-fetal-placental unit are dysregulated in obesity. Consequently, the transport of essential long-chain PUFAs for fetal brain development is disturbed. The mother’s gut microbiota is vital in maintaining postnatal energy homeostasis and maternal-fetal immune competence. Obesity during pregnancy changes the gut microbiota, affecting fetal brain development. Obesity in pregnancy can induce placental and intrauterine inflammation and thus influence the neurodevelopmental outcomes of the offspring. Several epidemiological studies observed an association between maternal obesity and adverse neurodevelopment. This review discusses the effects of maternal obesity and gut microbiota on fetal neurodevelopment outcomes. In addition, the possible mechanisms of the impacts of obesity and gut microbiota on fetal brain development are discussed.
Collapse
|
20
|
Early life adversity shapes neural circuit function during sensitive postnatal developmental periods. Transl Psychiatry 2022; 12:306. [PMID: 35915071 PMCID: PMC9343623 DOI: 10.1038/s41398-022-02092-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
Early life adversity (ELA) is a major risk factor for mental illness, but the neurobiological mechanisms by which ELA increases the risk for future psychopathology are still poorly understood. Brain development is particularly malleable during prenatal and early postnatal life, when complex neural circuits are being formed and refined through an interplay of excitatory and inhibitory neural input, synaptogenesis, synaptic pruning, myelination, and neurogenesis. Adversity that influences these processes during sensitive periods of development can thus have long-lasting and pervasive effects on neural circuit maturation. In this review, we will discuss clinical and preclinical evidence for the impact of ELA on neural circuit formation with a focus on the early postnatal period, and how long-lasting impairments in these circuits can affect future behavior. We provide converging evidence from human and animal studies on how ELA alters the functional development of brain regions, neural circuits, and neurotransmitter systems that are crucial for cognition and affective behavior, including the hippocampus, the hypothalamus-pituitary-adrenal (HPA) axis, neural networks of fear responses and cognition, and the serotonin (5-HT) system. We also discuss how gene-by-environment (GxE) interactions can determine individual differences in susceptibility and resilience to ELA, as well as molecular pathways by which ELA regulates neural circuit development, for which we emphasize epigenetic mechanisms. Understanding the molecular and neurobiological mechanisms underlying ELA effects on brain function and psychopathology during early postnatal sensitive periods may have great potential to advance strategies to better treat or prevent psychiatric disorders that have their origin early in life.
Collapse
|
21
|
Walker SC, Cavieres A, Peñaloza-Sancho V, El-Deredy W, McGlone FP, Dagnino-Subiabre A. C-low threshold mechanoafferent targeted dynamic touch modulates stress resilience in rats exposed to chronic mild stress. Eur J Neurosci 2022; 55:2925-2938. [PMID: 32852872 DOI: 10.1111/ejn.14951] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/05/2020] [Accepted: 08/20/2020] [Indexed: 01/28/2023]
Abstract
Affiliative tactile interactions buffer social mammals against neurobiological and behavioral effects of stress. The aim of this study was to investigate the cutaneous mechanisms underlying such beneficial consequences of touch by determining whether daily stroking, specifically targeted to activate a velocity/force tuned class of low-threshold c-fiber mechanoreceptor (CLTM), confers resilience against established markers of chronic unpredictable mild stress (CMS). Adult male Sprague Dawley rats were exposed to 2 weeks of CMS. Throughout the CMS protocol, some rats were stroked daily, either at CLTM optimal velocity (5 cm/s) or outside the CLTM optimal range (30 cm/s). A third CMS exposed group did not receive any tactile stimulation. The effect of CMS on serum corticosterone levels, anxiety- and depressive-like behaviors in these three groups was assessed in comparison to a control group of non-CMS exposed rats. While stroking did not mitigate the effects of CMS on body weight gain, CLTM optimal velocity stroking did significantly reduce CMS-induced elevations in corticosterone following an acute forced-swim. Rats receiving CLTM optimal stroking also showed significantly fewer anxiety-like behaviors (elevated plus-maze) than the other CMS exposed rats. In terms of depressive-like behavior, whereas the same velocity-specific resilience was observed in a forced-swim test and social interaction test both groups of stroked rats spent significantly less time interacting than control rats, though they also spent significantly less time in the corner than non-stroked CMS rats. Together, these findings support the theory CLTMs play a functional role in regulating the physiological condition of the body.
Collapse
Affiliation(s)
- Susannah C Walker
- Research Centre for Brain & Behaviour, Liverpool John Moores University, Liverpool, UK
| | - Antonia Cavieres
- Laboratory of Stress Neurobiology, Faculty of Sciences, Center for Integrative Neurobiology and Pathophysiology, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
| | - Valentín Peñaloza-Sancho
- Laboratory of Stress Neurobiology, Faculty of Sciences, Center for Integrative Neurobiology and Pathophysiology, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
| | - Wael El-Deredy
- Center for Research and Development in Health Engineering, Universidad de Valparaíso, Valparaíso, Chile
| | - Francis P McGlone
- Research Centre for Brain & Behaviour, Liverpool John Moores University, Liverpool, UK.,Institute of Psychology, Health & Society, University of Liverpool, Liverpool, UK
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, Faculty of Sciences, Center for Integrative Neurobiology and Pathophysiology, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
22
|
Śmierciak N, Szwajca M, Popiela TJ, Bryll A, Karcz P, Donicz P, Turek A, Krzyściak W, Pilecki M. Redefining the Cut-Off Ranges for TSH Based on the Clinical Picture, Results of Neuroimaging and Laboratory Tests in Unsupervised Cluster Analysis as Individualized Diagnosis of Early Schizophrenia. J Pers Med 2022; 12:jpm12020247. [PMID: 35207735 PMCID: PMC8874519 DOI: 10.3390/jpm12020247] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 11/16/2022] Open
Abstract
Thyroid abnormalities, including mild forms of hypothyroidism and hyperthyroidism, are reported as risk factors for the development of a number of neuropsychiatric disorders, including schizophrenia. The diagnostic process still takes into account the extreme ranges of the accepted reference values for serum TSH since the concentration of free thyroxine in the serum does not change by definition. TSH mU/L cut-off values in psychiatric patients are currently clinically considered in the case of extremely high serum TSH levels (>4.0 mU/L). The results obtained in this study suggest that the clinically significant value has a lower TSH cut-off point with an upper limit of 2–2.5 mU/L. The criteria for the differential diagnosis of patients with schizophrenia, however, mainly take into account statutory reference ranges without a background related to the history of thyroid diseases in the family. The results indicate the need to lower the upper cut-off values for TSH among patients with early psychosis, which is related to the potential clinical significance of the obtained values both in the field of clinical evaluation and neuroimaging and laboratory evaluation parameters. The cut-off points obtained with the prior available knowledge coincided with the values established in the unsupervised clustering method, which further confirms the legitimacy of their use in the individualized diagnosis strategy of schizophrenia.
Collapse
Affiliation(s)
- Natalia Śmierciak
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
| | - Marta Szwajca
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
| | - Tadeusz J. Popiela
- Department of Radiology, Jagiellonian University Medical College, Kopernika 19, 31-501 Krakow, Poland;
- Correspondence: (T.J.P.); (W.K.); (M.P.)
| | - Amira Bryll
- Department of Radiology, Jagiellonian University Medical College, Kopernika 19, 31-501 Krakow, Poland;
| | - Paulina Karcz
- Department of Electroradiology, Jagiellonian University Medical College, Michałowskiego 12, 31-126 Krakow, Poland;
| | - Paulina Donicz
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
| | - Aleksander Turek
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Łazarza 16, 31-530 Krakow, Poland
| | - Wirginia Krzyściak
- Department of Medical Diagnostics, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
- Correspondence: (T.J.P.); (W.K.); (M.P.)
| | - Maciej Pilecki
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 21a, 31-501 Krakow, Poland; (N.Ś.); (M.S.); (P.D.); (A.T.)
- Correspondence: (T.J.P.); (W.K.); (M.P.)
| |
Collapse
|
23
|
Espina JEC, Bagamasbad PD. Synergistic gene regulation by thyroid hormone and glucocorticoid in the hippocampus. VITAMINS AND HORMONES 2021; 118:35-81. [PMID: 35180933 DOI: 10.1016/bs.vh.2021.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The hippocampus is considered the center for learning and memory in the brain, and its development and function is greatly affected by the thyroid and stress axes. Thyroid hormone (TH) and glucocorticoids (GC) are known to have a synergistic effect on developmental programs across several vertebrate species, and their effects on hippocampal structure and function are well-documented. However, there are few studies that focus on the processes and genes that are cooperatively regulated by the two hormone axes. Cross-regulation of the thyroid and stress axes in the hippocampus occurs on multiple levels such that TH can regulate the expression of the GC receptor (GR) while GC can modulate tissue sensitivity to TH by controlling the expression of TH receptor (TR) and enzymes involved in TH biosynthesis. Thyroid hormone and GC are also known to synergistically regulate the transcription of genes associated with neuronal function and development. Synergistic gene regulation by TH and GC may occur through the direct, cooperative action of TR and GR on common target genes, or by indirect mechanisms involving gene regulatory cascades activated by TR and GR. In this chapter, we describe the known physiological effects and underlying molecular mechanisms of TH and GC synergistic gene regulation in the hippocampus.
Collapse
Affiliation(s)
- Jose Ezekiel C Espina
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines
| | - Pia D Bagamasbad
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines.
| |
Collapse
|
24
|
Roubinov D, Meaney MJ, Boyce WT. Change of pace: How developmental tempo varies to accommodate failed provision of early needs. Neurosci Biobehav Rev 2021; 131:120-134. [PMID: 34547365 PMCID: PMC8648258 DOI: 10.1016/j.neubiorev.2021.09.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 07/30/2021] [Accepted: 09/16/2021] [Indexed: 01/13/2023]
Abstract
The interplay of genes and environments (GxE) is a fundamental source of variation in behavioral and developmental outcomes. Although the role of developmental time (T) in the unfolding of such interactions has yet to be fully considered, GxE operates within a temporal frame of reference across multiple timescales and degrees of biological complexity. Here, we consider GxExT interactions to understand adversity-induced developmental acceleration or deceleration whereby environmental conditions hasten or hinder children's development. To date, developmental pace changes have been largely explained through a focus on the individual: for example, how adversity "wears down" aging biological systems or how adversity accelerates or decelerates maturation to optimize reproductive fitness. We broaden such theories by positing shifts in developmental pace in response to the parent-child dyad's capacity or incapacity for meeting children's early, physiological and safety needs. We describe empirical evidence and potential neurobiological mechanisms supporting this new conceptualization of developmental acceleration and deceleration. We conclude with suggestions for future research on the developmental consequences of early adverse exposures.
Collapse
Affiliation(s)
- Danielle Roubinov
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, United States.
| | - Michael J Meaney
- Department of Psychiatry and Sackler Program for Epigenetics and Psychobiology, McGill University, Montreal, Quebec, H3H 1R4, Canada; Child and Brain Development Program, CIFAR, Toronto, Ontario, M5G 1M1, Canada; Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A ⁎STAR), 117609, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
| | - W Thomas Boyce
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, United States; Child and Brain Development Program, CIFAR, Toronto, Ontario, M5G 1M1, Canada; Department of Pediatrics, University of California, San Francisco, United States
| |
Collapse
|
25
|
Batra A, Latsko M, Portella AK, Silveira PP. Early adversity and insulin: neuroendocrine programming beyond glucocorticoids. Trends Endocrinol Metab 2021; 32:1031-1043. [PMID: 34635400 DOI: 10.1016/j.tem.2021.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023]
Abstract
Exposure to direct or contextual adversities during early life programs the functioning of the brain and other biological systems, contributing to the development of physical as well as mental health issues in the long term. While the role of glucocorticoids in mediating the outcomes of early adversity has been explored for many years, less attention has been given to insulin. Beyond its metabolic effects in the periphery, central insulin action affects synaptic plasticity, brain neurotransmission, and executive functions. Knowledge about the interactions between the peripheral metabolism and brain function from a developmental perspective can contribute to prevention and diagnosis programs, as well as early interventions for vulnerable populations.
Collapse
Affiliation(s)
- Aashita Batra
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
| | - Maeson Latsko
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Healthy Brains for Healthy Lives, McGill University, Montreal, QC, Canada
| | - Andre Krumel Portella
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Patricia P Silveira
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
26
|
Van Puyvelde M, Staring L, Schaffers J, Rivas-Smits C, Groenendijk L, Smeyers L, Collette L, Schoofs A, Van den Bossche N, McGlone F. Why do we hunger for touch? The impact of daily gentle touch stimulation on maternal-infant physiological and behavioral regulation and resilience. Infant Ment Health J 2021; 42:823-838. [PMID: 34752649 DOI: 10.1002/imhj.21949] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We report the impact of a Gentle Touch Stimulation (GTS) program. Forty-three mothers provided daily 10-min GTS with C-tactile (CT) afferent optimal stroking touch, for 4 weeks to their 3-12 weeks old infants. CT-afferents are cutaneous unmyelinated, low-threshold mechanosensitive nerves hypothesized to underly the regulatory impact of affective touch. We compared physiological and behavioral responses during a no-touch-baseline (BL), static-touch-baseline (BL-T), intervention/control (GTS/CTRL), Still Face (SF) and Reunion (RU) condition for GTS-infants versus a control-group (CTRL) at the start (T1) and end of (T2) of the program. We collected mother-infant ECG, respiration, cortisol, video-recordings, and diary-reports. At T1, physiological arousal significantly increased during SF in both groups, that is, decreased respiratory sinus arrhythmia (RSA) and R-R interval (RRI). At T2, GTS-infants showed significantly increased RSA, RRI, decreased respiration during GTS, buffering SF-arousal and allowing complete recovery during RU; CTRL-infants showed higher SF-arousal and small recovery, under initial BL-levels. Maternal cardio-respiratory showed a metabolic investment during RU. Cortisol and behavioral analyses showed higher arousal in CTRL-infants than GTS-infants at T2. We suggest that the combination of phasic short-term and tonic long-term responses to CT-optimal stroking touch, delivered in a structured daily manner, contribute to the building of infant stress regulation and resilience.
Collapse
Affiliation(s)
- Martine Van Puyvelde
- VIPER Research Unit, LIFE Department, Royal Military Academy, Brussels, Belgium.,Experimental and Applied Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Louise Staring
- Experimental and Applied Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jana Schaffers
- Experimental and Applied Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cristina Rivas-Smits
- Experimental and Applied Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Leysa Groenendijk
- Experimental and Applied Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laura Smeyers
- Experimental and Applied Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laetitia Collette
- Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Anneke Schoofs
- Experimental and Applied Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nora Van den Bossche
- Experimental and Applied Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Francis McGlone
- School of Natural Sciences & Psychology, Faculty of Science, Liverpool John Moores University, Liverpool, UK.,Institute of Psychology, Health & Society, University of Liverpool, Liverpool, UK
| |
Collapse
|
27
|
Pallarés ME, Monteleone MC, Pastor V, Grillo Balboa J, Alzamendi A, Brocco MA, Antonelli MC. Early-Life Stress Reprograms Stress-Coping Abilities in Male and Female Juvenile Rats. Mol Neurobiol 2021; 58:5837-5856. [PMID: 34409559 DOI: 10.1007/s12035-021-02527-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/06/2021] [Indexed: 01/06/2023]
Abstract
Prenatal stress (PS) is a major risk factor for the development of emotional disorders in adulthood that may be mediated by an altered hypothalamic-pituitary-adrenal axis response to stress. Although the early onset of stress-related disorders is recognized as a major public health problem, to date, there are relatively few studies that have examined the incidence of early-life stressors in younger individuals. In this study, we assessed PS impact on the stress-coping response of juvenile offspring in behavioral tests and in the induced molecular changes in the hippocampus. Furthermore, we assessed if pregnancy stress could be driving changes in patterns of maternal behavior during early lactation. We found that PS modified stress-coping abilities of both sex offspring. In the hippocampus, PS increased the expression of bdnf-IV and crfr1 and induced sex difference changes on glucocorticoids and BDNF mRNA receptor levels. PS changed the hippocampal epigenetic landscape mainly in male offspring. Stress during pregnancy enhanced pup-directed behavior of stressed dams. Our study indicates that exposure to PS, in addition to enhanced maternal behavior, induces dynamic neurobehavioral variations at juvenile ages of the offspring that should be considered adaptive or maladaptive, depending on the characteristics of the confronting environment. Our present results highlight the importance to further explore risk factors that appear early in life that will be important to allow timely prevention strategies to later vulnerability to stress-related disorders.
Collapse
MESH Headings
- Animals
- Female
- Male
- Pregnancy
- Rats
- Adaptation, Psychological
- Anxiety/etiology
- Anxiety/genetics
- Anxiety/physiopathology
- Brain-Derived Neurotrophic Factor/biosynthesis
- Brain-Derived Neurotrophic Factor/genetics
- Corticosterone/blood
- Corticotropin-Releasing Hormone/biosynthesis
- Corticotropin-Releasing Hormone/genetics
- Elevated Plus Maze Test
- Gene Expression Regulation
- Glucocorticoids/biosynthesis
- Glucocorticoids/genetics
- Hippocampus/embryology
- Hippocampus/physiology
- Hypothalamo-Hypophyseal System/embryology
- Hypothalamo-Hypophyseal System/physiopathology
- Lactation/physiology
- Lactation/psychology
- Maternal Behavior
- Pituitary-Adrenal System/embryology
- Pituitary-Adrenal System/physiopathology
- Pregnancy Complications/physiopathology
- Pregnancy Complications/psychology
- Prenatal Exposure Delayed Effects
- Rats, Wistar
- Receptor, trkB/biosynthesis
- Receptor, trkB/genetics
- Receptors, Corticotropin-Releasing Hormone/biosynthesis
- Receptors, Corticotropin-Releasing Hormone/genetics
- Receptors, Glucocorticoid/biosynthesis
- Receptors, Glucocorticoid/genetics
- Restraint, Physical/adverse effects
- Sex Characteristics
- Stress, Physiological/physiology
- Stress, Psychological/physiopathology
- Swimming
Collapse
Affiliation(s)
- María Eugenia Pallarés
- Laboratorio de Neuroprogramación Perinatal del Neurodesarrollo, Instituto de Biología Celular Y Neurociencias "Prof. Eduardo De Robertis" (IBCN)- Facultad de Medicina, Universidad de Buenos Aires, 2155 Paraguay St. CABA, C1121ABG, Buenos Aires, Argentina.
| | - Melisa Carolina Monteleone
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Verónica Pastor
- Laboratorio de Neuroprogramación Perinatal del Neurodesarrollo, Instituto de Biología Celular Y Neurociencias "Prof. Eduardo De Robertis" (IBCN)- Facultad de Medicina, Universidad de Buenos Aires, 2155 Paraguay St. CABA, C1121ABG, Buenos Aires, Argentina
| | - Jazmín Grillo Balboa
- Laboratorio de Neuroprogramación Perinatal del Neurodesarrollo, Instituto de Biología Celular Y Neurociencias "Prof. Eduardo De Robertis" (IBCN)- Facultad de Medicina, Universidad de Buenos Aires, 2155 Paraguay St. CABA, C1121ABG, Buenos Aires, Argentina
| | - Ana Alzamendi
- Instituto Multidisciplinario de Biología Celular, Universidad Nacional de La Plata, Buenos Aires, Argentina
| | - Marcela Adriana Brocco
- Instituto de Investigaciones Biotecnológicas (IIB), Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Marta Cristina Antonelli
- Laboratorio de Neuroprogramación Perinatal del Neurodesarrollo, Instituto de Biología Celular Y Neurociencias "Prof. Eduardo De Robertis" (IBCN)- Facultad de Medicina, Universidad de Buenos Aires, 2155 Paraguay St. CABA, C1121ABG, Buenos Aires, Argentina
| |
Collapse
|
28
|
Parra-Montes de Oca MA, Sotelo-Rivera I, Gutiérrez-Mata A, Charli JL, Joseph-Bravo P. Sex Dimorphic Responses of the Hypothalamus-Pituitary-Thyroid Axis to Energy Demands and Stress. Front Endocrinol (Lausanne) 2021; 12:746924. [PMID: 34745011 PMCID: PMC8565401 DOI: 10.3389/fendo.2021.746924] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
The hypothalamus-pituitary-thyroid-axis (HPT) is one of the main neuroendocrine axes that control energy expenditure. The activity of hypophysiotropic thyrotropin releasing hormone (TRH) neurons is modulated by nutritional status, energy demands and stress, all of which are sex dependent. Sex dimorphism has been associated with sex steroids whose concentration vary along the life-span, but also to sex chromosomes that define not only sexual characteristics but the expression of relevant genes. In this review we describe sex differences in basal HPT axis activity and in its response to stress and to metabolic challenges in experimental animals at different stages of development, as well as some of the limited information available on humans. Literature review was accomplished by searching in Pubmed under the following words: "sex dimorphic" or "sex differences" or "female" or "women" and "thyrotropin" or "thyroid hormones" or "deiodinases" and "energy homeostasis" or "stress". The most representative articles were discussed, and to reduce the number of references, selected reviews were cited.
Collapse
Affiliation(s)
| | | | | | | | - Patricia Joseph-Bravo
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico
| |
Collapse
|
29
|
Duclot F, Kabbaj M. Epigenetics of Aggression. Curr Top Behav Neurosci 2021; 54:283-310. [PMID: 34595741 DOI: 10.1007/7854_2021_252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Aggression is a complex behavioral trait modulated by both genetic and environmental influences on gene expression. By controlling gene expression in a reversible yet potentially lasting manner in response to environmental stimulation, epigenetic mechanisms represent prime candidates in explaining both individual differences in aggression and the development of elevated aggressive behaviors following life adversity. In this manuscript, we review the evidence for an epigenetic basis in the development and expression of aggression in both humans and related preclinical animal models. In particular, we discuss reports linking DNA methylation, histone post-translational modifications, as well as non-coding RNA, to the regulation of a variety of genes implicated in the neurobiology of aggression including neuropeptides, the serotoninergic and dopaminergic systems, and stress response related systems. While clinical reports do reveal interesting patterns of DNA methylation underlying individual differences and experience-induced aggressive behaviors, they do, in general, face the challenge of linking peripheral observations to central nervous system regulations. Preclinical studies, on the other hand, provide detailed mechanistic insights into the epigenetic reprogramming of gene expression following life adversities. Although the functional link to aggression remains unclear in most, these studies together do highlight the involvement of epigenetic events driven by DNA methylation, histone modifications, and non-coding RNA in the neuroadaptations underlying the development and expression of aggression.
Collapse
Affiliation(s)
- Florian Duclot
- Department of Biomedical Sciences and Program in Neuroscience, Florida State University, Tallahassee, FL, USA.
| | - Mohamed Kabbaj
- Department of Biomedical Sciences and Program in Neuroscience, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
30
|
Malanchini M, Engelhardt LE, Raffington LA, Sabhlok A, Grotzinger AD, Briley DA, Madole JW, Freis SM, Patterson MW, Harden KP, Tucker-Drob EM. Weak and uneven associations of home, neighborhood, and school environments with stress hormone output across multiple timescales. Mol Psychiatry 2021; 26:4823-4838. [PMID: 32366955 PMCID: PMC9030635 DOI: 10.1038/s41380-020-0747-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 01/15/2023]
Abstract
The progression of lifelong trajectories of socioeconomic inequalities in health and mortality begins in childhood. Dysregulation in cortisol, a stress hormone that is the primary output of the hypothalamus-pituitary-adrenal (HPA) axis, has been hypothesized to be a mechanism for how early environmental adversity compromises health. However, despite the popularity of cortisol as a biomarker for stress and adversity, little is known about whether cortisol output differs in children being raised in socioeconomically disadvantaged environments. Here, we show that there are few differences between advantaged and disadvantaged children in their cortisol output. In 8-14-year-old children from the population-based Texas Twin Project, we measured cortisol output at three different timescales: (a) diurnal fluctuation in salivary cortisol (n = 400), (b) salivary cortisol reactivity and recovery after exposure to the Trier Social Stress Test (n = 444), and (c) cortisol concentration in hair (n = 1210). These measures converged on two moderately correlated, yet distinguishable, dimensions of HPA function. We tested differences in cortisol output across nine aspects of social disadvantage at the home (e.g., family socioeconomic status), school (e.g., average levels of academic achievement), and neighborhood (e.g., concentrated poverty). Children living in neighborhoods with higher concentrated poverty had higher diurnal cortisol output, as measured in saliva; otherwise, child cortisol output was unrelated to any other aspect of social disadvantage. Overall, we find limited support for alteration in HPA axis functioning as a general mechanism for the health consequences of socioeconomic inequality in childhood.
Collapse
Affiliation(s)
- Margherita Malanchini
- Department of Biological and Experimental Psychology, Queen Mary University of London, London, UK.
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA.
- Population research Center, The University of Texas at Austin, Austin, TX, USA.
| | - Laura E Engelhardt
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - Laurel A Raffington
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
- Population research Center, The University of Texas at Austin, Austin, TX, USA
| | - Aditi Sabhlok
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - Andrew D Grotzinger
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - Daniel A Briley
- Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - James W Madole
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - Samantha M Freis
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - Megan W Patterson
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - K Paige Harden
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
- Population research Center, The University of Texas at Austin, Austin, TX, USA
| | - Elliot M Tucker-Drob
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA.
- Population research Center, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
31
|
Jaimes-Hoy L, Pérez-Maldonado A, Narváez Bahena E, de la Cruz Guarneros N, Rodríguez-Rodríguez A, Charli JL, Soberón X, Joseph-Bravo P. Sex Dimorphic Changes in Trh Gene Methylation and Thyroid-Axis Response to Energy Demands in Maternally Separated Rats. Endocrinology 2021; 162:bqab110. [PMID: 34043769 DOI: 10.1210/endocr/bqab110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 12/18/2022]
Abstract
The hypothalamus-pituitary-thyroid (HPT) axis regulates energy balance through the pleiotropic action of thyroid hormones. HPT basal activity and stimulation by cold or voluntary exercise are repressed by previous chronic stress in adults. Maternal separation (MS) modifies HPT basal activity; we thus studied the response of the axis to energy demands and analyzed possible epigenetic changes on Trh promoter. Nonhandled (NH) or MS male Wistar rats were cold exposed 1 h at adulthood; Trh expression in the hypothalamic paraventricular nucleus (PVN) and serum thyrotropin (TSH) concentration were increased only in NH rats. Two weeks of voluntary exercise decreased fat mass and increased Trh expression, and thyroid hormones concentration changed proportionally to running distance in NH male rats and MS male rats. Although NH females ran more than MS and much more than males, exercise decreased body weight and fat mass only in NH rats with no change on any parameter of the HPT axis but increased Pomc expression in arcuate-nucleus of NH and Npy in MS females. Overall, the methylation pattern of PVN Trh gene promoter was similar in NH males and females; MS modified methylation of specific CpG sites, a thyroid hormone receptor (THR)-binding site present after the initiation site was hypomethylated in MS males; in MS females, the THR binding site of the proximal promoter (site 4) and 2 sites in the first intron were hypermethylated. Our studies showed that, in a sex-dimorphic manner, MS blunted the responses of HPT axis to energy demands in adult animals and caused methylation changes on Trh promoter that could alter T3 feedback.
Collapse
Affiliation(s)
- Lorraine Jaimes-Hoy
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Adrián Pérez-Maldonado
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Elian Narváez Bahena
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Natalia de la Cruz Guarneros
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Adair Rodríguez-Rodríguez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Jean-Louis Charli
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Xavier Soberón
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
- Instituto Nacional de Medicina Genómica, Ciudad de México, México
| | - Patricia Joseph-Bravo
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| |
Collapse
|
32
|
Dick A, Chen A. The role of TET proteins in stress-induced neuroepigenetic and behavioural adaptations. Neurobiol Stress 2021; 15:100352. [PMID: 34189192 PMCID: PMC8220100 DOI: 10.1016/j.ynstr.2021.100352] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 05/21/2021] [Accepted: 06/05/2021] [Indexed: 12/27/2022] Open
Abstract
Over the past decade, critical, non-redundant roles of the ten-eleven translocation (TET) family of dioxygenase enzymes have been identified in the brain during developmental and postnatal stages. Specifically, TET-mediated active demethylation, involving the iterative oxidation of 5-methylcytosine to 5-hydroxymethylcytosine and subsequent oxidative derivatives, is dynamically regulated in response to environmental stimuli such as neuronal activity, learning and memory processes, and stressor exposure. Such changes may therefore perpetuate stable and dynamic transcriptional patterns within neuronal populations required for neuroplasticity and behavioural adaptation. In this review, we will highlight recent evidence supporting a role of TET protein function and active demethylation in stress-induced neuroepigenetic and behavioural adaptations. We further explore potential mechanisms by which TET proteins may mediate both the basal and pathological embedding of stressful life experiences within the brain of relevance to stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Alec Dick
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- Corresponding author.
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- The Ruhman Family Laboratory for Research on the Neurobiology of Stress, Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
33
|
Lauby SC, Fleming AS, McGowan PO. Beyond maternal care: The effects of extra-maternal influences within the maternal environment on offspring neurodevelopment and later-life behavior. Neurosci Biobehav Rev 2021; 127:492-501. [PMID: 33905789 DOI: 10.1016/j.neubiorev.2021.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/08/2021] [Accepted: 04/17/2021] [Indexed: 01/26/2023]
Abstract
The early-life maternal environment has a profound and persistent effect on offspring neuroendocrine function, neurotransmitter systems, and behavior. Studies using rodent models suggest that early-life maternal care can influence the 'developmental programming' of offspring in part through altered epigenetic regulation of specific genes. The exploration of epigenetic regulation of these genes as a biological mechanism has been important to our understanding of how animals adapt to their environments and how these developmental trajectories may be altered. However, other non-maternal factors have been shown to act directly, or to interact with maternal care, to influence later-life phenotype. Based on accumulating evidence, including our research, we discuss other important influences on the developmental programming of offspring. We highlight early-life variations in temperature exposure and offspring genotype x environment interactions as prominent examples. We conclude with recommendations for future investigations on how early-life maternal care and extra-maternal influences lead to persistent changes in the brain and behavior of the offspring throughout development.
Collapse
Affiliation(s)
- Samantha C Lauby
- Department of Biological Sciences, University of Toronto Scarborough Campus, Scarborough, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Alison S Fleming
- Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada.
| | - Patrick O McGowan
- Department of Biological Sciences, University of Toronto Scarborough Campus, Scarborough, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
34
|
Stettler PR, F Antunes D, Taborsky B. The serotonin 1A receptor modulates the social behaviour within groups of a cooperatively-breeding cichlid. Horm Behav 2021; 129:104918. [PMID: 33428923 DOI: 10.1016/j.yhbeh.2020.104918] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/06/2020] [Accepted: 12/23/2020] [Indexed: 11/24/2022]
Abstract
The neurotransmitter serotonin (5-HT) reduces aggressive behaviour in a number of vertebrates, and the 5-HT1A receptor is known to be involved in this regulation. However, the role of this receptor in the modulation of sociopositive behaviour remains largely unknown. Here we investigated the role of the 5-HT1A receptor in the regulation of aggressive, submissive and affiliative behaviour in the cooperatively-breeding cichlid Neolamprologus pulcher. In two experiments, we performed intramuscular injections of a 5-HT1A agonist (8-OH-DPAT) and antagonist (Way-100635) followed by recordings of social behaviour of injected fish within their social groups. We determined the concentrations and post-injection times when the drugs had the greatest effect on social behaviour. We recorded spontaneous social behaviour in both experiments. In the second experiment we also recorded behaviour after social groups received a territorial challenge by live presentations of either conspecifics or egg predators. The 5-HT1A agonist caused an increase in aggression and a decrease in submission and affiliation, whereas the antagonist had the opposite effects. Thus, the 5-HT1A receptor plays an important regulatory role not only for aggressive but also sociopositive behaviour.
Collapse
Affiliation(s)
- Pia R Stettler
- Division of Behavioural Ecology, Institute of Ecology and Evolution, University of Bern, Wohlenstrasse 50A, 3032 Hinterkappelen, Switzerland.
| | - Diogo F Antunes
- Division of Behavioural Ecology, Institute of Ecology and Evolution, University of Bern, Wohlenstrasse 50A, 3032 Hinterkappelen, Switzerland.
| | - Barbara Taborsky
- Division of Behavioural Ecology, Institute of Ecology and Evolution, University of Bern, Wohlenstrasse 50A, 3032 Hinterkappelen, Switzerland.
| |
Collapse
|
35
|
Tiwari P, Fanibunda SE, Kapri D, Vasaya S, Pati S, Vaidya VA. GPCR signaling: role in mediating the effects of early adversity in psychiatric disorders. FEBS J 2021; 288:2602-2621. [DOI: 10.1111/febs.15738] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/11/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Praachi Tiwari
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Sashaina E. Fanibunda
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
- Medical Research Centre Kasturba Health Society Mumbai India
| | - Darshana Kapri
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Shweta Vasaya
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Sthitapranjya Pati
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| | - Vidita A. Vaidya
- Department of Biological Sciences Tata Institute of Fundamental Research Mumbai India
| |
Collapse
|
36
|
Roy B, Dwivedi Y. Modeling endophenotypes of suicidal behavior in animals. Neurosci Biobehav Rev 2021; 128:819-827. [PMID: 33421543 DOI: 10.1016/j.neubiorev.2020.12.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/25/2020] [Accepted: 12/31/2020] [Indexed: 12/26/2022]
Abstract
Suicide is a major public health concern. One of the common contributors to the increased risk for suicide is the genetic constitution of individuals, which determines certain endophenotypic traits used as quantifiable measure of neurobiological functions. Therefore, a logical deconstruction of the originating endophenotypes associated with suicidal risk could provide a better understanding of this complex disorder. In this regard, non-human animals can be a useful resource to test endophenotypes of suicidal behavior and the neurobiology underlying these endophenotypes. In this review, we have focused on the neurobiological abnormalities, primarily genetic and epigenetic abnormalities, associated with suicidal behavior and the scope of their modeling in animals. This can substantially advance the current understanding of suicidal behavior manifested with certain trait-based endophenotypes and may provide an opportunity to test novel hypotheses as well as aid in the development of treatment opportunities and risk assessment.
Collapse
Affiliation(s)
- Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, 1720 7(th) Avenue South, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, 1720 7(th) Avenue South, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
37
|
Aleixo JF, Pereira MRF, Montagnini BG, Pereira MJD, Forcato S, Moreira EG, Ceravolo GS, Vieira ML, Kiss ACI, Gerardin DCC. Effect of paracetamol treatment on maternal care and reproductive outcomes in female rat offspring. Reprod Fertil Dev 2020; 32:1311-1325. [PMID: 33308393 DOI: 10.1071/rd20007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 11/06/2020] [Indexed: 12/25/2022] Open
Abstract
Paracetamol (PAR) is one of the most commonly used drugs by pregnant women because it is considered safe for the mother and fetus. However, PAR is transferred into breast milk and crosses the blood-placental barrier, being present in the progeny during important stages of development. Intrauterine exposure to PAR may decrease the anogenital distance and follicle reserve in female rodent offspring. Therefore, the aim of the present study was to evaluate whether maternal PAR treatment altered the reproductive behaviour of dams and the sexual development of female rat offspring. Pregnant Wistar rats were gavaged daily with 350mg kg-1 day-1 PAR or water during gestation (from Gestation Day (GD) 6 until delivery) or during gestation and lactation (from GD6 until weaning). Maternal PAR treatment had maternal effects (increased grooming behaviour), and resulted in impaired sexual behaviour, decreased follicle reserve and increased plasma oestradiol concentrations in female offspring.
Collapse
Affiliation(s)
- Jeberson F Aleixo
- Department of Physiological Sciences. State University of Londrina, Rodovia Celso Garcia Cid, PR 445Km 380, 86051-980, Londrina, Paraná, Brazil
| | - Marina R F Pereira
- Department of Physiological Sciences. State University of Londrina, Rodovia Celso Garcia Cid, PR 445Km 380, 86051-980, Londrina, Paraná, Brazil
| | - Bruno G Montagnini
- Department of Physiological Sciences. State University of Londrina, Rodovia Celso Garcia Cid, PR 445Km 380, 86051-980, Londrina, Paraná, Brazil
| | - Matheus Junior D Pereira
- Department of Physiological Sciences. State University of Londrina, Rodovia Celso Garcia Cid, PR 445Km 380, 86051-980, Londrina, Paraná, Brazil
| | - Simone Forcato
- Department of Physiological Sciences. State University of Londrina, Rodovia Celso Garcia Cid, PR 445Km 380, 86051-980, Londrina, Paraná, Brazil
| | - Estefânia G Moreira
- Department of Physiological Sciences. State University of Londrina, Rodovia Celso Garcia Cid, PR 445Km 380, 86051-980, Londrina, Paraná, Brazil
| | - Graziela S Ceravolo
- Department of Physiological Sciences. State University of Londrina, Rodovia Celso Garcia Cid, PR 445Km 380, 86051-980, Londrina, Paraná, Brazil
| | - Milene L Vieira
- University Center Philadelphia, Alagoas Street, 2050, 86010-520, Londrina, Paraná, Brazil
| | - Ana C I Kiss
- Department of Physiology, Botucatu Biosciences Institute, São Paulo State University, Distrito de Rubião Júnior s/n, 18618-000, Botucatu, São Paulo, Brazil
| | - Daniela C C Gerardin
- Department of Physiological Sciences. State University of Londrina, Rodovia Celso Garcia Cid, PR 445Km 380, 86051-980, Londrina, Paraná, Brazil; and Corresponding author.
| |
Collapse
|
38
|
Lauby SC, McGowan PO. Early life variations in temperature exposure affect the epigenetic regulation of the paraventricular nucleus in female rat pups. Proc Biol Sci 2020; 287:20201991. [PMID: 33109014 PMCID: PMC7661289 DOI: 10.1098/rspb.2020.1991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/06/2020] [Indexed: 11/12/2022] Open
Abstract
Early life maternal care received has a profound effect on later-life behaviour in adult offspring, and previous studies have suggested epigenetic mechanisms are involved. Changes in thyroid hormone receptor signalling may be related to differences in maternal care received and DNA methylation modifications. We investigated the effects of variations in temperature exposure (a proxy of maternal contact) and licking-like tactile stimulation on these processes in week-old female rat pups. We assessed thyroid hormone receptor signalling by measuring circulating triiodothyronine and transcript abundance of thyroid hormone receptors and the thyroid hormone-responsive genes DNA methyltransferase 3a and oxytocin in the paraventricular nucleus of the hypothalamus. DNA methylation of the oxytocin promoter was assessed in relation to changes in thyroid hormone receptor binding. Repeated room temperature exposure was associated with a decrease in thyroid hormone receptor signalling measures relative to nest temperature exposure, while acute room temperature exposure was associated with an increase. Repeated room temperature exposure also increased thyroid hormone receptor binding and DNA methylation at the oxytocin promoter. These findings suggest that repeated room temperature exposure may affect DNA methylation levels as a consequence of alterations in thyroid hormone receptor signalling.
Collapse
Affiliation(s)
- Samantha C. Lauby
- Department of Biological Sciences, University of Toronto, Scarborough Campus, SW548, 1265 Military Trail, Scarborough, Toronto, Ontario, Canada M1C 1A4
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Patrick O. McGowan
- Department of Biological Sciences, University of Toronto, Scarborough Campus, SW548, 1265 Military Trail, Scarborough, Toronto, Ontario, Canada M1C 1A4
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
39
|
Early-life short-term environmental enrichment counteracts the effects of stress on anxiety-like behavior, brain-derived neurotrophic factor and nuclear translocation of glucocorticoid receptors in the basolateral amygdala. Sci Rep 2020; 10:14053. [PMID: 32820184 PMCID: PMC7441150 DOI: 10.1038/s41598-020-70875-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/30/2020] [Indexed: 01/16/2023] Open
Abstract
Early life is a decisive stage for the development of physiological and psychological characteristics of an individual. Any stress or disruption of healthy development at this stage has serious long-lasting consequences for the remaining life. Unfortunately, early life stress is a common occurrence in humans and other animals. In this context, we investigated if the provision of environmental enrichment during the pre-weaning phase of rat pups and dams could alter the consequences of early-life maternal-separation stress. Pre-weaning enrichment rescued the effects of maternal separation on the excess secretion of adrenal stress hormones and anxiety-like behavior during adulthood. Enrichment also reduced the effect of stress on the spine density of basolateral amygdala neurons, a brain region critical for stress-induced facilitation of emotional behaviors. Pre-weaning enrichment, provided during early-life, blunted the effects of maternal separation stress on decreased intra-nuclear translocation of glucocorticoid receptors within the amygdala neurons when tested later in adulthood. Early-life, pre-weaning environmental enrichment also increased the amount of brain-derived neurotrophic factor within adult basolateral amygdala. Our observations showed that environmental manipulation during early formative years could be utilized to build lifelong resilience to stress. Complex naturalistic housing and sensory enrichment is, thus, an useful buffer against an impoverished and stressful childhood.
Collapse
|
40
|
Montirosso R, Rosa E, Giorda R, Fazzi E, Orcesi S, Cavallini A, Provenzi L. Early Parenting Intervention - Biobehavioral Outcomes in infants with Neurodevelopmental Disabilities (EPI-BOND): study protocol for an Italian multicentre randomised controlled trial. BMJ Open 2020; 10:e035249. [PMID: 32699128 PMCID: PMC7375429 DOI: 10.1136/bmjopen-2019-035249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Neurodevelopmental disability (ND) represents an adverse condition for infants' socio-emotional and behavioural development as well as for caregiving (eg, parental sensitivity) and mother-infant interaction. Adverse exposures are associated with altered neuroendocrine hormones concentrations (eg, oxytocin and cortisol) and epigenetic regulation (eg, methylation of stress-related genes), which in turn may contribute to less-than-optimal mother-infant interaction. Parental sensitivity is a protective factor for childrens' development and early parental interventions (eg, video-feedback intervention) can promote parental caregiving and better developmental outcomes in children. The present multi-centric and longitudinal randomised controlled trial aims to assess if and to which extent early VFI could benefit both infants and mothers in terms of behavioural outcomes as well as neuroendocrine and epigenetic regulation. METHODS AND ANALYSIS Dyads will be randomly assigned to the video-feedback Intervention Group or Control Group ('dummy' intervention: telephone calls). Infants with ND aged 3 to 18 months will be recruited from three major child neuropsychiatric units in northern Italy. A multi-layer approach to intervention effects will include videotapes of mother-infant interaction, maternal reports as well as saliva samples for hormones concentrations and target-gene methylation analysis (eg, BDNF, NR3C1, OXTR and SCL6A4) that will be obtained at each of the four assessment sessions: T0, baseline; T1, post-intervention; T2, short-term follow-up (3 month); T3, long-term follow-up (6 month). Primary effectiveness measures will be infant socio-emotional behaviour and maternal sensitivity. Neuroendocrine hormones concentrations and DNA methylation status of target genes will be secondary outcomes. Feasibility, moderation and confounding variables will be measured and controlled between the two groups. ETHICS AND DISSEMINATION Ethics approval has been obtained in all three participating units. Results of the main trial and each of the secondary endpoints will be submitted for publication in peer-reviewed journals and international conferences. TRIAL REGISTRATION NUMBER NCT03853564; Pre-results.
Collapse
Affiliation(s)
- Rosario Montirosso
- 0-3 Center for the at-Risk Infant, Scientific Institute IRCCS E. Medea, Bosisio Parini, Lecco, Italy
| | - Elisa Rosa
- 0-3 Center for the at-Risk Infant, Scientific Institute IRCCS E. Medea, Bosisio Parini, Lecco, Italy
| | - Roberto Giorda
- Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Lecco, Italy
| | - Elisa Fazzi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Unit of Child and Adolescence Neuropsychiatry, ASST Spedali Civili, Brescia, Italy
| | - Simona Orcesi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Anna Cavallini
- Neuropsychiatry and Neurorehabilitation Unit, Scientific Institute IRCCS E. Medea, Bosisio Parini, Lecco, Italy
| | - Livio Provenzi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
41
|
Ilchibaeva TV, Tsybko AS, Kondaurova EM, Kovetskaya AI, Kozhemyakina RV, Naumenko VS. Expression Patterns of Serotonin Receptors 1А and 7 in the Brain of Rats with Genetically Determined Fear-Induced Aggressive Behavior or the Lack of Aggression. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420020051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Abstract
Epigenetic mechanisms govern the transcription of the genome. Research with model systems reveals that environmental conditions can directly influence epigenetic mechanisms that are associated with interindividual differences in gene expression in brain and neural function. In this review, we provide a brief overview of epigenetic mechanisms and research with relevant rodent models. We emphasize more recent translational research programs in epigenetics as well as the challenges inherent in the integration of epigenetics into developmental and clinical psychology. Our objectives are to present an update with respect to the translational relevance of epigenetics for the study of psychopathology and to consider the state of current research with respect to its potential importance for clinical research and practice in mental health.
Collapse
Affiliation(s)
- Kieran J O'Donnell
- Department of Psychiatry and Sackler Program for Epigenetics and Psychobiology, McGill University, Montreal, Quebec H4H 1R3, Canada; .,Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec H3H 1R4, Canada.,Child and Brain Development Program, CIFAR, Toronto, Ontario M5G 1M1, Canada
| | - Michael J Meaney
- Department of Psychiatry and Sackler Program for Epigenetics and Psychobiology, McGill University, Montreal, Quebec H4H 1R3, Canada; .,Child and Brain Development Program, CIFAR, Toronto, Ontario M5G 1M1, Canada.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), 117609 Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore
| |
Collapse
|
43
|
Environment and early life: Decisive factors for stress-resilience and vulnerability. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 150:155-185. [DOI: 10.1016/bs.irn.2019.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Fu Y, Depue RA. A novel neurobehavioral framework of the effects of positive early postnatal experience on incentive and consummatory reward sensitivity. Neurosci Biobehav Rev 2019; 107:615-640. [DOI: 10.1016/j.neubiorev.2019.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 09/08/2019] [Accepted: 09/17/2019] [Indexed: 12/22/2022]
|
45
|
Provenzi L, Brambilla M, Scotto di Minico G, Montirosso R, Borgatti R. Maternal caregiving and DNA methylation in human infants and children: Systematic review. GENES BRAIN AND BEHAVIOR 2019; 19:e12616. [DOI: 10.1111/gbb.12616] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/18/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Livio Provenzi
- Scientific Institute IRCCS E. Medea0‐3 Centre for the at‐Risk Infant Bosisio Parini Lecco Italy
| | - Maddalena Brambilla
- Scientific Institute IRCCS E. Medea0‐3 Centre for the at‐Risk Infant Bosisio Parini Lecco Italy
| | - Giunia Scotto di Minico
- Scientific Institute IRCCS E. Medea0‐3 Centre for the at‐Risk Infant Bosisio Parini Lecco Italy
| | - Rosario Montirosso
- Scientific Institute IRCCS E. Medea0‐3 Centre for the at‐Risk Infant Bosisio Parini Lecco Italy
| | - Renato Borgatti
- Scientific Institute IRCCS E. MedeaChild Neuropsychiatry and Neurorehabilitation Unit Bosisio Parini Lecco Italy
| |
Collapse
|
46
|
Safi-Stibler S, Gabory A. Epigenetics and the Developmental Origins of Health and Disease: Parental environment signalling to the epigenome, critical time windows and sculpting the adult phenotype. Semin Cell Dev Biol 2019; 97:172-180. [PMID: 31587964 DOI: 10.1016/j.semcdb.2019.09.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 09/19/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023]
Abstract
The literature about Developmental Origins of Health and Disease (DOHaD) studies is considerably growing. Maternal and paternal environment, during all the development of the individual from gametogenesis to weaning and beyond, as well as the psychosocial environment in childhood and teenage, can shape the adult and the elderly person's susceptibility to her/his own environment and diseases. This non-conventional, non-genetic, inheritance is underlain by several mechanisms among which epigenetics is obviously central, due to the notion of memory of early decisional events during development even when this stimulus is gone, that is implied in Waddington's developmental concept. This review first summarizes the different mechanisms by which the environment can model the epigenome: receptor signalling, energy metabolism and signal mechanotransduction from extracellular matrix to chromatin. Then an overview of the epigenetic changes in response to maternal environment during the vulnerability time windows, gametogenesis, early development, placentation and foetal growth, and postnatal period, is described, with the specific example of overnutrition and food deprivation. The implication of epigenetics in DOHaD is obvious, however the precise causal chain from early environment to the epigenome modifications to the phenotype still needs to be deciphered.
Collapse
Affiliation(s)
- Sofiane Safi-Stibler
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350, Jouy-en-Josas, France; Sorbonne Université, Collège Doctoral, F-75005, Paris, France
| | - Anne Gabory
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350, Jouy-en-Josas, France.
| |
Collapse
|
47
|
Lauby SC, Chatterjee D, Pan P, McGowan PO, Fleming AS. Inter-individual maternal care received and genotype interactions affect dopaminergic phenotypes in female rat offspring. J Neuroendocrinol 2019; 31:e12706. [PMID: 30860615 DOI: 10.1111/jne.12706] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/07/2019] [Accepted: 03/07/2019] [Indexed: 12/18/2022]
Abstract
Rat mothers exhibit natural variations in care and can shape offspring adult behaviour and their maternal care by affecting the dopaminergic system. We explored whether genotype and gene × environment interactions are involved in these processes in nulliparous female offspring. We assessed maternal licking/grooming toward individual female pups during the first week postpartum and dopamine-related behaviour of the offspring in adulthood. Behaviours explored included strategy shifting, impulsive action and sucrose preference. Single nucleotide polymorphisms in the dopamine receptor 2, dopamine transporter and catechol-O-methyltransferase genes were examined in relation to offspring behaviour and baseline dopamine turnover in select brain regions. Dopamine receptor 2 (RS107017253) variation moderated, or interacted with, the relationship between early-life licking received and behaviour. Specifically, offspring with the A/A genotype showed a significant correlation between early-life licking received and behaviour. Offspring with the A/G and G/G genotypes did not show this relationship. Dopamine transporter gene variation affected offspring behaviour regardless of early-life licking received. Our findings suggest that genotype can directly affect dopamine-related behaviours and alter the sensitivity of offspring to the maternal environment. This could be informative on how maternal care is transmitted between generations of female offspring.
Collapse
Affiliation(s)
- Samantha C Lauby
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Diptendu Chatterjee
- The Peter Gilgan Centre for Research and Learning, Sickkids Hospital, Toronto, Ontario, Canada
| | - Pauline Pan
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Patrick O McGowan
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Alison S Fleming
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
48
|
Timothy A, Benegal V, Shankarappa B, Saxena S, Jain S, Purushottam M. Influence of early adversity on cortisol reactivity, SLC6A4 methylation and externalizing behavior in children of alcoholics. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109649. [PMID: 31082414 DOI: 10.1016/j.pnpbp.2019.109649] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Children of parents with alcoholism face considerable stress, and often have externalizing behaviors. Early adversity is known to affect DNA methylation and the functioning of the HPA axis. We investigated the association of early adversity with cortisol reactivity, 5HTTLPR genotype, site specific DNA methylation in the SLC6A4 gene and externalizing behavior in children of alcoholics (COA), and a matched sample of control children. METHODS We examined children of alcoholics (N = 50) and age matched control children (N = 50) for exposure to early adversity (both prenatal and postnatal), assessed their salivary cortisol reactivity and evaluated their levels of emotional and behavioral difficulty in terms of externalizing and internalizing behavior. Site-specific DNA methylation at a previously characterized SLC6A4 region was determined in salivary DNA using pyrosequencing. The 5HTTLPR region of the SLC6A4 gene was also genotyped. RESULTS COA had significantly higher experience of early adversity than control children. Cortisol reactivity was reduced in COA, and negatively correlated with early adversity. Both early adversity and cortisol reactivity correlated with externalizing behavior. SLC6A4 methylation was higher in COA, and correlated with early adversity. SLC6A4 genotype did not show association with any of the variables. CONCLUSION Our study provides further evidence that early adversity is associated with blunted cortisol reactivity, increased site-specific CpG DNA methylation at the SLC6A4 gene, and high externalizing behavior.
Collapse
Affiliation(s)
- Anurag Timothy
- Centre for Addiction Medicine, National Institute of Mental Health and Neurosciences, Bangalore 560029, India; Department of Psychiatry, Base Hospital Delhi Cantt, New Delhi 110010, India
| | - Vivek Benegal
- Centre for Addiction Medicine, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Bhagyalakshmi Shankarappa
- Molecular Genetics Lab, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore 560029, India; St Johns Medical College Hospital, Bangalore, India
| | - Sachin Saxena
- Department of Psychiatry, Base Hospital Delhi Cantt, New Delhi 110010, India
| | - Sanjeev Jain
- Molecular Genetics Lab, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Meera Purushottam
- Molecular Genetics Lab, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| |
Collapse
|
49
|
Bagamasbad PD, Espina JEC, Knoedler JR, Subramani A, Harden AJ, Denver RJ. Coordinated transcriptional regulation by thyroid hormone and glucocorticoid interaction in adult mouse hippocampus-derived neuronal cells. PLoS One 2019; 14:e0220378. [PMID: 31348800 PMCID: PMC6660079 DOI: 10.1371/journal.pone.0220378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/15/2019] [Indexed: 12/04/2022] Open
Abstract
The hippocampus is a well-known target of thyroid hormone (TH; e.g., 3,5,3'-triiodothyronine-T3) and glucocorticoid (GC; e.g., corticosterone-CORT) action. Despite evidence that TH and GC play critical roles in neural development and function, few studies have identified genes and patterns of gene regulation influenced by the interaction of these hormones at a genome-wide scale. In this study we investigated gene regulation by T3, CORT, and T3 + CORT in the mouse hippocampus-derived cell line HT-22. We treated cells with T3, CORT, or T3 + CORT for 4 hr before cell harvest and RNA isolation for microarray analysis. We identified 9 genes regulated by T3, 432 genes by CORT, and 412 genes by T3 + CORT. Among the 432 CORT-regulated genes, there were 203 genes that exhibited an altered CORT response in the presence of T3, suggesting that T3 plays a significant role in modulating CORT-regulated genes. We also found 80 genes synergistically induced, and 73 genes synergistically repressed by T3 + CORT treatment. We performed in silico analysis using publicly available mouse neuronal chromatin immunoprecipitation-sequencing datasets and identified a considerable number of synergistically regulated genes with TH receptor and GC receptor peaks mapping within 1 kb of chromatin marks indicative of hormone-responsive enhancer regions. Functional annotation clustering of synergistically regulated genes reveal the relevance of proteasomal-dependent degradation, neuroprotective effect of growth hormones, and neuroinflammatory responses as key pathways to how TH and GC may coordinately influence learning and memory. Taken together, our transcriptome data represents a promising exploratory dataset for further study of common molecular mechanisms behind synergistic TH and GC gene regulation, and identify specific genes and their role in processes mediated by cross-talk between the thyroid and stress axes in a mammalian hippocampal model system.
Collapse
Affiliation(s)
- Pia D. Bagamasbad
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, United States of America
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines
| | - Jose Ezekiel C. Espina
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines
| | - Joseph R. Knoedler
- Neuroscience Graduate Program, The University of Michigan, Ann Arbor, Michigan, United States of America
| | - Arasakumar Subramani
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ariel J. Harden
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, United States of America
| | - Robert J. Denver
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
50
|
Zhang TY, Shahrokh D, Hellstrom IC, Wen X, Diorio J, Breuillaud L, Caldji C, Meaney MJ. Brain-Derived Neurotrophic Factor in the Nucleus Accumbens Mediates Individual Differences in Behavioral Responses to a Natural, Social Reward. Mol Neurobiol 2019; 57:290-301. [PMID: 31327126 DOI: 10.1007/s12035-019-01699-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/09/2019] [Indexed: 01/17/2023]
Abstract
BDNF-oxytocin interactions in the brain are implicated in mammalian maternal behavior. We found that BDNF gene expression is increased in the hippocampus of rat mothers that show increased pup licking/grooming (high LG mothers) compared to low LG mothers. High LG mothers also showed increased BDNF protein levels in the nucleus accumbens (nAcc). Immunoneutralization of BDNF in the nAcc eliminated the differences in pup LG between high and low LG mothers. Oxytocin antagonist in the ventral hippocampus significantly decreased the frequency of maternal LG behavior. Oxytocin antagonist significantly prevented the oxytocin-induced BDNF gene expression in primary hippocampal cell cultures. We suggest that oxytocin-induced regulation of BDNF in the nAcc provides a neuroendocrine basis for both individual differences in maternal behavior and resilience to the stress of reproduction in female mammals.
Collapse
Affiliation(s)
- Tie-Yuan Zhang
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H1R3, Canada. .,Sackler Program for Epigenetics & Psychobiology at McGill University, Montreal, Quebec, H4H1R3, Canada. .,Ludmer Centre for Neuroinformatics and Mental health, McGill University, Montreal, Quebec, H4H1R3, Canada.
| | - Dara Shahrokh
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H1R3, Canada
| | - Ian C Hellstrom
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H1R3, Canada
| | - Xianglan Wen
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H1R3, Canada.,Sackler Program for Epigenetics & Psychobiology at McGill University, Montreal, Quebec, H4H1R3, Canada
| | - Josie Diorio
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H1R3, Canada.,Sackler Program for Epigenetics & Psychobiology at McGill University, Montreal, Quebec, H4H1R3, Canada
| | - Lionel Breuillaud
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H1R3, Canada
| | - Christian Caldji
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H1R3, Canada
| | - Michael J Meaney
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H1R3, Canada. .,Sackler Program for Epigenetics & Psychobiology at McGill University, Montreal, Quebec, H4H1R3, Canada. .,Ludmer Centre for Neuroinformatics and Mental health, McGill University, Montreal, Quebec, H4H1R3, Canada. .,Singapore Institute for Clinical Sciences, Singapore, 117609, Singapore.
| |
Collapse
|