1
|
Percelay S, Lahogue C, Billard JM, Freret T, Boulouard M, Bouet V. The 3-hit animal models of schizophrenia: Improving strategy to decipher and treat the disease? Neurosci Biobehav Rev 2024; 157:105526. [PMID: 38176632 DOI: 10.1016/j.neubiorev.2023.105526] [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: 10/12/2023] [Revised: 12/08/2023] [Accepted: 12/23/2023] [Indexed: 01/06/2024]
Abstract
Schizophrenia is a complex disease related to combination and interactions between genetic and environmental factors, with an epigenetic influence. After the development of the first mono-factorial animal models of schizophrenia (1-hit), that reproduced patterns of either positive, negative and/or cognitive symptoms, more complex models combining two factors (2-hit) have been developed to better fit with the multifactorial etiology of the disease. In the two past decades, a new way to design animal models of schizophrenia have emerged by adding a third hit (3-hit). This review aims to discuss the relevance of the risk factors chosen for the tuning of the 3-hit animal models, as well as the validities measurements and their contribution to schizophrenia understanding. We intended to establish a comprehensive overview to help in the choice of factors for the design of multiple-hit animal models of schizophrenia.
Collapse
Affiliation(s)
- Solenn Percelay
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France
| | - Caroline Lahogue
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France.
| | - Jean-Marie Billard
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France
| | - Thomas Freret
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France
| | - Michel Boulouard
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France
| | - Valentine Bouet
- Normandie Univ, UNICAEN, INSERM, CYCERON, CHU Caen, COMETE UMR 1075, 14000 Caen, France.
| |
Collapse
|
2
|
Sharma R, Kumarasamy M, Parihar VK, Ravichandiran V, Kumar N. Monoamine Oxidase: A Potential Link in Papez Circuit to Generalized Anxiety Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:638-655. [PMID: 37055898 DOI: 10.2174/1871527322666230412105711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/01/2023] [Accepted: 02/09/2023] [Indexed: 04/15/2023]
Abstract
Anxiety is a common mental illness that affects a large number of people around the world, and its treatment is often based on the use of pharmacological substances such as benzodiazepines, serotonin, and 5-hydroxytyrosine (MAO) neurotransmitters. MAO neurotransmitters levels are deciding factors in the biological effects. This review summarizes the current understanding of the MAO system and its role in the modulation of anxiety-related brain circuits and behavior. The MAO-A polymorphisms have been implicated in the susceptibility to generalized anxiety disorder (GAD) in several investigations. The 5-HT system is involved in a wide range of physiological and behavioral processes, involving anxiety, aggressiveness, stress reactions, and other elements of emotional intensity. Among these, 5-HT, NA, and DA are the traditional 5-HT neurons that govern a range of biological activities, including sleep, alertness, eating, thermoregulation, pains, emotion, and memory, as anticipated considering their broad projection distribution in distinct brain locations. The DNMTs (DNA methyltransferase) protein family, which increasingly leads a prominent role in epigenetics, is connected with lower transcriptional activity and activates DNA methylation. In this paper, we provide an overview of the current state of the art in the elucidation of the brain's complex functions in the regulation of anxiety.
Collapse
Affiliation(s)
- Ravikant Sharma
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali- 844102, Bihar, India
| | - Murali Kumarasamy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali- 844102, Bihar, India
| | - Vipan Kumar Parihar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali-844102, Bihar, India
| | - V Ravichandiran
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali- 844102, Bihar, India
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali-844102, Bihar, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali-844102, Bihar, India
| |
Collapse
|
3
|
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
|
4
|
Asraf K, Zaidan H, Natoor B, Gaisler-Salomon I. Synergistic, long-term effects of glutamate dehydrogenase 1 deficiency and mild stress on cognitive function and mPFC gene and miRNA expression. Transl Psychiatry 2023; 13:248. [PMID: 37419882 DOI: 10.1038/s41398-023-02534-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
Glutamate abnormalities in the medial prefrontal cortex (mPFC) are associated with cognitive deficits. We previously showed that homozygous deletion of CNS glutamate dehydrogenase 1 (Glud1), a metabolic enzyme critical for glutamate metabolism, leads to schizophrenia-like behavioral abnormalities and increased mPFC glutamate; mice heterozygous for CNS Glud1 deletion (C-Glud1+/- mice) showed no cognitive or molecular abnormalities. Here, we examined the protracted behavioral and molecular effects of mild injection stress on C-Glud1+/- mice. We found spatial and reversal learning deficits, as well as large-scale mPFC transcriptional changes in pathways associated with glutamate and GABA signaling, in stress-exposed C-Glud1+/- mice, but not in their stress-naïve or C-Glud1+/+ littermates. These effects were observed several weeks following stress exposure, and the expression levels of specific glutamatergic and GABAergic genes differentiated between high and low reversal learning performance. An increase in miR203-5p expression immediately following stress may provide a translational regulatory mechanism to account for the delayed effect of stress exposure on cognitive function. Our findings show that chronic glutamate abnormalities interact with acute stress to induce cognitive deficits, and resonate with gene x environment theories of schizophrenia. Stress-exposed C-Glud1+/- mice may model a schizophrenia high-risk population, which is uniquely sensitive to stress-related 'trigger' events.
Collapse
Affiliation(s)
- Kfir Asraf
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Hiba Zaidan
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Baylasan Natoor
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Inna Gaisler-Salomon
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel.
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
5
|
Matrisciano F. Epigenetic regulation of metabotropic glutamate 2/3 receptors: Potential role for ultra-resistant schizophrenia? Pharmacol Biochem Behav 2023:173589. [PMID: 37348609 DOI: 10.1016/j.pbb.2023.173589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
Schizophrenia is a severe and debilitating psychiatric disorder characterized by early cognitive deficits, emotional and behavioral abnormalities resulted by a dysfunctional gene x environment interaction. Genetic and epigenetic abnormalities in cortical parvalbumin-positive GABAergic interneurons lead to alterations in glutamate-mediated excitatory neurotransmission, synaptic plasticity, and neuronal development. Epigenetic alterations during pregnancy or early phases of postnatal life are associated with schizophrenia vulnerability as well as inflammatory processes which are at the basis of brain pathology. An epigenetic animal model of schizophrenia showed specific changes in promoter DNA methylation activity of genes related to schizophrenia such as reelin, BDNF and GAD67, and altered expression and function of mGlu2/3 receptors in the frontal cortex. Although antipsychotic medications represent the main treatment for schizophrenia and generally show an optimal efficacy profile for positive symptoms and relatively poor efficacy for negative or cognitive symptoms, a considerable percentage of individuals show poor response, do not achieve a complete remission, and approximately 30 % of patients show treatment-resistance. Here, we explore the potential role of epigenetic abnormalities linked to metabotropic glutamate 2/3 receptors changes in expression and function as key molecular factors underlying the difference in response to antipsychotics.
Collapse
Affiliation(s)
- Francesco Matrisciano
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago (UIC), Chicago, IL, USA.
| |
Collapse
|
6
|
van Oers K, van den Heuvel K, Sepers B. The Epigenetics of Animal Personality. Neurosci Biobehav Rev 2023; 150:105194. [PMID: 37094740 DOI: 10.1016/j.neubiorev.2023.105194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 04/26/2023]
Abstract
Animal personality, consistent individual differences in behaviour, is an important concept for understanding how individuals vary in how they cope with environmental challenges. In order to understand the evolutionary significance of animal personality, it is crucial to understand the underlying regulatory mechanisms. Epigenetic marks such as DNA methylation are hypothesised to play a major role in explaining variation in phenotypic changes in response to environmental alterations. Several characteristics of DNA methylation also align well with the concept of animal personality. In this review paper, we summarise the current literature on the role that molecular epigenetic mechanisms may have in explaining personality variation. We elaborate on the potential for epigenetic mechanisms to explain behavioural variation, behavioural development and temporal consistency in behaviour. We then suggest future routes for this emerging field and point to potential pitfalls that may be encountered. We conclude that a more inclusive approach is needed for studying the epigenetics of animal personality and that epigenetic mechanisms cannot be studied without considering the genetic background.
Collapse
Affiliation(s)
- Kees van Oers
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands; Behavioural Ecology Group, Wageningen University & Research (WUR), Wageningen, the Netherlands.
| | - Krista van den Heuvel
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands; Behavioural Ecology Group, Wageningen University & Research (WUR), Wageningen, the Netherlands
| | - Bernice Sepers
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands; Behavioural Ecology Group, Wageningen University & Research (WUR), Wageningen, the Netherlands
| |
Collapse
|
7
|
Epigenetics in fetal alcohol spectrum disorder. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:211-239. [PMID: 37019593 DOI: 10.1016/bs.pmbts.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
During pregnancy, alcohol abuse and its detrimental effects on developing offspring are major public health, economic and social challenges. The prominent characteristic attributes of alcohol (ethanol) abuse during pregnancy in humans are neurobehavioral impairments in offspring due to damage to the central nervous system (CNS), causing structural and behavioral impairments that are together named fetal alcohol spectrum disorder (FASD). Development-specific alcohol exposure paradigms were established to recapitulate the human FASD phenotypes and establish the underlying mechanisms. These animal studies have offered some critical molecular and cellular underpinnings likely to account for the neurobehavioral impairments associated with prenatal ethanol exposure. Although the pathogenesis of FASD remains unclear, emerging literature proposes that the various genomic and epigenetic components that cause the imbalance in gene expression can significantly contribute to the development of this disease. These studies acknowledged numerous immediate and enduring epigenetic modifications, such as methylation of DNA, post-translational modifications (PTMs) of histone proteins, and regulatory networks related to RNA, using many molecular approaches. Methylated DNA profiles, PTMs of histone proteins, and RNA-regulated expression of genes are essential for synaptic and cognitive behavior. Thus, offering a solution to many neuronal and behavioral impairments reported in FASD. In the current chapter, we review the recent advances in different epigenetic modifications that cause the pathogenesis of FASD. The information discussed can help better explain the pathogenesis of FASD and thereby might provide a basis for finding novel therapeutic targets and innovative treatment strategies.
Collapse
|
8
|
Paoli C, Misztak P, Mazzini G, Musazzi L. DNA Methylation in Depression and Depressive-Like Phenotype: Biomarker or Target of Pharmacological Intervention? Curr Neuropharmacol 2022; 20:2267-2291. [PMID: 35105292 PMCID: PMC9890294 DOI: 10.2174/1570159x20666220201084536] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/18/2022] [Accepted: 01/26/2022] [Indexed: 12/29/2022] Open
Abstract
Major depressive disorder (MDD) is a debilitating psychiatric disorder, the third leading global cause of disability. Regarding aetiopathogenetic mechanisms involved in the onset of depressive disorders, the interaction between genetic vulnerability traits and environmental factors is believed to play a major role. Although much is still to be elucidated about the mechanisms through which the environment can interact with genetic background shaping the disease risk, there is a general agreement about a key role of epigenetic marking. In this narrative review, we focused on the association between changes in DNA methylation patterns and MDD or depressive-like phenotype in animal models, as well as mechanisms of response to antidepressant drugs. We discussed studies presenting DNA methylation changes at specific genes of interest and profiling analyses in both patients and animal models of depression. Overall, we collected evidence showing that DNA methylation could not only be considered as a promising epigenetic biomarker of pathology but could also help in predicting antidepressant treatment efficacy. Finally, we discussed the hypothesis that specific changes in DNA methylation signature could play a role in aetiopathogenetic processes as well as in the induction of antidepressant effect.
Collapse
Affiliation(s)
- Caterina Paoli
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- School of Pharmacy, Pharmacy Unit, University of Camerino, 62032 Camerino, Italy
| | - Paulina Misztak
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Giulia Mazzini
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Laura Musazzi
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
9
|
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: 47] [Impact Index Per Article: 23.5] [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
|
10
|
Clinton SM, Unroe KA, Shupe EA, McCoy CR, Glover ME. Resilience to Stress: Lessons from Rodents about Nature versus Nurture. Neuroscientist 2022; 28:283-298. [PMID: 33567987 PMCID: PMC11092422 DOI: 10.1177/1073858421989357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Individual differences in human temperament influence how we respond to stress and can confer vulnerability (or resilience) to emotional disorders. For example, high levels of behavioral inhibition in children predict increased risk of mood and anxiety disorders in later life. The biological underpinnings of temperament are unknown, although improved understanding can offer insight into the pathogenesis of emotional disorders. Our laboratory has used a rat model of temperamental differences to study neurodevelopmental factors that lead to a highly inhibited, stress vulnerable phenotype. Selective breeding for high versus low behavioral response to novelty created two rat strains that exhibit dramatic behavior differences over multiple domains relevant to emotional disorders. Low novelty responder (bLR) rats exhibit high levels of behavioral inhibition, passive stress coping, anhedonia, decreased sociability and vulnerability to chronic stress compared to high novelty responders (bHRs). On the other hand, bHRs exhibit high levels of behavioral dis-inhibition, active coping, and aggression. This review article summarizes our work with the bHR/bLR model showing the developmental emergence of the bHR/bLR phenotypes, the role the environment plays in shaping it, and the involvement of epigenetic processes such as DNA methylation that mediate differences in emotionality and stress reactivity.
Collapse
Affiliation(s)
- Sarah M. Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Keaton A. Unroe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Elizabeth A. Shupe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Chelsea R. McCoy
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Matthew E. Glover
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| |
Collapse
|
11
|
Thumfart KM, Jawaid A, Bright K, Flachsmann M, Mansuy IM. Epigenetics of childhood trauma: Long term sequelae and potential for treatment. Neurosci Biobehav Rev 2021; 132:1049-1066. [PMID: 34742726 DOI: 10.1016/j.neubiorev.2021.10.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/17/2021] [Accepted: 10/29/2021] [Indexed: 12/17/2022]
Abstract
Childhood trauma (CT) can have persistent effects on the brain and is one of the major risk factors for neuropsychiatric diseases in adulthood. Recent advances in the field of epigenetics suggest that epigenetic factors such as DNA methylation and histone modifications, as well as regulatory processes involving non-coding RNA are associated with the long-term sequelae of CT. This narrative review summarizes current knowledge on the epigenetic basis of CT and describes studies in animal models and human subjects examining how the epigenome and transcriptome are modified by CT in the brain. It discusses psychological and pharmacological interventions that can counteract epigenetic changes induced by CT and the need to establish longitudinal assessment after CT for developing more effective diagnostics and treatment strategies based on epigenetic targets.
Collapse
Affiliation(s)
- Kristina M Thumfart
- Laboratory of Neuroepigenetics, Brain Research Institute, University of Zürich and Institute for Neuroscience of the Swiss Federal Institute of Technology, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - Ali Jawaid
- Laboratory of Neuroepigenetics, Brain Research Institute, University of Zürich and Institute for Neuroscience of the Swiss Federal Institute of Technology, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland; Laboratory for Translational Research in Neuropsychiatric Disorders (TREND), BRAINCITY: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Ludwika Pasteura 3, Warsaw, 02-093, Poland
| | - Kristina Bright
- Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Marc Flachsmann
- Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Isabelle M Mansuy
- Laboratory of Neuroepigenetics, Brain Research Institute, University of Zürich and Institute for Neuroscience of the Swiss Federal Institute of Technology, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.
| |
Collapse
|
12
|
Ibrahim P, Almeida D, Nagy C, Turecki G. Molecular impacts of childhood abuse on the human brain. Neurobiol Stress 2021; 15:100343. [PMID: 34141833 PMCID: PMC8187840 DOI: 10.1016/j.ynstr.2021.100343] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/24/2021] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
Childhood abuse (CA) is a prevalent global health concern, increasing the risk of negative mental health outcomes later in life. In the literature, CA is commonly defined as physical, sexual, and emotional abuse, as well as neglect. Several mental disorders have been associated with CA, including depression, bipolar disorder, schizophrenia, and post-traumatic stress disorder, along with an increased risk of suicide. It is thought that traumatic life events occurring during childhood and adolescence may have a significant impact on essential brain functions, which may persist throughout adulthood. The interaction between the brain and the external environment can be mediated by epigenetic alterations in gene expression, and there is a growing body of evidence to show that such changes occur as a function of CA. Disruptions in the HPA axis, myelination, plasticity, and signaling have been identified in individuals with a history of CA. Understanding the molecular impact of CA on the brain is essential for the development of treatment and prevention measures. In this review, we will summarize studies that highlight the molecular changes associated with CA in the human brain, along with supporting evidence from peripheral studies and animal models. We will also discuss some of the limitations surrounding the study of CA and propose extracellular vesicles as a promising future approach in the field.
Collapse
Affiliation(s)
- Pascal Ibrahim
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Daniel Almeida
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
13
|
Joaquim RM, Guatimosim RF, Araújo RJDS, Nardi AE, Veras AB, Alves VDM. Vulnerability biomarkers for mental illness and suicide risk: Regards for the development of pharmacological and psychological therapies. CURRENT RESEARCH IN BEHAVIORAL SCIENCES 2021. [DOI: 10.1016/j.crbeha.2021.100050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
14
|
Lee JS, Kang W, Kang Y, Kim A, Han KM, Tae WS, Ham BJ. Alterations in the Occipital Cortex of Drug-Naïve Adults With Major Depressive Disorder: A Surface-Based Analysis of Surface Area and Cortical Thickness. Psychiatry Investig 2021; 18:1025-1033. [PMID: 34666430 PMCID: PMC8542746 DOI: 10.30773/pi.2021.0099] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/27/2021] [Accepted: 07/22/2021] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Advances in surface-based morphometric methods have allowed researchers to separate cortical volume into cortical thickness (CTh) and surface area (SA). Although CTh alterations in major depressive disorder (MDD) have been observed in numerous studies, few studies have described significant SA alterations. Our study aimed to measure patients' SAs and to compare it with their CTh to examine whether SA exhibits alteration patterns that differ from those of CTh in drug-naïve patients with MDD. METHODS A total of 71 drug-naïve MDD patients and 111 healthy controls underwent structural magnetic resonance imaging, and SA and CTh were analyzed between the groups. RESULTS We found a smaller SA in the left superior occipital gyrus (L-SOG) in drug-naïve patients with MDD. In the CTh analysis, the bilateral fusiform gyrus, left middle occipital gyrus, left temporal superior gyrus, and right posterior cingulate showed thinner cortices in patients with MDD, while the CTh of the bilateral SOG, right straight gyrus, right posterior cingulate, and left lingual gyrus were increased. CONCLUSION Compared with the bilateral occipito-temporal changes in CTh, SA alterations in patients with MDD were confined to the L-SOG. These findings may improve our understanding of the neurobiological mechanisms of SA alteration in relation to MDD.
Collapse
Affiliation(s)
- Jee Soo Lee
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Wooyoung Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Youbin Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Aram Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyu-Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo-Suk Tae
- Brain Convergence Research Center, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
Labonté B, Abdallah K, Maussion G, Yerko V, Yang J, Bittar T, Quessy F, Golden SA, Navarro L, Checknita D, Gigek C, Lopez JP, Neve RL, Russo SJ, Tremblay RE, Côté G, Meaney MJ, Mechawar N, Nestler EJ, Turecki G. Regulation of impulsive and aggressive behaviours by a novel lncRNA. Mol Psychiatry 2021; 26:3751-3764. [PMID: 31907380 PMCID: PMC7436429 DOI: 10.1038/s41380-019-0637-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/26/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022]
Abstract
High impulsive and aggressive traits associate with poor behavioural self-control. Despite their importance in predicting behavioural negative outcomes including suicide, the molecular mechanisms underlying the expression of impulsive and aggressive traits remain poorly understood. Here, we identified and characterized a novel long noncoding RNA (lncRNA), acting as a regulator of the monoamine oxidase A (MAOA) gene in the brain, and named it MAOA-associated lncRNA (MAALIN). Our results show that in the brain of suicide completers, MAALIN is regulated by a combination of epigenetic mechanisms including DNA methylation and chromatin modifications. Elevated MAALIN in the dentate gyrus of impulsive-aggressive suicides was associated with lower MAOA expression. Viral overexpression of MAALIN in neuroprogenitor cells decreased MAOA expression while CRISPR-mediated knock out resulted in elevated MAOA expression. Using viral-mediated gene transfer, we confirmed that MAALIN in the hippocampus significantly decreases MAOA expression and exacerbates the expression of impulsive-aggressive behavioural traits in CD1 aggressive mice. Overall, our findings suggest that variations in DNA methylation mediate the differential expression of a novel lncRNA that acts on MAOA expression to regulate impulsive-aggressive behaviours.
Collapse
Affiliation(s)
- Benoit Labonté
- Centre de Recherche CERVO, Department of Neuroscience and Psychiatry, Laval University, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada.
| | - Khaled Abdallah
- Centre de Recherche CERVO, Department of Neuroscience and Psychiatry, Laval University, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Gilles Maussion
- McGill Group for Suicide Studies, Douglas Research Centre, Department of Psychiatry, McGill University, Frank B. Common Pavilion, 6875 LaSalle Blvd., Montreal, QC, H4H 1R3, Canada
| | - Volodymyr Yerko
- McGill Group for Suicide Studies, Douglas Research Centre, Department of Psychiatry, McGill University, Frank B. Common Pavilion, 6875 LaSalle Blvd., Montreal, QC, H4H 1R3, Canada
| | - Jennie Yang
- McGill Group for Suicide Studies, Douglas Research Centre, Department of Psychiatry, McGill University, Frank B. Common Pavilion, 6875 LaSalle Blvd., Montreal, QC, H4H 1R3, Canada
| | - Thibault Bittar
- Centre de Recherche CERVO, Department of Neuroscience and Psychiatry, Laval University, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Francis Quessy
- Centre de Recherche CERVO, Department of Neuroscience and Psychiatry, Laval University, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Sam A Golden
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luis Navarro
- Unitat de Genètica Molecular Institut de Biomedicina de València, Valencia, Spain
| | - Dave Checknita
- McGill Group for Suicide Studies, Douglas Research Centre, Department of Psychiatry, McGill University, Frank B. Common Pavilion, 6875 LaSalle Blvd., Montreal, QC, H4H 1R3, Canada
| | - Carolina Gigek
- McGill Group for Suicide Studies, Douglas Research Centre, Department of Psychiatry, McGill University, Frank B. Common Pavilion, 6875 LaSalle Blvd., Montreal, QC, H4H 1R3, Canada
| | - Juan Pablo Lopez
- McGill Group for Suicide Studies, Douglas Research Centre, Department of Psychiatry, McGill University, Frank B. Common Pavilion, 6875 LaSalle Blvd., Montreal, QC, H4H 1R3, Canada
| | - Rachael L Neve
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Richard E Tremblay
- Research Unit on Children's Psychosocial Maladjustment, Université de Montréal, Montreal, QC, Canada
| | - Gilles Côté
- Philippe-Pinel Institute of Montreal, University of Montreal, Montreal, QC, Canada
| | - Michael J Meaney
- McGill Group for Suicide Studies, Douglas Research Centre, Department of Psychiatry, McGill University, Frank B. Common Pavilion, 6875 LaSalle Blvd., Montreal, QC, H4H 1R3, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Research Centre, Department of Psychiatry, McGill University, Frank B. Common Pavilion, 6875 LaSalle Blvd., Montreal, QC, H4H 1R3, Canada
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Research Centre, Department of Psychiatry, McGill University, Frank B. Common Pavilion, 6875 LaSalle Blvd., Montreal, QC, H4H 1R3, Canada.
| |
Collapse
|
16
|
Cocaine-related DNA methylation in caudate neurons alters 3D chromatin structure of the IRXA gene cluster. Mol Psychiatry 2021; 26:3134-3151. [PMID: 33046833 PMCID: PMC8039060 DOI: 10.1038/s41380-020-00909-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/14/2020] [Accepted: 10/01/2020] [Indexed: 02/01/2023]
Abstract
Epigenetic mechanisms, like those involving DNA methylation, are thought to mediate the relationship between chronic cocaine dependence and molecular changes in addiction-related neurocircuitry, but have been understudied in human brain. We initially used reduced representation bisulfite sequencing (RRBS) to generate a methylome-wide profile of cocaine dependence in human post-mortem caudate tissue. We focused on the Iroquois Homeobox A (IRXA) gene cluster, where hypomethylation in exon 3 of IRX2 in neuronal nuclei was associated with cocaine dependence. We replicated this finding in an independent cohort and found similar results in the dorsal striatum from cocaine self-administering mice. Using epigenome editing and 3C assays, we demonstrated a causal relationship between methylation within the IRX2 gene body, CTCF protein binding, three-dimensional (3D) chromatin interaction, and gene expression. Together, these findings suggest that cocaine-related hypomethylation of IRX2 contributes to the development and maintenance of cocaine dependence through alterations in 3D chromatin structure in the caudate nucleus.
Collapse
|
17
|
Alfonso S, Gesto M, Sadoul B. Temperature increase and its effects on fish stress physiology in the context of global warming. JOURNAL OF FISH BIOLOGY 2021; 98:1496-1508. [PMID: 33111333 DOI: 10.1111/jfb.14599] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 05/07/2023]
Abstract
The capacity of fishes to cope with environmental variation is considered to be a main determinant of their fitness and is partly determined by their stress physiology. By 2100, global ocean temperature is expected to rise by 1-4°C, with potential consequences for stress physiology. Global warming is affecting animal populations worldwide through chronic temperature increases and an increase in the frequency of extreme heatwave events. As ectotherms, fishes are expected to be particularly vulnerable to global warming. Although little information is available about the effects of global warming on stress physiology in nature, multiple studies describe the consequences of temperature increases on stress physiology in controlled laboratory conditions, providing insight into what can be expected in the wild. Chronic temperature increase constitutes a physiological load that can alter the ability of fishes to cope with additional stressors, which might compromise their fitness. In addition, rapid temperature increases are known to induce acute stress responses in fishes and might be of ecological relevance in particular situations. This review summarizes knowledge about effects of temperature increases on the stress physiology of fishes and discusses these in the context of global warming.
Collapse
Affiliation(s)
- Sébastien Alfonso
- COISPA Tecnologia & Ricerca, Stazione Sperimentale per lo Studio delle Risorse del Mare, Bari, Italy
| | - Manuel Gesto
- Section for Aquaculture, DTU Aqua, Technical University of Denmark, Hirtshals, Denmark
| | - Bastien Sadoul
- MARBEC, Ifremer, IRD, UM2, CNRS, Sète, France
- ESE, Ecology and Ecosystem Health, Institut Agro, INRAE, Rennes Cedex, France
| |
Collapse
|
18
|
Hung JH, Cheng HY, Tsai YC, Pan HA, Omar HA, Chiu CC, Su YM, Lin YM, Teng YN. LRWD1 expression is regulated through DNA methylation in human testicular embryonal carcinoma cells. Basic Clin Androl 2021; 31:12. [PMID: 34011267 PMCID: PMC8136200 DOI: 10.1186/s12610-021-00130-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Sperm growth and maturation are correlated with the expression levels of Leucine-rich repeat and WD repeat-containing protein 1 (LRWD1), a widely expressed protein in the human testicles. The decrease in LRWD1 cellular level was linked to the reduction in cell growth and mitosis and the rise in cell microtubule atrophy rates. Since DNA methylation has a major regulatory role in gene expression, this study aimed at exploring the effect of the modulation of DNA methylation on LRWD1 expression levels. RESULTS The results revealed the presence of a CpG island up of 298 bps (- 253 ~ + 45) upon LRWD1 promoter in NT2/D1 cells. The hypermethylation of the LRWD1 promoter was linked to a reduction in the transcription activity in NT2/D1 cells, as indicated by luciferase reporter assay. The methylation activator, floxuridine, confirmed the decrease in the LRWD1 promoter transcriptional activity. On the other hand, 5-Aza-2'-deoxycytidine (5-Aza-dc, methylation inhibitor), significantly augmented LRWD1 promoter activity and the expression levels of mRNA and proteins. Furthermore, DNA methylation status of LRWD1 promoter in human sperm genomic DNA samples was analyzed. The results indicated that methylation of LRWD1 promoter was correlated to sperm activity. CONCLUSIONS Thus, the regulation of LRWD1 expression is correlated with the methylation status of LRWD1 promoter, which played a significant role in the modulation of spermatogenesis, sperm motility, and vitality. Based on these results, the methylation status of LRWD1 promoter may serve as a novel molecular diagnostic marker or a therapeutic target in males' infertility.
Collapse
Affiliation(s)
- Jui-Hsiang Hung
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Han-Yi Cheng
- Department of Biological Sciences and Technology, National University of Tainan, No.33, Sec. 2, Shulin St., West Central District, Tainan City, 700, Taiwan
| | - Yung-Chieh Tsai
- Department of Obstetrics and Gynecology, Chi-Mei Medical Center; Department of Sport Management, and Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | | | - Hany A Omar
- Sharjah Institute for Medical Research and College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yin-Mei Su
- Department of Biological Sciences and Technology, National University of Tainan, No.33, Sec. 2, Shulin St., West Central District, Tainan City, 700, Taiwan
| | - Yung-Ming Lin
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, No.33, Sec. 2, Shulin St., West Central District, Tainan City, 700, Taiwan.
| |
Collapse
|
19
|
Lewis CR, Breitenstein RS, Henderson A, Sowards HA, Piras IS, Huentelman MJ, Doane LD, Lemery-Chalfant K. Harsh Parenting Predicts Novel HPA Receptor Gene Methylation and NR3C1 Methylation Predicts Cortisol Daily Slope in Middle Childhood. Cell Mol Neurobiol 2021; 41:783-793. [PMID: 32472381 DOI: 10.1007/s10571-020-00885-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/23/2020] [Indexed: 01/16/2023]
Abstract
Adverse experiences in childhood are associated with altered hypothalamic-pituitary-adrenal (HPA) axis function and negative health outcomes throughout life. It is now commonly accepted that abuse and neglect can alter epigenetic regulation of HPA genes. Accumulated evidence suggests harsh parenting practices such as spanking are also strong predictors of negative health outcomes. We predicted harsh parenting at 2.5 years old would predict HPA gene DNA methylation similarly to abuse and neglect, and cortisol output at 8.5 years old. Saliva samples were collected three times a day across 3 days to estimate cortisol diurnal slopes. Methylation was quantified using the Illumina Infinium MethylationEPIC array BeadChip (850 K) with DNA collected from buccal cells. We used principal components analysis to compute a summary statistic for CpG sites across candidate genes. The first and second components were used as outcome variables in mixed linear regression analyses with harsh parenting as a predictor variable. We found harsh parenting significantly predicted methylation of several HPA axis genes, including novel gene associations with AVPRB1, CRHR1, CRHR2, and MC2R (FDR corrected p < 0.05). Further, we found NR3C1 methylation predicted a steeper diurnal cortisol slope. Our results extend the current literature by demonstrating harsh parenting may influence DNA methylation similarly to more extreme early life experiences such as abuse and neglect. Further, we show NR3C1 methylation is associated with diurnal HPA function. Elucidating the molecular consequences of harsh parenting on health can inform best parenting practices and provide potential treatment targets for common complex disorders.
Collapse
Affiliation(s)
- Candace R Lewis
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA.
- Psychology, Arizona State University, Tempe, AZ, USA.
| | | | - Adrienne Henderson
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | | | - Ignazio S Piras
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | - Matthew J Huentelman
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | - Leah D Doane
- Psychology, Arizona State University, Tempe, AZ, USA
| | | |
Collapse
|
20
|
Richetto J, Meyer U. Epigenetic Modifications in Schizophrenia and Related Disorders: Molecular Scars of Environmental Exposures and Source of Phenotypic Variability. Biol Psychiatry 2021; 89:215-226. [PMID: 32381277 DOI: 10.1016/j.biopsych.2020.03.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/19/2020] [Accepted: 03/16/2020] [Indexed: 12/18/2022]
Abstract
Epigenetic modifications are increasingly recognized to play a role in the etiology and pathophysiology of schizophrenia and other psychiatric disorders with developmental origins. Here, we summarize clinical and preclinical findings of epigenetic alterations in schizophrenia and relevant disease models and discuss their putative origin. Recent findings suggest that certain schizophrenia risk loci can influence stochastic variation in gene expression through epigenetic processes, highlighting the intricate interaction between genetic and epigenetic control of neurodevelopmental trajectories. In addition, a substantial portion of epigenetic alterations in schizophrenia and related disorders may be acquired through environmental factors and may be manifested as molecular "scars." Some of these scars can influence brain functions throughout the entire lifespan and may even be transmitted across generations via epigenetic germline inheritance. Epigenetic modifications, whether caused by genetic or environmental factors, are plausible molecular sources of phenotypic heterogeneity and offer a target for therapeutic interventions. The further elucidation of epigenetic modifications thus may increase our knowledge regarding schizophrenia's heterogeneous etiology and pathophysiology and, in the long term, may advance personalized treatments through the use of biomarker-guided epigenetic interventions.
Collapse
Affiliation(s)
- Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, and Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, and Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Kozhuharova P, Diaconescu AO, Allen P. Reduced cortical GABA and glutamate in high schizotypy. Psychopharmacology (Berl) 2021; 238:2459-2470. [PMID: 34146134 PMCID: PMC8373725 DOI: 10.1007/s00213-021-05867-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 05/02/2021] [Indexed: 12/04/2022]
Abstract
RATIONALE Abnormal functioning of the inhibitory gamma-aminobutyric acid (GABA) and excitatory (glutamate) systems is proposed to play a role in the development of schizophrenia spectrum disorder. Although results are mixed, previous 1H-magnetic resonance spectroscopy (MRS) studies in schizophrenia and clinical high-risk samples report these metabolites are altered in comparison to healthy controls. Currently, however, there are few studies of these metabolites in schizotypy samples, a personality dimension associated with the experience of schizophrenia and psychosis-like symptoms. OBJECTIVES We investigated if GABA and glutamate metabolite concentrations are altered in people with high schizotypy. We also explored the relationship between resilience to stress, GABA metabolite concentrations and schizotypy. METHODS We used MRS to examine GABA and glutamate levels in the medial prefrontal cortex in people with low and high schizotypy traits as assessed with the Schizotypal Personality Questionnaire. Resilience to stress was assessed using the Connor-Davidson Resilience Scale. RESULTS Compared to individuals with low schizotypy traits, high schizotypy individuals showed lower cortical prefrontal GABA (F (1,38) = 5.18, p = 0.03, η2 = 0.09) and glutamate metabolite levels (F (1, 49) = 6.25, p = 0.02, η2 = 0.02). Furthermore, participants with high GABA and high resilience levels were significantly more likely to be in the low schizotypy group than participants with low GABA and high resilience or high GABA and low resilience (95% CI 1.07-1.34, p < .001). CONCLUSIONS These findings demonstrate that subclinical schizotypal traits are associated with abnormal functioning of both inhibitory and excitatory systems and suggest that these transmitters are implicated in a personality trait believed to be on a continuum with psychosis.
Collapse
Affiliation(s)
- Petya Kozhuharova
- Centre for Cognition, Neuroscience and Neuroimaging, Department of Psychology, University of Roehampton, Holybourne Ave, Roehampton, London, SW15 4JD, UK.
| | - Andreea O Diaconescu
- Department of Psychiatry, Brain and Therapeutics, Krembil Centre for Neuroinformatics, CAMH, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Paul Allen
- Centre for Cognition, Neuroscience and Neuroimaging, Department of Psychology, University of Roehampton, Holybourne Ave, Roehampton, London, SW15 4JD, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
22
|
Catale C, Bussone S, Lo Iacono L, Viscomi MT, Palacios D, Troisi A, Carola V. Exposure to different early-life stress experiences results in differentially altered DNA methylation in the brain and immune system. Neurobiol Stress 2020; 13:100249. [PMID: 33344704 PMCID: PMC7739045 DOI: 10.1016/j.ynstr.2020.100249] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/15/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
The existence of a proportional relationship between the number of early-life stress (ELS) events experienced and the impoverishment of child mental health has been hypothesized. However, different types of ELS experiences may be associated with different neuro-psycho-biological impacts, due to differences in the intrinsic nature of the stress. DNA methylation is one of the molecular mechanisms that have been implicated in the "translation" of ELS exposure into neurobiological and behavioral abnormalities during adulthood. Here, we investigated whether different ELS experiences resulted in differential impacts on global DNA methylation levels in the brain and blood samples from mice and humans. ELS exposure in mice resulted in observable changes in adulthood, with exposure to social isolation inducing more dramatic alterations in global DNA methylation levels in several brain structures compared with exposure to a social threatening environment. Moreover, these two types of stress resulted in differential impacts on the epigenetic programming of different brain regions and cellular populations, namely microglia. In a pilot clinical study, blood global DNA methylation levels and exposure to childhood neglect or abuse were investigated in patients presenting with major depressive disorder or substance use disorder. A significant effect of the mental health diagnosis on global methylation levels was observed, but no effect of either childhood abuse or neglect was detected. These findings demonstrate that different types of ELS have differential impacts on epigenetic programming, through DNA methylation in specific brain regions, and that these differential impacts are associated with the different behavioral outcomes observed after ELS experiences.
Collapse
Affiliation(s)
- Clarissa Catale
- Department of Psychology, Ph.D. Program in "Behavioral Neuroscience", Sapienza University of Rome, Rome, Italy
| | - Silvia Bussone
- Department of Dynamic and Clinical Psychology, Sapienza University of Rome, Rome, Italy
| | - Luisa Lo Iacono
- Department of Psychology, Sapienza University of Rome, Rome, Italy.,IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health, Section of Histology and Embryology, University "Cattolica Del S. Cuore", Rome, Italy
| | | | - Alfonso Troisi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Valeria Carola
- Department of Dynamic and Clinical Psychology, Sapienza University of Rome, Rome, Italy.,IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
23
|
Abstract
The Norway rat has important impacts on our life. They are amongst the most used research subjects, resulting in ground-breaking advances. At the same time, wild rats live in close association with us, leading to various adverse interactions. In face of this relevance, it is surprising how little is known about their natural behaviour. While recent laboratory studies revealed their complex social skills, little is known about their social behaviour in the wild. An integration of these different scientific approaches is crucial to understand their social life, which will enable us to design more valid research paradigms, develop more effective management strategies, and to provide better welfare standards. Hence, I first summarise the literature on their natural social behaviour. Second, I provide an overview of recent developments concerning their social cognition. Third, I illustrate why an integration of these areas would be beneficial to optimise our interactions with them.
Collapse
Affiliation(s)
- Manon K Schweinfurth
- School of Psychology and Neuroscience, University of St AndrewsSt AndrewsUnited Kingdom
| |
Collapse
|
24
|
Zhong H, Rong J, Zhu C, Liang M, Li Y, Zhou R. Epigenetic Modifications of GABAergic Interneurons Contribute to Deficits in Adult Hippocampus Neurogenesis and Depression-Like Behavior in Prenatally Stressed Mice. Int J Neuropsychopharmacol 2020; 23:274-285. [PMID: 32211762 PMCID: PMC7177164 DOI: 10.1093/ijnp/pyaa020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/15/2020] [Accepted: 03/19/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Prenatal stress (PRS) is considered a risk factor for depressive disorder. Adult hippocampal neurogenesis is believed to play a role in the regulation of affective behaviors. GABAergic interneuron is a key modulator in adult hippocampal neurogenesis. Growing evidence indicates that PRS has adverse effects on adult hippocampal neurogenesis and DNA epigenetic modifications of the GABAergic system. The aim of this study was to investigate whether epigenetic GABAergic dysfunction participates in the negative impact of PRS on adult hippocampal neurogenesis and related emotional behaviors. METHODS Behavioral tests were used to explore PRS-induced depression-like behaviors of adult female mice. Immunohistochemistry staining, real-time reverse transcription-polymerase chain reaction, western blot, and chromatin immunoprecipitation were employed to detect adult neurogenesis and epigenetic changes of the GABAergic system in the hippocampus of PRS mice. RESULTS PRS mice developed a depression phenotype accompanied by the inhibited maturation of hippocampal newborn neurons. Compared with control mice, PRS mice showed decreased expression of glutamic acid decarboxylase 67 at the mRNA and protein levels. GABAA receptor agonist phenobarbital could rectify the decrease of 5-bromo-2-deoxyuridine/neuronal nuclei double-positive (BrdU+/NeuN+) cells in PRS mice. PRS mice also showed increased expression of DNA methyltransferase 1 and increased binding of DNA methyltransferase 1 to glutamic acid decarboxylase 67 promoter region. The treatment with DNA methyltransferase 1 inhibitor 5-aza-deoxycytidine restored the decrease of BrdU+/NeuN+ cells and depression-like behaviors in PRS mice via improving GABAergic system. CONCLUSIONS The present results indicate that epigenetic changes of the GABAergic system are responsible for adult hippocampus neurogenesis and depression-like behaviors in PRS mice.
Collapse
Affiliation(s)
- Haiquan Zhong
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Jing Rong
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Chunting Zhu
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Min Liang
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Yingchun Li
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Rong Zhou
- Department of Physiology, Nanjing Medical University, Jiangsu, China,Correspondence: Rong Zhou, PhD, Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing City, Jiangsu Province, China 211166 ()
| |
Collapse
|
25
|
Fabri M, Fortuna S. Maria Montessori and Neuroscience: The Trailblazing Insights of an Exceptional Mind. Neuroscientist 2020; 26:394-401. [PMID: 32111132 DOI: 10.1177/1073858420902677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This comment presents Maria Montessori (1870-1952) and highlights that her child-centered method of education is based on brilliant intuitions, which were confirmed by neuroscience research many decades later, such as the distinction of three critical periods in children's psychobiological development; the importance of the environment in supporting cerebral development and in promoting learning, as well as of affective stimulation in psychological growth and maturation; the specific neural structure of humans that specifically enables the acquisition of a language; the vital role of fine object manipulation in neuropsychological development, and of the physical exercise in brain and nervous system development.
Collapse
Affiliation(s)
- Mara Fabri
- Department of Clinical and Experimental Medicine, Marche Polytechnic University, Ancona, Italy
| | - Stefania Fortuna
- Department of Molecular and Clinical Sciences, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
26
|
Abstract
Depression is one of the most common psychiatric disorders affecting public health. Studies over the past years suggest that the methylations of some specific genes such as BDNF, SLC6A4, and NR3C1 play an important role in the development of depression. Recently, epigenetic evidences suggest that the expression levels of DNA methyltransferases differ in several brain areas including the prefrontal cortex, hippocampus, amygdala, and nucleus accumbens in depression patients and animal models, but the potential link between the expression levels of DNA methylatransferases and the methylations of specific genes needs further investigation to clarify the pathogenesis of depression.
Collapse
Affiliation(s)
- Zhenghao Duan
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jie Lu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
27
|
Lin E, Tsai SJ. Gene-Environment Interactions and Role of Epigenetics in Anxiety Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1191:93-102. [PMID: 32002924 DOI: 10.1007/978-981-32-9705-0_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Several environmental risk factors such as early adverse childhood experiences, stress, and stressful life events are associated with anxiety disorders. Current approaches such as epigenetics and gene-environment interactions were used to identify candidate biomarkers for anxiety disorders to assess determinants of disease. In this chapter, in relation to gene-environment interactions, a variety of association studies regarding anxiety disorders were surveyed. We then showed supporting results from recent association studies such as human studies and animal models in terms of the epigenetic contribution to disease susceptibility to anxiety disorders. At last, future directions and limitations are highlighted. With the advances in multi-omics technologies, innovative ideas regarding disease prevention and drug responsiveness in anxiety disorders require further research in epigenetics and gene-environment interactions.
Collapse
Affiliation(s)
- Eugene Lin
- Department of Biostatistics, University of Washington, Seattle, WA, USA.,Department of Electrical & Computer Engineering, University of Washington, Seattle, WA, USA.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan. .,Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan. .,Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
28
|
Shaw JC, Crombie GK, Zakar T, Palliser HK, Hirst JJ. Perinatal compromise contributes to programming of GABAergic and glutamatergic systems leading to long-term effects on offspring behaviour. J Neuroendocrinol 2020; 32:e12814. [PMID: 31758712 DOI: 10.1111/jne.12814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/30/2019] [Accepted: 11/20/2019] [Indexed: 01/01/2023]
Abstract
Extensive evidence now shows that adversity during the perinatal period is a significant risk factor for the development of neurodevelopmental disorders long after the causative event. Despite stemming from a variety of causes, perinatal compromise appears to have similar effects on the developing brain, thereby resulting in behavioural disorders of a similar nature. These behavioural disorders occur in a sex-dependent manner, with males affected more by externalising behaviours such as attention deficit hyperactivity disorder (ADHD) and females by internalising behaviours such as anxiety. Regardless of the causative event or the sex of the offspring, these disorders may begin in childhood or adolescence but extend into adulthood. A mechanism by which adverse events in the perinatal period impact later in life behaviour has been shown to be the changing epigenetic landscape. Methylation of the GAD1/GAD67 gene, which encodes the key glutamate-to-GABA-synthesising enzyme glutamate decarboxylase 1, resulting in increased levels of glutamate, is one epigenetic mechanism that may account for a tendency towards excitation in disorders such as ADHD. Exposure of the fetus or the neonate to high levels of cortisol may be the mediator between perinatal compromise and poor behavioural outcomes because evidence suggests that increased glucocorticoid exposure triggers widespread changes in the epigenetic landscape. This review summarises the current evidence and recent literature about the impact of various perinatal insults on the epigenome and the common mechanisms that may explain the similarity of behavioural outcomes occurring following diverse perinatal compromise.
Collapse
Affiliation(s)
- Julia C Shaw
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Gabrielle K Crombie
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Tamas Zakar
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
29
|
Zhuang YY, Xiang L, Wen XR, Shen RJ, Zhao N, Zheng SS, Han RY, Qu J, Lu F, Jin ZB. Slc7a14 Is Indispensable in Zebrafish Retinas. Front Cell Dev Biol 2019; 7:333. [PMID: 31921845 PMCID: PMC6920099 DOI: 10.3389/fcell.2019.00333] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Abstract
Previous study has identified SLC7A14 as a new causative gene of retinitis pigmentosa (RP). However, the role of SLC7A14 has not been fully characterized. The goal of this study was to investigate the biological features of slc7a14 in zebrafish. To determine the expression of slc7a14 in developing zebrafish, we performed in situ hybridization (ISH) and quantitative real-time PCR. Morpholino knockdown and overexpression experiments were performed to study the role of slc7a14 in zebrafish retinas. Immunostaining was carried out to observe structural changes. Visual motor responses (VMR) and optokinetic responses (OKR) were analyzed to assess visual behaviors. Terminal deoxynucleotidyl transferase (dUTP) nick-end labeling (TUNEL) staining was performed to survey apoptotic retinal cells. We found that slc7a14 was highly expressed in neuronal tissues, including the brain, spinal cord and retina, and that the expression levels increased during early embryogenesis. Consistently, ISH showed a similar expression pattern. Knockdown of slc7a14 led to dose-dependent microphthalmia that was reversed by overexpression. The immunostaining results revealed that the rod-specific protein zpr-3 and the retinal pigment epithelium-specific protein zpr-2 (decreased to 44.48%) were significantly suppressed in the slc7a14-silenced morphants. Notably, visual behaviors (the VMR and the OKR) were severely impaired in the slc7a14-deficient morphant, especially the VMR OFF response. In addition, apoptotic cells were observed in the retina at 3 days post fertilization (dpf) and 5 dpf by TUNEL assay. Our results demonstrated that slc7a14 is essential for visually mediated behaviors in zebrafish. Temporary silencing of slc7a14 in larvae led to severe visual impairments, consistent with the manifestations observed in RP patients. Our findings provide further insights into the genetic mechanisms of RP predisposition caused by SLC7A14 mutations.
Collapse
Affiliation(s)
- You-Yuan Zhuang
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| | - Lue Xiang
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| | - Xin-Ran Wen
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| | - Ren-Juan Shen
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| | - Ning Zhao
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| | - Si-Si Zheng
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| | - Ru-Yi Han
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| | - Jia Qu
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| | - Fan Lu
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| | - Zi-Bing Jin
- Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ophthalmology, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, China
| |
Collapse
|
30
|
Tomasi J, Lisoway AJ, Zai CC, Harripaul R, Müller DJ, Zai GCM, McCabe RE, Richter MA, Kennedy JL, Tiwari AK. Towards precision medicine in generalized anxiety disorder: Review of genetics and pharmaco(epi)genetics. J Psychiatr Res 2019; 119:33-47. [PMID: 31563039 DOI: 10.1016/j.jpsychires.2019.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023]
Abstract
Generalized anxiety disorder (GAD) is a prevalent and chronic mental disorder that elicits widespread functional impairment. Given the high degree of non-response/partial response among patients with GAD to available pharmacological treatments, there is a strong need for novel approaches that can optimize outcomes, and lead to medications that are safer and more effective. Although investigations have identified interesting targets predicting treatment response through pharmacogenetics (PGx), pharmaco-epigenetics, and neuroimaging methods, these studies are often solitary, not replicated, and carry several limitations. This review provides an overview of the current status of GAD genetics and PGx and presents potential strategies to improve treatment response by combining better phenotyping with PGx and improved analytical methods. These strategies carry the dual benefit of delivering data on biomarkers of treatment response as well as pointing to disease mechanisms through the biology of the markers associated with response. Overall, these efforts can serve to identify clinical, genetic, and epigenetic factors that can be incorporated into a pharmaco(epi)genetic test that may ultimately improve treatment response and reduce the socioeconomic burden of GAD.
Collapse
Affiliation(s)
- Julia Tomasi
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Amanda J Lisoway
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Clement C Zai
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Ricardo Harripaul
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Molecular Neuropsychiatry & Development (MiND) Lab, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Daniel J Müller
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Gwyneth C M Zai
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; General Adult Psychiatry and Health Systems Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Randi E McCabe
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Anxiety Treatment and Research Clinic, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada
| | - Margaret A Richter
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Frederick W. Thompson Anxiety Disorders Centre, Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - James L Kennedy
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Arun K Tiwari
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
31
|
Alese OO, Mabandla MV. Transgenerational deep sequencing revealed hypermethylation of hippocampal mGluR1 gene with altered mRNA expression of mGluR5 and mGluR3 associated with behavioral changes in Sprague Dawley rats with history of prolonged febrile seizure. PLoS One 2019; 14:e0225034. [PMID: 31710636 PMCID: PMC6844483 DOI: 10.1371/journal.pone.0225034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/27/2019] [Indexed: 02/04/2023] Open
Abstract
The impact of febrile seizure has been shown to transcend immediate generation with the alteration of glutamatergic pathway being implicated. However, transgenerational effects of this neurological disorder particularly prolonged febrile seizure (PFS) on neurobehavioral study and methylation profile is unknown. We therefore hypothesized that transgenerational impact of prolonged febrile seizure is dependent on methylation of hippocampal mGluR1 gene. Prolonged febrile seizure was induced on post-natal day (PND) 14, by injecting lipopolysaccharide (LPS; 217μg/kg ip) and kainic acid (KA; 1.83 mg/kg ip). Sucrose preference test (SPT) and Forced swim test (FST) were carried out in the first generation (F0) of animals at PND37 and PND60. The F0 rats were decapitated at PND 14, 37 and 60 which corresponded to childhood, adolescent and adulthood respectively and their hippocampal tissue collected. The second generation (F1) rats were obtained by mating F0 generation at PND 60 across different groups, F1 rats were subjected to SPT and FST test on PND 37 only. Decapitation of F1rats and collection of hippocampal tissues were done on PND 14 and 37. Assessment of mGluR5 and mGluR3 mRNA was done with PCR while mGluR1 methylation profile was assessed with the Quantitative MassARRAY analysis. Results showed that PFS significantly leads to decreased sucrose consumption in the SPT and increased immobility time in the FST in both generations of rats. It also leads to significant decrease in mGluR5 mRNA expression with a resultant increased expression of mGluR3 mRNA expression and hypermethylation of mGluR1 gene across both generations of rats. This study suggested that PFS led to behavioral changes which could be transmitted on to the next generation in rats.
Collapse
MESH Headings
- Animals
- Base Sequence
- Behavior, Animal
- DNA Methylation/genetics
- High-Throughput Nucleotide Sequencing
- Hippocampus/metabolism
- Immobilization
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptor, Metabotropic Glutamate 5/genetics
- Receptor, Metabotropic Glutamate 5/metabolism
- Receptors, Metabotropic Glutamate/genetics
- Receptors, Metabotropic Glutamate/metabolism
- Seizures, Febrile/genetics
- Sucrose
- Swimming
Collapse
Affiliation(s)
- Oluwole Ojo Alese
- Department of Human Physiology, College of Health Sciences, University of Kwazulu-Natal, Durban, South Africa
| | - Musa V. Mabandla
- Department of Human Physiology, College of Health Sciences, University of Kwazulu-Natal, Durban, South Africa
| |
Collapse
|
32
|
Yue ST, Zhang J, Ma DH. [Research advances in the effect of environmental stress in the neonatal intensive care unit on the neurodevelopment of preterm infants and its epigenetics]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:1144-1147. [PMID: 31753099 PMCID: PMC7389294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/06/2019] [Indexed: 11/12/2023]
Abstract
Adverse environmental stimulation in the neonatal intensive care unit (NICU) can affect neurodevelopment through epigenetic modification and thus has adverse effects on the long-term developmental outcome of preterm infants. Developmental care can reverse epigenetic changes in genes and promote neurodevelopment in preterm infants. This article reviews the influence of environmental stress in the NICU and developmental care on neurodevelopment in preterm infants, as well as related epigenetic effects, in order to provide a reference for epigenetic studies of preterm infants.
Collapse
Affiliation(s)
- Shao-Ting Yue
- Department of Nursing, Wuhan University School of Health Sciences, Wuhan 430071, China.
| | | | | |
Collapse
|
33
|
Yue ST, Zhang J, Ma DH. [Research advances in the effect of environmental stress in the neonatal intensive care unit on the neurodevelopment of preterm infants and its epigenetics]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:1144-1147. [PMID: 31753099 PMCID: PMC7389294 DOI: 10.7499/j.issn.1008-8830.2019.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/06/2019] [Indexed: 06/10/2023]
Abstract
Adverse environmental stimulation in the neonatal intensive care unit (NICU) can affect neurodevelopment through epigenetic modification and thus has adverse effects on the long-term developmental outcome of preterm infants. Developmental care can reverse epigenetic changes in genes and promote neurodevelopment in preterm infants. This article reviews the influence of environmental stress in the NICU and developmental care on neurodevelopment in preterm infants, as well as related epigenetic effects, in order to provide a reference for epigenetic studies of preterm infants.
Collapse
Affiliation(s)
- Shao-Ting Yue
- Department of Nursing, Wuhan University School of Health Sciences, Wuhan 430071, China.
| | | | | |
Collapse
|
34
|
Early Postnatal Treatment with Valproate Induces Gad1 Promoter Remodeling in the Brain and Reduces Apnea Episodes in Mecp2-Null Mice. Int J Mol Sci 2019; 20:ijms20205177. [PMID: 31635390 PMCID: PMC6834123 DOI: 10.3390/ijms20205177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 11/17/2022] Open
Abstract
The deletion of Mecp2, the gene encoding methyl-CpG-binding protein 2, causes severe breathing defects and developmental anomalies in mammals. In Mecp2-null mice, impaired GABAergic neurotransmission is demonstrated at the early stage of life. GABAergic dysfunction in neurons in the rostral ventrolateral medulla (RVLM) is considered as a primary cause of breathing abnormality in Mecp2-null mice, but its molecular mechanism is unclear. Here, we report that mRNA expression levels of Gad1, which encodes glutamate decarboxylase 67 (GAD67), in the RVLM of Mecp2-null (Mecp2-/y, B6.129P2(C)-Mecp2tm1.1Bird/J) mice is closely related to the methylation status of its promoter, and valproate (VPA) can upregulate transcription from Gad1 through epigenetic mechanisms. The administration of VPA (300 mg/kg/day) together with L-carnitine (30 mg/kg/day) from day 8 to day 14 after birth increased Gad1 mRNA expression in the RVLM and reduced apnea counts in Mecp2-/y mice on postnatal day 15. Cytosine methylation levels in the Gad1 promoter were higher in the RVLM of Mecp2-/y mice compared to wild-type mice born to C57BL/6J females, while VPA treatment decreased the methylation levels in Mecp2-/y mice. Chromatin immunoprecipitation assay revealed that the VPA treatment reduced the binding of methyl-CpG binding domain protein 1 (MBD1) to the Gad1 promoter in Mecp2-/y mice. These results suggest that VPA improves breathing of Mecp2-/y mice by reducing the Gad1 promoter methylation, which potentially leads to the enhancement of GABAergic neurotransmission in the RVLM.
Collapse
|
35
|
Sild M, Booij L. Histone deacetylase 4 (HDAC4): a new player in anorexia nervosa? Mol Psychiatry 2019; 24:1425-1434. [PMID: 30742020 DOI: 10.1038/s41380-019-0366-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/20/2018] [Accepted: 01/23/2019] [Indexed: 12/26/2022]
Abstract
Anorexia nervosa (AN) and other eating disorders continue to constitute significant challenges for individual and public health. AN is thought to develop as a result of complex interactions between environmental triggers, psychological risk factors, sociocultural influences, and genetic vulnerability. Recent research developments have highlighted a novel potentially relevant component in the AN etiology-activity of the histone deacetylase 4 (HDAC4) gene that has emerged in several recent studies related to AN. HDAC4 is a member of the ubiquitously important family of epigenetic modifier enzymes called histone deacetylases and has been implicated in processes related to the formation and function of the central nervous system (CNS), bone, muscle, and metabolism. In a family affected by eating disorders, a missense mutation in HDAC4 (A786T) was found to segregate with the illness. The relevance of this mutation in eating-related behaviors was further confirmed with mouse models. Despite the fact that HDAC4 has not been identified as a significant signal in genome-wide association studies in AN, several studies have found significant or near-significant methylation differences in HDAC4 locus in peripheral tissues of actively ill AN patients in comparison with different control groups. Limitations of these studies include a lack of understanding of to what extent the changes in methylation are predictive of AN as such changes might also occur as a consequence of the disease. It remains to be determined how methylation in peripheral tissues correlates with that in the CNS and how different methylation patterns affect HDAC4 expression. The present review discusses the findings and potential roles of HDAC4 in AN. Its emerging roles in learning and neuroplasticity may be specific and relevant for the etiology of AN and potentially lead to novel therapeutic approaches.
Collapse
Affiliation(s)
- Mari Sild
- Department of Psychology, Concordia University, Montreal, QC, Canada.,CHU Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Linda Booij
- Department of Psychology, Concordia University, Montreal, QC, Canada. .,CHU Sainte-Justine Hospital Research Center, Montreal, QC, Canada. .,Department of Psychiatry, McGill University, Montreal, QC, Canada. .,Department of Psychiatry, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
36
|
Abstract
OBJECTIVE Depression is associated with various environmental risk factors such as stress, childhood maltreatment experiences, and stressful life events. Current approaches to assess the pathophysiology of depression, such as epigenetics and gene-environment (GxE) interactions, have been widely leveraged to determine plausible markers, genes, and variants for the risk of developing depression. METHODS We focus on the most recent developments for genomic research in epigenetics and GxE interactions. RESULTS In this review, we first survey a variety of association studies regarding depression with consideration of GxE interactions. We then illustrate evidence of epigenetic mechanisms such as DNA methylation, microRNAs, and histone modifications to influence depression in terms of animal models and human studies. Finally, we highlight their limitations and future directions. CONCLUSION In light of emerging technologies in artificial intelligence and machine learning, future research in epigenetics and GxE interactions promises to achieve novel innovations that may lead to disease prevention and future potential therapeutic treatments for depression.
Collapse
Affiliation(s)
- Eugene Lin
- Department of Biostatistics, University of Washington, Seattle, WA , USA.,Department of Electrical & Computer Engineering, University of Washington, Seattle, WA, USA.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan.,Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
37
|
Shen XF, Yuan HB, Wang GQ, Xue H, Liu YF, Zhang CX. Role of DNA hypomethylation in lateral habenular nucleus in the development of depressive-like behavior in rats. J Affect Disord 2019; 252:373-381. [PMID: 30999094 DOI: 10.1016/j.jad.2019.03.062] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/02/2019] [Accepted: 03/19/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Lateral habenula nucleus (LHb) has recently been noted for its role in stress-induced depressive disorder. Yet little is known about the mechanisms by which external stimuli or depression induces pathological alteration in the LHb. METHODS Chronic unpredictable mild stress (CUMS) was employed to model depressive-like behaviors in adult rats. We examined expressions of DNA methyltransferases (Dnmts) mRNA and protein and global DNA methylation levels in LHb of CUMS-induced depressive rats. Then 5-aza-2'-deoxycytidine (5-aza), a Dnmts inhibitor, was infused into the LHb of native rats to test the effects of hypomethylation in the LHb. The gene expressions in the LHb and the levels of 5-HT and its metabolite 5-hydroxyindoleacetic acid (5-HIAA) in dorsal raphe nucleus (DRN) were examined in 5-aza infusion rats by quantitative real-time PCR and high performance liquid chromatography, respectively. RESULTS Rats were exposed to CUMS for 21 days and depressive-like behaviors were induced as expected. We observed significant decrease in mRNA and protein expressions of Dnmt1 and DNA hypomethylation in LHb of depressive rats. These phenomenon suggests that CUMS-induced depressive-like behaviors are related with DNA hypomethylation in the LHb. Local 5-aza infusion into LHb of native rat resulted in global DNA hypomethylation in the LHb and induced depressive-like behaviors which are featured with lack of interest and investment in the environment, behavioral despair and anhedonia. Moreover, DNA hypomethylation in the LHb increased transcription of β calcium/calmodulin dependent protein kinase II and glutamate receptor 1 in the LHb and attenuated the levels of 5-HT and 5-HIAA in the DRN. Our data suggested that alteration of DNA methylation in the LHb may control 5-HT neuronal activity in the DRN to regulate emotional state. CONCLUSIONS DNA hypomethylation in the LHb is involved in the development of depressive-like behavior and suitable methylation state contributes to the emotional stabilization.
Collapse
Affiliation(s)
- Xiang-Feng Shen
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Hai-Bo Yuan
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, China
| | - Guo-Qiang Wang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Hui Xue
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Yong-Feng Liu
- Department of Molecular Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| | - Chun-Xiao Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
38
|
Bludau A, Royer M, Meister G, Neumann ID, Menon R. Epigenetic Regulation of the Social Brain. Trends Neurosci 2019; 42:471-484. [PMID: 31103351 DOI: 10.1016/j.tins.2019.04.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022]
Abstract
Social behavior, a highly adaptive and crucial component of mammalian life, is regulated by particularly sensitive regulatory brain mechanisms. Substantial evidence implicates classical epigenetic mechanisms including histone modifications, DNA methylation, and nucleosome remodeling as well as nonclassical mechanisms mediated by noncoding RNA in the regulation of social behavior. These mechanisms collectively form the 'epigenetic network' that orchestrates genomic integration of salient and transient social experiences. Consequently, its dysregulation has been linked to behavioral deficits and psychopathologies. This review focuses on the role of the epigenetic network in regulating the enduring effects of social experiences during early-life, adolescence, and adulthood. We discuss research in animal models, primarily rodents, and associations between dysregulation of epigenetic mechanisms and human psychopathologies, specifically autism spectrum disorder (ASD) and schizophrenia.
Collapse
Affiliation(s)
- Anna Bludau
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Melanie Royer
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany; Biochemistry Center Regensburg (BZR), Laboratory of RNA Biology, University of Regensburg, Regensburg, Germany
| | - Gunter Meister
- Biochemistry Center Regensburg (BZR), Laboratory of RNA Biology, University of Regensburg, Regensburg, Germany
| | - Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Rohit Menon
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
39
|
Chistiakov DA, Chekhonin VP. Early-life adversity-induced long-term epigenetic programming associated with early onset of chronic physical aggression: Studies in humans and animals. World J Biol Psychiatry 2019; 20:258-277. [PMID: 28441915 DOI: 10.1080/15622975.2017.1322714] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objectives: To examine whether chronic physical aggression (CPA) in adulthood can be epigenetically programmed early in life due to exposure to early-life adversity. Methods: Literature search of public databases such as PubMed/MEDLINE and Scopus. Results: Children/adolescents susceptible for CPA and exposed to early-life abuse fail to efficiently cope with stress that in turn results in the development of CPA later in life. This phenomenon was observed in humans and animal models of aggression. The susceptibility to aggression is a complex trait that is regulated by the interaction between environmental and genetic factors. Epigenetic mechanisms mediate this interaction. Subjects exposed to stress early in life exhibited long-term epigenetic programming that can influence their behaviour in adulthood. This programming affects expression of many genes not only in the brain but also in other systems such as neuroendocrine and immune. Conclusions: The propensity to adult CPA behaviour in subjects experienced to early-life adversity is mediated by epigenetic programming that involves long-term systemic epigenetic alterations in a whole genome.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- a Department of Fundamental and Applied Neurobiology , Serbsky Federal Medical Research Center of Psychiatry and Narcology , Moscow , Russia
| | - Vladimir P Chekhonin
- a Department of Fundamental and Applied Neurobiology , Serbsky Federal Medical Research Center of Psychiatry and Narcology , Moscow , Russia.,b Department of Medical Nanobiotechnology , Pirogov Russian State Medical University (RSMU) , Moscow , Russia
| |
Collapse
|
40
|
Labonté B, Jeong YH, Parise E, Issler O, Fatma M, Engmann O, Cho KA, Neve R, Nestler EJ, Koo JW. Gadd45b mediates depressive-like role through DNA demethylation. Sci Rep 2019; 9:4615. [PMID: 30874581 PMCID: PMC6420662 DOI: 10.1038/s41598-019-40844-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
Animal studies using chronic social defeat stress (CSDS) in mice showed that brain-derived neurotrophic factor (BDNF) signaling in the mesolimbic dopamine (DA) circuit is important for the development of social aversion. However, the downstream molecular targets after BDNF release from ventral tegmental area (VTA) DA terminals are unknown. Here, we show that depressive-like behaviors induced by CSDS are mediated in part by Gadd45b downstream of BDNF signaling in the nucleus accumbens (NAc). We show that Gadd45b mRNA levels are increased in susceptible but not resilient mice. Intra-NAc infusion of BDNF or optical stimulation of VTA DA terminals in NAc enhanced Gadd45b expression levels in the NAc. Importantly, Gadd45b downregulation reversed social avoidance in susceptible mice. Together, these data suggest that Gadd45b in NAc contributes to susceptibility to social stress. In addition, we investigated the function of Gadd45b in demethylating CpG islands of representative gene targets, which have been associated with a depressive phenotype in humans and animal models. We found that Gadd45b downregulation changes DNA methylation levels in a phenotype-, gene-, and locus-specific fashion. Together, these results highlight the contribution of Gadd45b and changes in DNA methylation in mediating the effects of social stress in the mesolimbic DA circuit.
Collapse
Affiliation(s)
- Benoit Labonté
- CERVO Brain Research Centre, Department of Neuroscience and Psychiatry, Faculty of Medicine, Laval University, Québec, Canada
| | - Yun Ha Jeong
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Korea
| | - Eric Parise
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Orna Issler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mena Fatma
- CERVO Brain Research Centre, Department of Neuroscience and Psychiatry, Faculty of Medicine, Laval University, Québec, Canada
| | - Olivia Engmann
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kyung-Ah Cho
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Korea
- Department of Molecular Medicine, Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Rachael Neve
- Viral Gene Transfer Core, McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA
| | - Eric J Nestler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ja Wook Koo
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Korea.
| |
Collapse
|
41
|
Abstract
Prenatal adversity shapes child neurodevelopment and risk for later mental health problems. The quality of the early care environment can buffer some of the negative effects of prenatal adversity on child development. Retrospective studies, in adult samples, highlight epigenetic modifications as sentinel markers of the quality of the early care environment; however, comparable data from pediatric cohorts are lacking. Participants were drawn from the Maternal Adversity Vulnerability and Neurodevelopment (MAVAN) study, a longitudinal cohort with measures of infant attachment, infant development, and child mental health. Children provided buccal epithelial samples (mean age = 6.99, SD = 1.33 years, n = 226), which were used for analyses of genome-wide DNA methylation and genetic variation. We used a series of linear models to describe the association between infant attachment and (a) measures of child outcome and (b) DNA methylation across the genome. Paired genetic data was used to determine the genetic contribution to DNA methylation at attachment-associated sites. Infant attachment style was associated with infant cognitive development (Mental Development Index) and behavior (Behavior Rating Scale) assessed with the Bayley Scales of Infant Development at 36 months. Infant attachment style moderated the effects of prenatal adversity on Behavior Rating Scale scores at 36 months. Infant attachment was also significantly associated with a principal component that accounted for 11.9% of the variation in genome-wide DNA methylation. These effects were most apparent when comparing children with a secure versus a disorganized attachment style and most pronounced in females. The availability of paired genetic data revealed that DNA methylation at approximately half of all infant attachment-associated sites was best explained by considering both infant attachment and child genetic variation. This study provides further evidence that infant attachment can buffer some of the negative effects of early adversity on measures of infant behavior. We also highlight the interplay between infant attachment and child genotype in shaping variation in DNA methylation. Such findings provide preliminary evidence for a molecular signature of infant attachment and may help inform attachment-focused early intervention programs.
Collapse
|
42
|
Increase of histone acetylation in the GABAergic neurons in the rostral ventromedial medulla associated with mechanical hypersensitivity after repeated restraint stress. Brain Res Bull 2018; 142:394-402. [DOI: 10.1016/j.brainresbull.2018.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 01/31/2023]
|
43
|
Shepard RD, Gouty S, Kassis H, Berenji A, Zhu W, Cox BM, Nugent FS. Targeting histone deacetylation for recovery of maternal deprivation-induced changes in BDNF and AKAP150 expression in the VTA. Exp Neurol 2018; 309:160-168. [PMID: 30102916 DOI: 10.1016/j.expneurol.2018.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/17/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022]
Abstract
Severe early life stressors increase the probability of developing psychiatric disorders later in life through modifications in neuronal circuits controlling brain monoaminergic signaling. Our previous work demonstrated that 24 h maternal deprivation (MD) in male Sprague Dawley rats modifies dopamine (DA) signaling from the ventral tegmental area (VTA) through changes at GABAergic synapses that were reversible by in vitro histone deacetylase (HDAC) inhibition which led to restoration of the scaffold A-kinase anchoring protein (AKAP150) signaling and subsequently recovered GABAergic plasticity (Authement et al., 2015). Using a combination of in situ hybridization, Western blots and immunohistochemistry, we confirmed that MD-induced epigenetic modifications at the level of histone acetylation were associated with an upregulation of HDAC2. MD also increased Akap5 mRNA levels in the VTA. Western blot analysis of AKAP150 protein expression showed an increase in synaptic levels of AKAP150 protein in the VTA with an accompanying decrease in synaptic levels of protein kinase A (PKA). Moreover, the abundance of mature brain-derived neurotrophic factor (BDNF) protein of VTA tissues from MD rats was significantly lower than in control groups. In vivo systemic injection with a selective class I HDAC inhibitor (CI-994) was sufficient to reverse MD-induced histone hypoacetylation in the VTA for 24 h after the injection. Furthermore, HDAC inhibition normalized the levels of mBDNF and AKAP150 proteins at 24 h. Our data suggest that HDAC-mediated targeting of BDNF and AKAP-dependent local signaling within VTA could provide novel therapeutics for prevention of later-life psychopathology.
Collapse
Affiliation(s)
- Ryan D Shepard
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Shawn Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Haifa Kassis
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Aylar Berenji
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - William Zhu
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Brian M Cox
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Fereshteh S Nugent
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA.
| |
Collapse
|
44
|
Lu J, Jiao Z, Yu Y, Zhang C, He X, Li Q, Xu D, Wang H. Programming for increased expression of hippocampal GAD67 mediated the hypersensitivity of the hypothalamic-pituitary-adrenal axis in male offspring rats with prenatal ethanol exposure. Cell Death Dis 2018; 9:659. [PMID: 29855476 PMCID: PMC5981620 DOI: 10.1038/s41419-018-0663-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/28/2018] [Accepted: 04/26/2018] [Indexed: 12/15/2022]
Abstract
An imbalance of excitatory and inhibitory signals in the brain has been proposed to be one of the main pathological features of various diseases related to hypothalamic-pituitary-adrenal axis (HPAA) dysfunction. Excessive glutamate release induces neuronal excitotoxicity, while glutamic acid decarboxylase (GAD) 67 promotes the transformation of excessive glutamate to γ-aminobutyric acid (GABA). Our previous studies demonstrated that prenatal ethanol exposure (PEE) causes foetal over-exposure to maternal corticosterone and hypersensitivity of the HPAA after birth, but its intrauterine programming mechanism is unknown. In this study, PEE was shown to lead to an enhanced potential excitatory ability of the hypothalamus and hypersensitivity of the HPAA, as well as mild abnormal hippocampal morphology, demethylation of the -1019 to -691-bp region in the hippocampal GAD67 promoter and upregulation of GAD67 expression accompanied by a reduction in glutamatergic neurons and increase in GABAergic neurons in PEE male offspring. Similar changes were also found in PEE male foetal rats. Furthermore, corticosterone increased the expression of the glucocorticoid receptor (GR) and GAD67 in foetal hippocampal H19-7 cells in a concentration-dependent manner, accompanied by demethylation of the GAD67 promoter, a decrease in glutamatergic neurons and increase in GABAergic neurons. The GR inhibitor, mifepristone, reversed the effects of corticosterone on H19-7 cells. These results suggested that PEE-induced excessive corticosterone can lead to upregulation of GAD67 through epigenetic modification mediated by the GR in the male foetal hippocampus, thereby weakening the negative regulation of the HPAA by the hippocampus and increasing the potential excitatory ability of the hypothalamus. These changes persisted until after birth, resulting in hypersensitivity of the HPAA. However, gender differences were observed in the hippocampal development, morphology and GAD67 expression associated with PEE. Programming for the increased expression of hippocampal GAD67 is a potential mechanism responsible for the hypersensitivity of the HPAA in PEE male rats.
Collapse
Affiliation(s)
- Juan Lu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, Hubei Province, China.,Gansu provincial hospital of TCM Affiliated to Gansu University of Chinese Medicine, Gansu, 730050, China
| | - Zhexiao Jiao
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, Hubei Province, China
| | - Ying Yu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, Hubei Province, China.,Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chong Zhang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, Hubei Province, China
| | - Xia He
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, Hubei Province, China
| | - Qiang Li
- Gansu provincial hospital of TCM Affiliated to Gansu University of Chinese Medicine, Gansu, 730050, China
| | - Dan Xu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, Hubei Province, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, Hubei Province, China.
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, Hubei Province, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, Hubei Province, China.
| |
Collapse
|
45
|
Burns SB, Szyszkowicz JK, Luheshi GN, Lutz PE, Turecki G. Plasticity of the epigenome during early-life stress. Semin Cell Dev Biol 2018; 77:115-132. [DOI: 10.1016/j.semcdb.2017.09.033] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/08/2017] [Accepted: 09/22/2017] [Indexed: 12/22/2022]
|
46
|
Begum G, Otsu M, Ahmed U, Ahmed Z, Stevens A, Fulton D. NF-Y-dependent regulation of glutamate receptor 4 expression and cell survival in cells of the oligodendrocyte lineage. Glia 2018; 66:1896-1914. [PMID: 29704264 PMCID: PMC6220837 DOI: 10.1002/glia.23446] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 03/14/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022]
Abstract
Glutamate receptor subunit 4 (GluA4) is highly expressed by neural cells sensitive to excitotoxicity, and is the predominant subunit expressed by oligodendrocyte precursor cells (OPC) during a key period of vulnerability to hypoxic‐ischemic injury. Therefore, transcriptional networks downstream of excitotoxic GluA4 activation represent a promising area for therapeutic intervention. In this work, we identify the CCAAT binding transcription factor NF‐Yb as a novel transcriptional regulator of Gria4 (GluA4 gene), and a controller of excitotoxic death in the oligodendroglial lineage. We describe a novel regulatory region within Gria4 containing CCAAT sequences whose binding by NF‐Yb is regulated by excitotoxicity. Excitotoxicity‐induced alterations in NF‐Yb binding are associated with changes in Gria4 transcription, while knockdown of NF‐Yb alters the transcription of reporter constructs containing this regulatory region. Data from immortalized and primary OPC reveal that RNAi and pharmacological disruption of NF‐Yb alter Gria4 transcription, with the latter inducing apoptosis and influencing a set of apoptotic genes similarly regulated during excitotoxicity. These data provide the first definition of a trans‐acting mechanism regulating Gria4, and identify the NF‐Y network as a potential source of pharmacological targets for promoting OPC survival.
Collapse
Affiliation(s)
- Ghazala Begum
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Masahiro Otsu
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Usman Ahmed
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Zubair Ahmed
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Adam Stevens
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9PL, United Kingdom
| | - Daniel Fulton
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| |
Collapse
|
47
|
Kilvitis HJ, Hanson H, Schrey AW, Martin LB. Epigenetic Potential as a Mechanism of Phenotypic Plasticity in Vertebrate Range Expansions. Integr Comp Biol 2018; 57:385-395. [PMID: 28859411 DOI: 10.1093/icb/icx082] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
SYNOPSIS During range expansions, organisms are often exposed to multiple pressures, including novel enemies (i.e., predators, competitors and/or parasites) and unfamiliar or limited resources. Additionally, small propagule sizes at range edges can result in genetic founder effects and bottlenecks, which can affect phenotypic diversity and thus selection. Despite these obstacles, individuals in expanding populations often thrive at the periphery of a range, and this success may be mediated by phenotypic plasticity. Increasing evidence suggests that epigenetic mechanisms may underlie such plasticity because they allow for more rapid phenotypic responses to novel environments than are possible via the accumulation of genetic variation. Here, we review how molecular epigenetic mechanisms could facilitate plasticity in range-expanding organisms, emphasizing the roles of DNA methylation and other epigenetic marks in the physiological regulatory networks that drive whole-organism performance. We focus on the hypothalamic-pituitary-adrenal (HPA) axis, arguing that epigenetically-mediated plasticity in the regulation of glucocorticoids in particular might strongly impact range expansions. We hypothesize that novel environments release and/or select for epigenetic potential in HPA variation and hence organismal performance and ultimately fitness.
Collapse
Affiliation(s)
- Holly J Kilvitis
- Department of Integrative Biology, University of South Florida, Tampa, FL 33620, USA
| | - Haley Hanson
- Department of Integrative Biology, University of South Florida, Tampa, FL 33620, USA
| | - Aaron W Schrey
- Department of Biology, Armstrong State University, Savannah, GA 31419, USA
| | - Lynn B Martin
- Department of Integrative Biology, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
48
|
Zhu C, Liang M, Li Y, Feng X, Hong J, Zhou R. Involvement of Epigenetic Modifications of GABAergic Interneurons in Basolateral Amygdala in Anxiety-like Phenotype of Prenatally Stressed Mice. Int J Neuropsychopharmacol 2018; 21:570-581. [PMID: 29471396 PMCID: PMC6007574 DOI: 10.1093/ijnp/pyy006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Prenatal stress is considered a risk factor for anxiety disorder. Downregulation in the expression of GABAergic gene, that is, glutamic acid decarboxylase 67, associated with DNA methyltransferase overexpression in GABAergic neurons has been regarded as a characteristic component of anxiety disorder. Prenatal stress has an adverse effect on the development of the basolateral amygdala, which is a key region in anxiety regulation. The aim of this study is to analyze the possibility of epigenetic alterations of GABAergic neurons in the basolateral amygdala participating in prenatal stress-induced anxiety. METHODS Behavioral tests were used to explore the prenatal stress-induced anxiety behaviors of female adult mice. Real-time RT-PCR, western blot, chromatin immunoprecipitation, and electrophysiological analysis were employed to detect epigenetic changes of GABAergic system in the basolateral amygdala. RESULTS Prenatal stress mice developed an anxiety-like phenotype accompanied by a significant increase of DNA methyltransferase 1 and a reduced expression of glutamic acid decarboxylase 67 in the basolateral amygdala. Prenatal stress mice also showed the increased binding of DNA methyltransferase 1 and methyl CpG binding protein 2 to glutamic acid decarboxylase 67 promoter region. The decrease of glutamic acid decarboxylase 67 transcript was paralleled by an enrichment of 5-methylcytosine in glutamic acid decarboxylase 67 promoter regions. Electrophysiological study revealed the increase of postsynaptic neuronal excitability in the cortical-basolateral amygdala synaptic transmission of prenatal stress mice. 5-Aza-deoxycytidine treatment restored the increased synaptic transmission and anxiety-like behaviors in prenatal stress mice via improving GABAergic system. CONCLUSION The above results suggest that DNA epigenetic modifications of GABAergic interneurons in the basolateral amygdala participate in the etiology of anxiety-like phenotype in prenatal stress mice.
Collapse
Affiliation(s)
- Chunting Zhu
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Min Liang
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Yingchun Li
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Xuejiao Feng
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Juan Hong
- Department of Physiology, Nanjing Medical University, Jiangsu, China
| | - Rong Zhou
- Department of Physiology, Nanjing Medical University, Jiangsu, China,Correspondence: Rong Zhou, PhD, Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing City, Jiangsu Province, China 211166 ()
| |
Collapse
|
49
|
Epigenetics and Early Life Adversity: Current Evidence and Considerations for Epigenetic Studies in the Context of Child Maltreatment. THE BIOLOGY OF EARLY LIFE STRESS 2018. [DOI: 10.1007/978-3-319-72589-5_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
50
|
Barnett Burns S, Almeida D, Turecki G. The Epigenetics of Early Life Adversity: Current Limitations and Possible Solutions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:343-425. [DOI: 10.1016/bs.pmbts.2018.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|