1
|
Shulhai AM, Munerati A, Menzella M, Palanza P, Esposito S, Street ME. Insights into pubertal development: a narrative review on the role of epigenetics. J Endocrinol Invest 2025; 48:817-830. [PMID: 39704935 PMCID: PMC11950117 DOI: 10.1007/s40618-024-02513-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE Puberty is a key phase of growth and development, characterized by psychophysical transformations. It is driven by a combination of genetic, hormonal, and environmental variables. Epigenetic mechanisms, including histone post-translational modifications and chromatin remodeling, microRNAs, and DNA methylation, play important roles in orchestrating the developmental processes. We describe environmental factors that may interact with genetics, and factors influencing puberty onset, focusing in particular on epigenetic mechanisms that can help understand the timing and variations that lead to precocious or delayed puberty. METHODS We conducted a narrative review of associations between puberty and epigenetic mechanisms through a comprehensive search of PubMed, Scopus, and Web of Science databases. RESULTS The chromatin landscape of genes as KISS1 has revealed dynamic changes in histone modifications as puberty approaches, influencing the stimulation or inhibition of gene expression critical for reproductive maturation. MiRNAs regulate gene expression, whereas DNA methylation affects activation or repression of gene transcription of genes involved in pubertal timing. Moreover, studies in animal models have provided insights into the role of DNA methylation and miRNAs in brain sexual differentiation, highlighting the active involvement of epigenetic mechanisms in shaping sexually dimorphic brain structures. CONCLUSION This review highlights the importance of understanding the complex interplay between epigenetic regulation and pubertal development, which can lead to new therapeutic options and shed light on the fundamental processes driving reproductive maturation.
Collapse
Affiliation(s)
- Anna-Mariia Shulhai
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, University of Parma, Parma, 43126, Italy
- Department of Pediatrics №2, Ivan Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Anna Munerati
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, University of Parma, Parma, 43126, Italy
| | - Marialaura Menzella
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, University of Parma, Parma, 43126, Italy
| | - Paola Palanza
- Unit of Neuroscience, Department of Medicine and Surgery, University of Parma, Parma, 43125, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, University of Parma, Parma, 43126, Italy
| | - Maria Elisabeth Street
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, University of Parma, Parma, 43126, Italy.
| |
Collapse
|
2
|
He X, Ma Q, Liu J, Lei P, Peng H, Lu W, Liu Y, Zhan X, Yan B, Ma X, Yang J. Investigating the shared genetic architecture between schizophrenia and sex hormone traits. Transl Psychiatry 2025; 15:83. [PMID: 40097391 PMCID: PMC11914697 DOI: 10.1038/s41398-025-03305-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 02/17/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Sex hormones are involved in schizophrenia pathogenesis; however, their direction and genetic overlap remain unknown. By leveraging summary statistics from large-scale genome-wide association studies, we quantified the shared genetic architecture between schizophrenia and four sex hormone traits. Linkage disequilibrium score regression and bivariate causal mixture modeling strategies showed significant positive correlations between sex hormone-binding globulin (SHBG), total testosterone, and schizophrenia, while bioavailable testosterone and schizophrenia were negatively correlated. Estradiol showed a weak positive correlation with schizophrenia, with little polygenic overlap. The conjunctional false discovery rate method identified 303 lead single-nucleotide polymorphisms (SNPs) in jointly shared genomic loci between schizophrenia and SHBG, with 130, 52, and 9 SNPs shared between schizophrenia and total testosterone, bioavailable testosterone, and estradiol, respectively. Functional annotation suggests that mitotic sister chromatid segregation and N-glycan biosynthesis may be involved in common mechanisms underlying sex hormone regulation and schizophrenia onset. In conclusion, this study clarified the inherent relationships between schizophrenia and sex hormone traits, highlighted the roles of mitotic sister chromatid segregation and N-glycan biosynthesis in the pathogenesis of schizophrenia, and delivered potential targets for further validation.
Collapse
Affiliation(s)
- Xiaoyan He
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qingyan Ma
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pu Lei
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huan Peng
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wen Lu
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yixin Liu
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xianyan Zhan
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bin Yan
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiancang Ma
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Jian Yang
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
3
|
Du S, Shang G, Tian X, Liu Z, Yang Y, Niu H, Bian J, Wu Y, Ma J. Effects of DNA Methylation of HPA-Axis Genes of F1 Juvenile Induced by Maternal Density Stress on Behavior and Immune Traits in Root Voles ( Microtus oeconomus)-A Field Experiment. Animals (Basel) 2024; 14:2467. [PMID: 39272253 PMCID: PMC11393846 DOI: 10.3390/ani14172467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
The literature shows that maternal stress can influence behavior and immune function in F1. Yet, most studies on these are from the laboratory, and replicated studies on the mechanisms by which maternal stress drives individual characteristics are still not fully understood in wild animals. We manipulated high- and low-density parental population density using large-scale field enclosures and examined behavior and immune traits. Within the field enclosures, we assessed anti-keyhole limpet hemocyanin immunoglobulin G (anti-KLH IgG) level, phytohemagglutinin (PHA) responses, hematology, cytokines, the depressive and anxiety-like behaviors and prevalence and intensity of coccidial infection. We then collected brain tissue from juvenile voles born at high or low density, quantified mRNA and protein expression of corticotropin-releasing hormone (CRH) and glucocorticoid receptor gene (NR3C1) and measured DNA methylation at CpG sites in a region that was highly conserved with the prairie vole CRH and NR3C1 promoter. At high density, we found that the F1 had a lower DNA methylation level of CRH and a higher DNA methylation level of NR3C1, which resulted in an increase in the expression levels of the CRH mRNA and protein expression and further reduced the expression levels of the NR3C1 mRNA and protein expression, and ultimately led to have delayed responses to acute immobilization stress. Juvenile voles born at high density also reduced anti-KLH IgG levels and PHA responses, increased cytokines, and depressive and anxiety-like behaviors, and the effects further led to higher coccidial infection. From the perspective of population density inducing the changes in behavior and immunity at the brain level, our results showed a physiological epigenetic mechanism for population self-regulation in voles. Our results indicate that altering the prenatal intrinsic stress environment can fundamentally impact behavior and immunity by DNA methylation of HPA-axis genes and can further drive population fluctuations in wild animals.
Collapse
Affiliation(s)
- Shouyang Du
- Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang 453003, China
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Guozhen Shang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China
- Qinghai Key Laboratory of Animal Ecological Genomics, Xining 810001, China
| | - Xin Tian
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Zihan Liu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Yanbin Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450007, China
| | - Hongxing Niu
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Jianghui Bian
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810001, China
- Qinghai Key Laboratory of Animal Ecological Genomics, Xining 810001, China
| | - Yan Wu
- School of Life and Environment Sciences, Hangzhou Normal University, Hangzhou 310012, China
| | - Jinyou Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| |
Collapse
|
4
|
Cattaneo A, Begni V, Zonca V, Riva MA. Early life adversities, psychopathologies and novel pharmacological strategies. Pharmacol Ther 2024; 260:108686. [PMID: 38969307 DOI: 10.1016/j.pharmthera.2024.108686] [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: 02/23/2024] [Revised: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Exposure to adversities during early life stages (early life adversities - ELA), ranging from pregnancy to adolescence, represents a major risk factor for the vulnerability to mental disorders. Hence, it is important to understand the molecular and functional underpinning of such relationship, in order to develop strategies aimed at reducing the psychopathologic burden associated with ELA, which may eventually lead to a significant improvement in clinical practice. In this review, we will initially recapitulate clinical and preclinical evidence supporting the link between ELA and psychopathology and we will primarily discuss the main biological mechanisms that have been described as potential mediators of the effects of ELA on the psychopathologic risk, including the role for genetic factors as well as sex differences. The knowledge emerging from these studies may be instrumental for the development of novel therapeutic strategies aimed not only at correcting the deficits that emerge from ELA exposure, but also in preventing the manifestation of a full-blown psychopathologic condition. With this respect, we will specifically focus on adolescence as a key time frame for disease onset as well as for early therapeutic intervention. We believe that incorporating clinical and preclinical research data in the context of early life adversities can be instrumental to elucidate the mechanisms contributing to the risk for psychopathology or that may promote resilience. This will ultimately allow the identification of 'at risk' individuals who may benefit from specific forms of interventions that, by interfering with disease trajectories, could result in more benign clinical outcomes.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Valentina Zonca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
5
|
Saavedra LPJ, Piovan S, Moreira VM, Gonçalves GD, Ferreira ARO, Ribeiro MVG, Peres MNC, Almeida DL, Raposo SR, da Silva MC, Barbosa LF, de Freitas Mathias PC. Epigenetic programming for obesity and noncommunicable disease: From womb to tomb. Rev Endocr Metab Disord 2024; 25:309-324. [PMID: 38040983 DOI: 10.1007/s11154-023-09854-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Several epidemiological, clinical and experimental studies in recent decades have shown the relationship between exposure to stressors during development and health outcomes later in life. The characterization of these susceptible phases, such as preconception, gestation, lactation and adolescence, and the understanding of factors that influence the risk of an adult individual for developing obesity, metabolic and cardiovascular diseases, is the focus of the DOHaD (Developmental Origins of Health and Disease) research line. In this sense, advancements in molecular biology techniques have contributed significantly to the understanding of the mechanisms underlying the observed phenotypes, their morphological and physiological alterations, having as a main driving factor the epigenetic modifications and their consequent modulation of gene expression. The present narrative review aimed to characterize the different susceptible phases of development and associated epigenetic modifications, and their implication in the development of non-communicable diseases. Additionally, we provide useful insights into interventions during development to counteract or prevent long-term programming for disease susceptibility.
Collapse
Affiliation(s)
- Lucas Paulo Jacinto Saavedra
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Silvano Piovan
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Veridiana Mota Moreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Gessica Dutra Gonçalves
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maria Natália Chimirri Peres
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Douglas Lopes Almeida
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Scarlett Rodrigues Raposo
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Mariane Carneiro da Silva
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Letícia Ferreira Barbosa
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Paulo Cezar de Freitas Mathias
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil.
| |
Collapse
|
6
|
Kundakovic M, Tickerhoof M. Epigenetic mechanisms underlying sex differences in the brain and behavior. Trends Neurosci 2024; 47:18-35. [PMID: 37968206 PMCID: PMC10841872 DOI: 10.1016/j.tins.2023.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/21/2023] [Accepted: 09/26/2023] [Indexed: 11/17/2023]
Abstract
Sex differences are found across brain regions, behaviors, and brain diseases. Sexual differentiation of the brain is initiated prenatally but it continues throughout life, as a result of the interaction of three major factors: gonadal hormones, sex chromosomes, and the environment. These factors are thought to act, in part, via epigenetic mechanisms which control chromatin and transcriptional states in brain cells. In this review, we discuss evidence that epigenetic mechanisms underlie sex-specific neurobehavioral changes during critical organizational periods, across the estrous cycle, and in response to diverse environments throughout life. We further identify future directions for the field that will provide novel mechanistic insights into brain sex differences, inform brain disease treatments and women's brain health in particular, and apply to people across genders.
Collapse
Affiliation(s)
- Marija Kundakovic
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA.
| | - Maria Tickerhoof
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| |
Collapse
|
7
|
Zinchuk MS, Druzhkova TA, Popova SB, Zhanina MY, Guekht AB, Gulyaeva NV. Early Adverse Family Experiences and Elevated Adrenocorticotropic Hormone Predict Non-Suicidal Self-Injury in Females with Non-Psychotic Mental Disorders and Suicidal Ideation. Biomedicines 2023; 11:3181. [PMID: 38137402 PMCID: PMC10740784 DOI: 10.3390/biomedicines11123181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Nonsuicidal self-injurious behavior (NSSI), prevalent in patients with non-psychotic mental disorders (NPMD), is associated with numerous adverse outcomes. Despite active research into the clinical and psychological aspects of NSSI, the underlying biological mechanisms remain obscure. Early adverse experiences are believed to induce long-lasting changes in neuroendocrine mechanisms of stress control playing a key role in NSSI development. The aim of the study was to evaluate parameters potentially predicting development of NSSI in female patients with NPMD and suicidal ideation. Eighty female patients over 18 years with NPMD and suicidal ideation (40 with and 40 without NSSI) and 48 age matching women without evidence of mental illness (healthy controls) were enrolled. Diagnostic interviews and self-report measures were used to assess childhood maltreatment, presence, frequency, and characteristics of suicidal and self-injurious thoughts and behaviors, the Beck Depression Inventory scale to assess severity of depression. Hypothalamic-pituitary-adrenal axis markers, hormones, and neurotrophic factors were measured in blood serum. The likelihood of developing NSSI in patients with NPMD and suicidal ideation was associated with early adverse family history and elevated adrenocorticotropic hormone levels. Dysregulation of hypothalamic-pituitary-adrenal axis as a result of early chronic stress experiences may represent critical biological mechanism promoting the development of NSSI behaviors in patients with NPMD.
Collapse
Affiliation(s)
- Mikhail S. Zinchuk
- Moscow Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia; (M.S.Z.); (S.B.P.); (M.Y.Z.)
| | - Tatiana A. Druzhkova
- Moscow Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia; (M.S.Z.); (S.B.P.); (M.Y.Z.)
| | - Sofya B. Popova
- Moscow Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia; (M.S.Z.); (S.B.P.); (M.Y.Z.)
| | - Marina Y. Zhanina
- Moscow Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia; (M.S.Z.); (S.B.P.); (M.Y.Z.)
- Department of Functional Biochemistry of Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | - Alla B. Guekht
- Moscow Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia; (M.S.Z.); (S.B.P.); (M.Y.Z.)
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, 119049 Moscow, Russia
| | - Natalia V. Gulyaeva
- Moscow Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia; (M.S.Z.); (S.B.P.); (M.Y.Z.)
- Department of Functional Biochemistry of Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| |
Collapse
|
8
|
Murlanova K, Pletnikov MV. Modeling psychotic disorders: Environment x environment interaction. Neurosci Biobehav Rev 2023; 152:105310. [PMID: 37437753 DOI: 10.1016/j.neubiorev.2023.105310] [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: 12/14/2022] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Schizophrenia is a major psychotic disorder with multifactorial etiology that includes interactions between genetic vulnerability and environmental risk factors. In addition, interplay of multiple environmental adversities affects neurodevelopment and may increase the individual risk of developing schizophrenia. Consistent with the two-hit hypothesis of schizophrenia, we review rodent models that combine maternal immune activation as the first hit with other adverse environmental exposures as the second hit. We discuss the strengths and pitfalls of the current animal models of environment x environment interplay and propose some future directions to advance the field.
Collapse
Affiliation(s)
- Kateryna Murlanova
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
9
|
Kovacs-Balint ZA, Raper J, Richardson R, Gopakumar A, Kettimuthu KP, Higgins M, Feczko E, Earl E, Ethun KF, Li L, Styner M, Fair D, Bachevalier J, Sanchez MM. The role of puberty on physical and brain development: A longitudinal study in male Rhesus Macaques. Dev Cogn Neurosci 2023; 60:101237. [PMID: 37031512 PMCID: PMC10114189 DOI: 10.1016/j.dcn.2023.101237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 02/20/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
This study examined the role of male pubertal maturation on physical growth and development of neurocircuits that regulate stress, emotional and cognitive control using a translational nonhuman primate model. We collected longitudinal data from male macaques between pre- and peri-puberty, including measures of physical growth, pubertal maturation (testicular volume, blood testosterone -T- concentrations) and brain structural and resting-state functional MRI scans to examine developmental changes in amygdala (AMY), hippocampus (HIPPO), prefrontal cortex (PFC), as well as functional connectivity (FC) between those regions. Physical growth and pubertal measures increased from pre- to peri-puberty. The indexes of pubertal maturation -testicular size and T- were correlated at peri-puberty, but not at pre-puberty (23 months). Our findings also showed ICV, AMY, HIPPO and total PFC volumetric growth, but with region-specific changes in PFC. Surprisingly, FC in these neural circuits only showed developmental changes from pre- to peri-puberty for HIPPO-orbitofrontal FC. Finally, testicular size was a better predictor of brain structural maturation than T levels -suggesting gonadal hormones-independent mechanisms-, whereas T was a strong predictor of functional connectivity development. We expect that these neural circuits will show more drastic pubertal-dependent maturation, including stronger associations with pubertal measures later, during and after male puberty.
Collapse
Affiliation(s)
- Z A Kovacs-Balint
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.
| | - J Raper
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Dept. of Pediatrics, Emory University, Atlanta, GA 30322, USA
| | - R Richardson
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - A Gopakumar
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - K P Kettimuthu
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - M Higgins
- Office of Nursing Research, Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA 30322, USA
| | - E Feczko
- Dept. of Pediatrics, University of Minnesota, Minneapolis, MN 55414, USA; Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN 55414, USA
| | - E Earl
- Dept. of Behavioral Neuroscience, Oregon Health & Sciences University, Portland, OR 97239, USA
| | - K F Ethun
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - L Li
- Dept. of Pediatrics, Emory University, Atlanta, GA 30322, USA; Marcus Autism Center; Children's Healthcare of Atlanta, GA, USA
| | - M Styner
- Dept. of Psychiatry, University of North Carolina, Chapel Hill, NC 27514, USA
| | - D Fair
- Dept. of Pediatrics, University of Minnesota, Minneapolis, MN 55414, USA; Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN 55414, USA
| | - J Bachevalier
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - M M Sanchez
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Dept. of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Impact of Nut Consumption on Cognition across the Lifespan. Nutrients 2023; 15:nu15041000. [PMID: 36839359 PMCID: PMC9965316 DOI: 10.3390/nu15041000] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Cognitive health is a life-long concern affected by modifiable risk factors, including lifestyle choices, such as dietary intake, with serious implications for quality of life, morbidity, and mortality worldwide. In addition, nuts are a nutrient-dense food that contain a number of potentially neuroprotective components, including monounsaturated and polyunsaturated fatty acids, fiber, B-vitamins, non-sodium minerals, and highly bioactive polyphenols. However, increased nut consumption relates to a lower cardiovascular risk and a lower burden of cardiovascular risk factors that are shared with neurodegenerative disorders, which is why nuts have been hypothesized to be beneficial for brain health. The present narrative review discusses up-to-date epidemiological, clinical trial, and mechanistic evidence of the effect of exposure to nuts on cognitive performance. While limited and inconclusive, available evidence suggests a possible role for nuts in the maintenance of cognitive health and prevention of cognitive decline in individuals across the lifespan, particularly in older adults and those at higher risk. Walnuts, as a rich source of the plant-based polyunsaturated omega-3 fatty acid alpha-linolenic acid, are the nut type most promising for cognitive health. Given the limited definitive evidence available to date, especially regarding cognitive health biomarkers and hard outcomes, future studies are needed to better elucidate the impact of nuts on the maintenance of cognitive health, as well as the prevention and management of cognitive decline and dementia, including Alzheimer disease.
Collapse
|
11
|
Perera BPU, Morgan RK, Polemi KM, Sala-Hamrick KE, Svoboda LK, Dolinoy DC. PIWI-Interacting RNA (piRNA) and Epigenetic Editing in Environmental Health Sciences. Curr Environ Health Rep 2022; 9:650-660. [PMID: 35917009 DOI: 10.1007/s40572-022-00372-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW: The epigenome modulates gene expression in response to environmental stimuli. Modifications to the epigenome are potentially reversible, making them a promising therapeutic approach to mitigate environmental exposure effects on human health. This review details currently available genome and epigenome editing technologies and highlights ncRNA, including piRNA, as potential tools for targeted epigenome editing. RECENT FINDINGS: Zinc finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN), and clustered regularly interspaced short palindromic repeats (CRISPR) associated nuclease (CRISPR/Cas) research has significantly advanced genome editing technology, with broad promise in genetic research and targeted therapies. Initial epigenome-directed therapies relied on global modification and suffered from limited specificity. Adapted from current genome editing tools, zinc finger protein (ZFP), TALE, and CRISPR/nuclease-deactivated Cas (dCas) systems now confer locus-specific epigenome editing, with promising applicability in the field of environmental health sciences. However, high incidence of off-target effects and time taken for screening limit their use. FUTURE DEVELOPMENT: ncRNA serve as a versatile biomarker with well-characterized regulatory mechanisms that can easily be adapted to edit the epigenome. For instance, the transposon silencing mechanism of germline PIWI-interacting RNAs (piRNA) could be engineered to specifically methylate a given gene, overcoming pitfalls of current global modifiers. Future developments in epigenome editing technologies will inform risk assessment through mechanistic investigation and serve as potential modes of intervention to mitigate environmentally induced adverse health outcomes later in life.
Collapse
Affiliation(s)
- Bambarendage P U Perera
- School of Public Health, Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA.
| | - Rachel K Morgan
- School of Public Health, Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Katelyn M Polemi
- School of Public Health, Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Kimmie E Sala-Hamrick
- School of Public Health, Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Laurie K Svoboda
- School of Public Health, Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Dana C Dolinoy
- School of Public Health, Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
- School of Public Health, Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
12
|
Saengkaew T, Howard SR. Genetics of pubertal delay. Clin Endocrinol (Oxf) 2022; 97:473-482. [PMID: 34617615 PMCID: PMC9543006 DOI: 10.1111/cen.14606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
The timing of pubertal development is strongly influenced by the genetic background, and clinical presentations of delayed puberty are often found within families with clear patterns of inheritance. The discovery of the underlying genetic regulators of such conditions, in recent years through next generation sequencing, has advanced the understanding of the pathogenesis of disorders of pubertal timing and the potential for genetic testing to assist diagnosis for patients with these conditions. This review covers the significant advances in the understanding of the biological mechanisms of delayed puberty that have occurred in the last two decades.
Collapse
Affiliation(s)
- Tansit Saengkaew
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Endocrinology Unit, Department of Paediatrics, Faculty of MedicinePrince of Songkla UniversitySongkhlaThailand
| | - Sasha R. Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
13
|
Murlanova K, Hasegawa Y, Kamiya A, Pletnikov MV. Cannabis effects on the adolescent brain. CANNABIS AND THE DEVELOPING BRAIN 2022:283-330. [DOI: 10.1016/b978-0-12-823490-7.00007-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Rainville JR, Lipuma T, Hodes GE. Translating the Transcriptome: Sex Differences in the Mechanisms of Depression and Stress, Revisited. Biol Psychiatry 2022; 91:25-35. [PMID: 33865609 PMCID: PMC10197090 DOI: 10.1016/j.biopsych.2021.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 12/28/2022]
Abstract
The past decade has produced a plethora of studies examining sex differences in the transcriptional profiles of stress and mood disorders. As we move forward from accepting the existence of extensive molecular sex differences in the brain to exploring the purpose of these sex differences, our approach must become more systemic and less reductionist. Earlier studies have examined specific brain regions and/or cell types. To use this knowledge to develop the next generation of personalized medicine, we need to comprehend how transcriptional changes across the brain and/or the body relate to each other. We provide an overview of the relationships between baseline and depression/stress-related transcriptional sex differences and explore contributions of preclinically identified mechanisms and their impacts on behavior.
Collapse
Affiliation(s)
- Jennifer R Rainville
- Department of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia
| | - Timothy Lipuma
- Department of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia
| | - Georgia E Hodes
- Department of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia.
| |
Collapse
|
15
|
Sex differences in the genetic regulation of the blood transcriptome response to glucocorticoid receptor activation. Transl Psychiatry 2021; 11:632. [PMID: 34903727 PMCID: PMC8669026 DOI: 10.1038/s41398-021-01756-2] [Citation(s) in RCA: 13] [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: 07/05/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
Substantial sex differences have been reported in the physiological response to stress at multiple levels, including the release of the stress hormone, cortisol. Here, we explore the genomic variants in 93 females and 196 males regulating the initial transcriptional response to cortisol via glucocorticoid receptor (GR) activation. Gene expression levels in peripheral blood were obtained before and after GR-stimulation with the selective GR agonist dexamethasone to identify differential expression following GR-activation. Sex stratified analyses revealed that while the transcripts responsive to GR-stimulation were mostly overlapping between males and females, the quantitative trait loci (eQTLs) regulation differential transcription to GR-stimulation was distinct. Sex-stratified eQTL SNPs (eSNPs) were located in different functional genomic elements and sex-stratified transcripts were enriched within postmortem brain transcriptional profiles associated with Major Depressive Disorder (MDD) specifically in males and females in the cingulate cortex. Female eSNPs were enriched among SNPs linked to MDD in genome-wide association studies. Finally, transcriptional sensitive genetic profile scores derived from sex-stratified eSNPS regulating differential transcription to GR-stimulation were predictive of depression status and depressive symptoms in a sex-concordant manner in a child and adolescent cohort (n = 584). These results suggest the potential of eQTLs regulating differential transcription to GR-stimulation as biomarkers of sex-specific biological risk for stress-related psychiatric disorders.
Collapse
|
16
|
Bilecki W, Wawrzczak-Bargieła A, Majcher-Maślanka I, Chmelova M, Maćkowiak M. Inhibition of BET Proteins during Adolescence Affects Prefrontal Cortical Development: Relevance to Schizophrenia. Int J Mol Sci 2021; 22:ijms22168710. [PMID: 34445411 PMCID: PMC8395847 DOI: 10.3390/ijms22168710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 01/10/2023] Open
Abstract
Background: The present study investigated the role of proteins from the bromodomain and extra-terminal (BET) family in schizophrenia-like abnormalities in a neurodevelopmental model of schizophrenia induced by prenatal methylazoxymethanol (MAM) administration (MAM-E17). Methods: An inhibitor of BET proteins, JQ1, was administered during adolescence on postnatal days (P) 23–P29, and behavioural responses (sensorimotor gating, recognition memory) and prefrontal cortical (mPFC) function (long-term potentiation (LTP), molecular and proteomic analyses) studies were performed in adult males and females. Results: Deficits in sensorimotor gating and recognition memory were observed only in MAM-treated males. However, adolescent JQ1 treatment affected animals of both sexes in the control but not MAM-treated groups and reduced behavioural responses in both sexes. An electrophysiological study showed LTP impairments only in male MAM-treated animals, and JQ1 did not affect LTP in the mPFC. In contrast, MAM did not affect activity-dependent gene expression, but JQ1 altered gene expression in both sexes. A proteomic study revealed alterations in MAM-treated groups mainly in males, while JQ1 affected both sexes. Conclusions: MAM-induced schizophrenia-like abnormalities were observed only in males, while adolescent JQ1 treatment affected memory recognition and altered the molecular and proteomic landscape in the mPFC of both sexes. Thus, transient adolescent inhibition of the BET family might prompt permanent alterations in the mPFC.
Collapse
|
17
|
Brocato E, Wolstenholme JT. Neuroepigenetic consequences of adolescent ethanol exposure. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:45-84. [PMID: 34696879 DOI: 10.1016/bs.irn.2021.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Adolescence is a critical developmental period characterized by ongoing brain maturation processes including myelination and synaptic pruning. Adolescents experience heightened reward sensitivity, sensation seeking, impulsivity, and diminished inhibitory self-control, which contribute to increased participation in risky behaviors, including the initiation of alcohol use. Ethanol exposure in adolescence alters memory and cognition, anxiety-like behavior, and ethanol sensitivity as well as brain myelination and dendritic spine morphology, with effects lasting into adulthood. Emerging evidence suggests that epigenetic modifications may explain these lasting effects. Focusing on the amygdala, prefrontal cortex and hippocampus, we review studies investigating the epigenetic consequences of adolescent ethanol exposure. Ethanol metabolism globally increases donor substrates for histone acetylation and histone and DNA methylation, and this chapter discusses how this can further impact epigenetic programming of the adolescent brain. Elucidation of the mechanisms through which ethanol can alter the epigenetic code at specific transcripts may provide therapeutic targets for intervention.
Collapse
Affiliation(s)
- Emily Brocato
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States; VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
18
|
Carpenter MD, Manners MT, Heller EA, Blendy JA. Adolescent oxycodone exposure inhibits withdrawal-induced expression of genes associated with the dopamine transmission. Addict Biol 2021; 26:e12994. [PMID: 33325096 DOI: 10.1111/adb.12994] [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] [Received: 05/12/2020] [Revised: 10/09/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022]
Abstract
Prescription opioid misuse is a major public health concern among children and adolescents in the United States. Opioids are the most commonly abused drugs and are the fastest growing drug problem among adolescents. In humans and animals, adolescence is a particularly sensitive period associated with an increased response to drugs of abuse. Our previous studies indicate that oxycodone exposure during adolescence increases morphine reward in adulthood. How early drug exposure mediates long-term changes in the brain and behavior is not known, but epigenetic regulation is a likely mechanism. To address this question, we exposed mice to oxycodone or saline during adolescence and examined epigenetic modifications at genes associated with dopamine activity during adulthood at early and late withdrawal, in the ventral tegmental area (VTA). We then compared these with alterations in the VTA of adult-treated mice following an equivalent duration of exposure and withdrawal to determine if the effects of oxycodone are age dependent. We observed persistence of adolescent-like gene expression following adolescent oxycodone exposure relative to age-matched saline exposed controls, although dopamine-related gene expression was transiently activated at 1 day of withdrawal. Following prolonged withdrawal enrichment of the repressive histone mark, H3K27me3, was maintained, consistent with inhibition of gene regulation following adolescent exposure. By contrast, mice exposed to oxycodone as adults showed loss of the repressive mark and increased gene expression following 28 days of withdrawal following oxycodone exposure. Together, our findings provide evidence that adolescent oxycodone exposure has long-term epigenetic consequences in VTA of the developing brain.
Collapse
Affiliation(s)
- Marco D. Carpenter
- Department of Systems Pharmacology and Translational Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Institute for Translational Medicine and Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Penn Epigenetics Institute, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Melissa T. Manners
- Department of Systems Pharmacology and Translational Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Department of Biological Sciences University of the Sciences Philadelphia Pennsylvania USA
| | - Elizabeth A. Heller
- Department of Systems Pharmacology and Translational Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Institute for Translational Medicine and Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Penn Epigenetics Institute, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Julie A. Blendy
- Department of Systems Pharmacology and Translational Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Institute for Translational Medicine and Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
| |
Collapse
|
19
|
Bacon ER, Brinton RD. Epigenetics of the developing and aging brain: Mechanisms that regulate onset and outcomes of brain reorganization. Neurosci Biobehav Rev 2021; 125:503-516. [PMID: 33657435 DOI: 10.1016/j.neubiorev.2021.02.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Brain development is a life-long process that encompasses several critical periods of transition, during which significant cognitive changes occur. Embryonic development, puberty, and reproductive senescence are all periods of transition that are hypersensitive to environmental factors. Rather than isolated episodes, each transition builds upon the last and is influenced by consequential changes that occur in the transition before it. Epigenetic marks, such as DNA methylation and histone modifications, provide mechanisms by which early events can influence development, cognition, and health outcomes. For example, parental environment influences imprinting patterns in gamete cells, which ultimately impacts gene expression in the embryo which may result in hypersensitivity to poor maternal nutrition during pregnancy, raising the risks for cognitive impairment later in life. This review explores how epigenetics induce and regulate critical periods, and also discusses how early environmental interactions prime a system towards a particular health outcome and influence susceptibility to disease or cognitive impairment throughout life.
Collapse
Affiliation(s)
- Eliza R Bacon
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA; The Center for Precision Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Roberta Diaz Brinton
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA; Center for Innovation in Brain Science, School of Medicine, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
20
|
How do we improve adolescent diet and physical activity in India and sub-Saharan Africa? Findings from the Transforming Adolescent Lives through Nutrition (TALENT) consortium. Public Health Nutr 2020; 24:5309-5317. [PMID: 33111660 DOI: 10.1017/s1368980020002244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Adolescent diet, physical activity and nutritional status are generally known to be sub-optimal. This is an introduction to a special issue of papers devoted to exploring factors affecting diet and physical activity in adolescents, including food insecure and vulnerable groups. SETTING Eight settings including urban, peri-urban and rural across sites from five different low- and middle-income countries. DESIGN Focus groups with adolescents and caregivers carried out by trained researchers. RESULTS Our results show that adolescents, even in poor settings, know about healthy diet and lifestyles. They want to have energy, feel happy, look good and live longer, but their desire for autonomy, a need to 'belong' in their peer group, plus vulnerability to marketing exploiting their aspirations, leads them to make unhealthy choices. They describe significant gender, culture and context-specific barriers. For example, urban adolescents had easy access to energy dense, unhealthy foods bought outside the home, whereas junk foods were only beginning to permeate rural sites. Among adolescents in Indian sites, pressure to excel in exams meant that academic studies were squeezing out physical activity time. CONCLUSIONS Interventions to improve adolescents' diets and physical activity levels must therefore address structural and environmental issues and influences in their homes and schools, since it is clear that their food and activity choices are the product of an interacting complex of factors. In the next phase of work, the Transforming Adolescent Lives through Nutrition consortium will employ groups of adolescents, caregivers and local stakeholders in each site to develop interventions to improve adolescent nutritional status.
Collapse
|
21
|
Creighton SD, Stefanelli G, Reda A, Zovkic IB. Epigenetic Mechanisms of Learning and Memory: Implications for Aging. Int J Mol Sci 2020; 21:E6918. [PMID: 32967185 PMCID: PMC7554829 DOI: 10.3390/ijms21186918] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
The neuronal epigenome is highly sensitive to external events and its function is vital for producing stable behavioral outcomes, such as the formation of long-lasting memories. The importance of epigenetic regulation in memory is now well established and growing evidence points to altered epigenome function in the aging brain as a contributing factor to age-related memory decline. In this review, we first summarize the typical role of epigenetic factors in memory processing in a healthy young brain, then discuss the aspects of this system that are altered with aging. There is general agreement that many epigenetic marks are modified with aging, but there are still substantial inconsistencies in the precise nature of these changes and their link with memory decline. Here, we discuss the potential source of age-related changes in the epigenome and their implications for therapeutic intervention in age-related cognitive decline.
Collapse
Affiliation(s)
- Samantha D. Creighton
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (S.D.C.); (G.S.)
| | - Gilda Stefanelli
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (S.D.C.); (G.S.)
| | - Anas Reda
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S, Canada;
| | - Iva B. Zovkic
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (S.D.C.); (G.S.)
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S, Canada;
| |
Collapse
|
22
|
Dulman RS, Wandling GM, Pandey SC. Epigenetic mechanisms underlying pathobiology of alcohol use disorder. CURRENT PATHOBIOLOGY REPORTS 2020; 8:61-73. [PMID: 33747641 DOI: 10.1007/s40139-020-00210-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Purpose of review Chronic alcohol use is a worldwide problem with multifaceted consequences including multiplying medical costs and sequelae, societal effects like drunk driving and assault, and lost economic productivity. These large-scale outcomes are driven by the consumption of ethanol, a small permeable molecule that has myriad effects in the human body, particularly in the liver and brain. In this review, we have summarized effects of acute and chronic alcohol consumption on epigenetic mechanisms that may drive pathobiology of Alcohol Use Disorder (AUD) while identifying areas of need for future research. Recent findings Epigenetics has emerged as an interesting field of biology at the intersection of genetics and the environment, and ethanol in particular has been identified as a potent modulator of the epigenome with various effects on DNA methylation, histone modifications, and non-coding RNAs. These changes alter chromatin dynamics and regulate gene expression that contribute to behavioral and physiological changes leading to the development of AUD psychopathology and cancer pathology. Summary Evidence and discussion presented here from preclinical results and available translational studies have increased our knowledge of the epigenetic effects of alcohol consumption. These studies have identified targets that can be used to develop better therapies to reduce chronic alcohol abuse and mitigate its societal burden and pathophysiology.
Collapse
Affiliation(s)
- Russell S Dulman
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Gabriela M Wandling
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA.,Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
23
|
Moore SR, Humphreys KL, Colich NL, Davis EG, Lin DTS, MacIsaac JL, Kobor MS, Gotlib IH. Distinctions between sex and time in patterns of DNA methylation across puberty. BMC Genomics 2020; 21:389. [PMID: 32493224 PMCID: PMC7268482 DOI: 10.1186/s12864-020-06789-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/20/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There are significant sex differences in human physiology and disease; the genomic sources of these differences, however, are not well understood. During puberty, a drastic neuroendocrine shift signals physical changes resulting in robust sex differences in human physiology. Here, we explore how shifting patterns of DNA methylation may inform these pathways of biological plasticity during the pubertal transition. In this study we analyzed DNA methylation (DNAm) in saliva at two time points across the pubertal transition within the same individuals. Our purpose was to compare two domains of DNAm patterns that may inform processes of sexual differentiation 1) sex related sites, which demonstrated differences between males from females and 2) time related sites in which DNAm shifted significantly between timepoints. We further explored the correlated network structure sex and time related DNAm networks and linked these patterns to pubertal stage, assays of salivary testosterone, a reliable diagnostic of free, unbound hormone that is available to act on target tissues, and overlap with androgen response elements. RESULTS Sites that differed by biological sex were largely independent of sites that underwent change across puberty. Time-related DNAm sites, but not sex-related sites, formed correlated networks that were associated with pubertal stage. Both time and sex DNAm networks reflected salivary testosterone levels that were enriched for androgen response elements, with sex-related DNAm networks being informative of testosterone levels above and beyond biological sex later in the pubertal transition. CONCLUSIONS These results inform our understanding of the distinction between sex- and time-related differences in DNAm during the critical period of puberty and highlight a novel linkage between correlated patterns of sex-related DNAm and levels of salivary testosterone.
Collapse
Affiliation(s)
- Sarah Rose Moore
- Department of Medical Genetics, University of British Columbia
- BC Children's Hospital Research Institute, 938 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada.
| | - Kathryn Leigh Humphreys
- Department of Psychology and Human Development, Vanderbilt University, 230 Appleton Pl, Nashville, TN, 37203, USA
| | - Natalie Lisanne Colich
- Department of Psychology, University of Washington Seattle, Guthrie Hall (GTH), 119A 98195-1525, Seattle, WA, 98105, USA
| | - Elena Goetz Davis
- Department of Psychology, Stanford University, 450 Jane Stanford Way, Stanford, CA, 94305, USA
| | - David Tse Shen Lin
- Department of Medical Genetics, University of British Columbia
- BC Children's Hospital Research Institute, 938 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
| | - Julia Lynn MacIsaac
- Department of Medical Genetics, University of British Columbia
- BC Children's Hospital Research Institute, 938 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
| | - Michael Steffen Kobor
- Department of Medical Genetics, University of British Columbia
- BC Children's Hospital Research Institute, 938 W 28th Ave, Vancouver, BC, V5Z 4H4, Canada
| | - Ian Henry Gotlib
- Department of Psychology, Stanford University, 450 Jane Stanford Way, Stanford, CA, 94305, USA
| |
Collapse
|
24
|
Abstract
The hippocampus is central to spatial learning and stress responsiveness, both of which differ in form and function in males versus females, yet precisely how the hippocampus contributes to these sex differences is largely unknown. In reproductively mature individuals, sex differences in the steroid hormone milieu undergirds many sex differences in hippocampal-related endpoints. However, there is also evidence for developmental programming of adult hippocampal function, with a central role for androgens as well as their aromatized byproduct, estrogens. These include sex differences in cell genesis, synapse formation, dendritic arborization, and excitatory/inhibitory balance. Enduring effects of steroid hormone modulation occur during two developmental epochs, the first being the classic perinatal critical period of sexual differentiation of the brain and the other being adolescence and the associated hormonal changes of puberty. The cellular mechanisms by which steroid hormones enduringly modify hippocampal form and function are poorly understood, but we here review what is known and highlight where attention should be focused.
Collapse
|
25
|
Csaba G. Reprogramming of the Immune System by Stress and Faulty Hormonal Imprinting. Clin Ther 2020; 42:983-992. [PMID: 32307123 DOI: 10.1016/j.clinthera.2020.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Hormonal imprinting is taking place perinatally at the first encounter between the developing hormone receptors and their target hormones. However, in this crucial period when the developmental window for physiological imprinting is open, other molecules, such as synthetic hormones and endocrine disruptors can bind to the receptors, leading to faulty imprinting with life-long consequences, especially to the immune system. This review presents the factors of stress and faulty hormonal imprinting that lead to reprogramming of the immune system. METHODS Relevant publications from Pubmed since 1990 were reviewed and synthesized. FINDINGS The developing immune system is rather sensitive to hormonal effects. Faulty hormonal imprinting is able to reprogram the original developmental program present in a given cell, with lifelong consequences, manifested in alteration of hormone binding by receptors, susceptibility to certain (non-infectious) diseases, and triggering of other diseases. As stress mobilizes the hypothalamic-pituitary-adrenal axis if it occurred during gestation or perinatally, it could lead to faulty hormonal imprinting in the immune system, manifested later as allergic and autoimmune diseases or weakness of normal immune defenses. Hormonal imprinting is an epigenetic process and is carried to the offspring without alteration of DNA base sequences. This means that any form of early-life stress alone or in association with hormonal imprinting could be associated with the developmental origin of health and disease (DOHaD). As puberty is also a period of reprogramming, stress or faulty imprinting can change the original (developmental) program, also with life-long consequences. IMPLICATIONS Considering the continuous differentiation of immune cells (from blast-cells) during the whole life, there is a possibility of late-imprinting or stress-activated reprogramming in the immune system at any periods of life, with later pathogenetic consequences.
Collapse
Affiliation(s)
- György Csaba
- Department of Genetics, Cell, and Immunobiology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
26
|
Salberg S, Noel M, Burke NN, Vinall J, Mychasiuk R. Utilization of a rodent model to examine the neurological effects of early life adversity on adolescent pain sensitivity. Dev Psychobiol 2020; 62:386-399. [DOI: 10.1002/dev.21922] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/19/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Sabrina Salberg
- Department of Psychology University of Calgary Calgary AB Canada
- Alberta Children’s Hospital Research Institute University of Calgary Calgary AB Canada
- Hotchkiss Brain Institute University of Calgary Calgary AB Canada
| | - Melanie Noel
- Department of Psychology University of Calgary Calgary AB Canada
- Alberta Children’s Hospital Research Institute University of Calgary Calgary AB Canada
- Hotchkiss Brain Institute University of Calgary Calgary AB Canada
| | - Nikita N. Burke
- Hotchkiss Brain Institute University of Calgary Calgary AB Canada
- Comparative Biology & Experimental Medicine, and Physiology & Pharmacology University of Calgary Calgary AB Canada
| | - Jillian Vinall
- Department of Anesthesia University of Calgary Calgary AB Canada
| | - Richelle Mychasiuk
- Department of Psychology University of Calgary Calgary AB Canada
- Alberta Children’s Hospital Research Institute University of Calgary Calgary AB Canada
- Hotchkiss Brain Institute University of Calgary Calgary AB Canada
| |
Collapse
|
27
|
Sze Y, Brunton PJ. Sex, stress and steroids. Eur J Neurosci 2019; 52:2487-2515. [DOI: 10.1111/ejn.14615] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
| | - Paula J. Brunton
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
- Zhejiang University‐University of Edinburgh Joint Institute Haining Zhejiang China
| |
Collapse
|
28
|
Howard SR, Dunkel L. Delayed Puberty-Phenotypic Diversity, Molecular Genetic Mechanisms, and Recent Discoveries. Endocr Rev 2019; 40:1285-1317. [PMID: 31220230 PMCID: PMC6736054 DOI: 10.1210/er.2018-00248] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/31/2019] [Indexed: 02/07/2023]
Abstract
This review presents a comprehensive discussion of the clinical condition of delayed puberty, a common presentation to the pediatric endocrinologist, which may present both diagnostic and prognostic challenges. Our understanding of the genetic control of pubertal timing has advanced thanks to active investigation in this field over the last two decades, but it remains in large part a fascinating and mysterious conundrum. The phenotype of delayed puberty is associated with adult health risks and common etiologies, and there is evidence for polygenic control of pubertal timing in the general population, sex-specificity, and epigenetic modulation. Moreover, much has been learned from comprehension of monogenic and digenic etiologies of pubertal delay and associated disorders and, in recent years, knowledge of oligogenic inheritance in conditions of GnRH deficiency. Recently there have been several novel discoveries in the field of self-limited delayed puberty, encompassing exciting developments linking this condition to both GnRH neuronal biology and metabolism and body mass. These data together highlight the fascinating heterogeneity of disorders underlying this phenotype and point to areas of future research where impactful developments can be made.
Collapse
Affiliation(s)
- Sasha R Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Leo Dunkel
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
29
|
Pittet F, Tyson C, Herrington JA, Houdelier C, Lumineau S. Postnatal care generates phenotypic behavioural correlations in the Japanese quail. Behav Ecol Sociobiol 2019. [DOI: 10.1007/s00265-019-2735-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
30
|
Post-translational histone modifications and their interaction with sex influence normal brain development and elaboration of neuropsychiatric disorders. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1968-1981. [DOI: 10.1016/j.bbadis.2018.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
|
31
|
Abstract
Delayed pubertal onset has many etiologies, but on average two-thirds of patients presenting with late puberty have self-limited (or constitutional) delayed puberty. Self-limited delayed puberty often has a strong familial basis. Segregation analyses from previous studies show complex models of inheritance, most commonly autosomal dominant, but also including autosomal recessive, bilineal, and X-linked. Sporadic cases are also observed. Despite this, the neuroendocrine mechanisms and genetic regulation remain unclear in the majority of patients with self-limited delayed puberty. Only rarely have mutations in genes known to cause aberrations of the hypothalamic-pituitary-gonadal axis been identified in cases of delayed puberty, and the majority of these are in relatives of patients with congenital hypogonadotropic hypogonadism (CHH), for example in the FGFR1 and GNRHR genes. Using next generation sequencing in a large family with isolated self-limited delayed puberty, a pathogenic mutation in the CHH gene HS6ST1 was found as the likely cause for this phenotype. Additionally, a study comparing the frequency of mutations in genes that cause GnRH deficiency between probands with CHH and probands with isolated self-limited delayed puberty identified that a significantly higher proportion of mutations with a greater degree of oligogenicity were seen in the CHH group. Mutations in the gene IGSF10 have been implicated in the pathogenesis of familial late puberty in a large Finnish cohort. IGSF10 disruption represents a fetal origin of delayed puberty, with dysregulation of GnRH neuronal migration during embryonic development presenting for the first time in adolescence as late puberty. Some patients with self-limited delayed puberty have distinct constitutional features of growth and puberty. Deleterious variants in FTO have been found in families with delayed puberty with extremely low BMI and maturational delay in growth in early childhood. Recent exciting evidence highlights the importance of epigenetic up-regulation of GnRH transcription by a network of miRNAs and transcription factors, including EAP1, during puberty. Whilst a fascinating heterogeneity of genetic defects have been shown to result in delayed and disordered puberty, and many are yet to be discovered, genetic testing may become a realistic diagnostic tool for the differentiation of conditions of delayed puberty.
Collapse
Affiliation(s)
- Sasha R. Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, London, United Kingdom
| |
Collapse
|
32
|
Okada N, Ando S, Sanada M, Hirata-Mogi S, Iijima Y, Sugiyama H, Shirakawa T, Yamagishi M, Kanehara A, Morita M, Yagi T, Hayashi N, Koshiyama D, Morita K, Sawada K, Ikegame T, Sugimoto N, Toriyama R, Masaoka M, Fujikawa S, Kanata S, Tada M, Kirihara K, Yahata N, Araki T, Jinde S, Kano Y, Koike S, Endo K, Yamasaki S, Nishida A, Hiraiwa-Hasegawa M, Bundo M, Iwamoto K, Tanaka SC, Kasai K. Population-neuroscience study of the Tokyo TEEN Cohort (pn-TTC): Cohort longitudinal study to explore the neurobiological substrates of adolescent psychological and behavioral development. Psychiatry Clin Neurosci 2019; 73:231-242. [PMID: 30588712 DOI: 10.1111/pcn.12814] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 12/06/2018] [Accepted: 12/25/2018] [Indexed: 12/14/2022]
Abstract
AIM Adolescence is a crucial stage of psychological development and is critically vulnerable to the onset of psychopathology. Our understanding of how the maturation of endocrine, epigenetics, and brain circuit may underlie psychological development in adolescence, however, has not been integrated. Here, we introduce our research project, the population-neuroscience study of the Tokyo TEEN Cohort (pn-TTC), a longitudinal study to explore the neurobiological substrates of development during adolescence. METHODS Participants in the first wave of the pn-TTC (pn-TTC-1) study were recruited from those of the TTC study, a large-scale epidemiological survey in which 3171 parent-adolescent pairs were recruited from the general population. Participants underwent psychological, cognitive, sociological, and physical assessment. Moreover, adolescents and their parents underwent magnetic resonance imaging (MRI; structural MRI, resting-state functional MRI, and magnetic resonance spectroscopy), and adolescents provided saliva samples for hormone analysis and for DNA analysis including epigenetics. Furthermore, the second wave (pn-TTC-2) followed similar methods as in the first wave. RESULTS A total of 301 parent-adolescent pairs participated in the pn-TTC-1 study. Moreover, 281 adolescents participated in the pn-TTC-2 study, 238 of whom were recruited from the pn-TTC-1 sample. The instruction for data request is available at: http://value.umin.jp/data-resource.html. CONCLUSION The pn-TTC project is a large-scale and population-neuroscience-based survey with a plan of longitudinal biennial follow up. Through this approach we seek to elucidate adolescent developmental mechanisms according to biopsychosocial models. This current biomarker research project, using minimally biased samples recruited from the general population, has the potential to expand the new research field of population neuroscience.
Collapse
Affiliation(s)
- Naohiro Okada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
| | - Shuntaro Ando
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Motoyuki Sanada
- Center for Applied Psychological Science, Kwansei Gakuin University, Nishinomiya, Japan
| | - Sachiko Hirata-Mogi
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yudai Iijima
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Physical and Health Education, Graduate School of Education, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Sugiyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Integrated Educational Sciences, Graduate School of Education, The University of Tokyo, Tokyo, Japan
| | - Toru Shirakawa
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mika Yamagishi
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akiko Kanehara
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaya Morita
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoko Yagi
- Department of Child Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noriyuki Hayashi
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daisuke Koshiyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kentaro Morita
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kingo Sawada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tempei Ikegame
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Molecular Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noriko Sugimoto
- Department of Child Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Rie Toriyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mio Masaoka
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinya Fujikawa
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sho Kanata
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Psychiatry, Teikyo University School of Medicine, Tokyo, Japan
| | - Mariko Tada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
| | - Kenji Kirihara
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noriaki Yahata
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tsuyoshi Araki
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Seiichiro Jinde
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukiko Kano
- Department of Child Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinsuke Koike
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan.,UTokyo Institute for Diversity and Adaptation of Human Mind (UTIDAHM), The University of Tokyo, Tokyo, Japan
| | - Kaori Endo
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Syudo Yamasaki
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Atsushi Nishida
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mariko Hiraiwa-Hasegawa
- Department of Evolutionary Studies of Biosystems, School of Advanced Sciences, Graduate University for Advanced Studies (SOKENDAI), Hayama, Japan
| | - Miki Bundo
- Department of Molecular Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuya Iwamoto
- Department of Molecular Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Saori C Tanaka
- Department of Computational Neurobiology, ATR Computational Neuroscience Laboratories, Kyoto, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
| |
Collapse
|
33
|
Azadi M, Azizi H, Haghparast A. Paternal exposure to morphine during adolescence induces reward-resistant phenotype to morphine in male offspring. Brain Res Bull 2019; 147:124-132. [DOI: 10.1016/j.brainresbull.2019.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 02/06/2019] [Indexed: 12/29/2022]
|
34
|
Csaba G. Hormonal Imprinting: The First Cellular-level Evidence of Epigenetic Inheritance and its Present State. Curr Genomics 2019; 20:409-418. [PMID: 32476998 PMCID: PMC7235388 DOI: 10.2174/1389202920666191116113524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 09/24/2019] [Accepted: 10/21/2019] [Indexed: 12/28/2022] Open
Abstract
Hormonal imprinting takes place perinatally at the first encounter between the developing hormone receptor and its target hormone. This process is needed for the normal function of the receptor-hormone pair and its effect is life-long. However, in this critical period, when the developmental window is open, related molecules (members of the same hormone family, synthetic hormones and hormone-like molecules, endocrine disruptors) also can be bound by the receptor, causing life-long faulty imprinting. In this case, the receptors’ binding capacity changes and alterations are caused at adult age in the sexual and behavioral sphere, in the brain and bones, inclination to diseases and manifestation of diseases, etc. Hereby, faulty hormonal imprinting is the basis of metabolic and immunological imprinting as well as the developmental origin of health and disease (DOHaD). Although the perinatal period is the most critical for faulty imprinting, there are other critical periods as weaning and adolescence, when the original imprinting can be modified or new imprintings develop. Hormonal imprinting is an epigenetic process, without changing the base sequence of DNA, it is inherited in the cell line of the imprinted cells and also transgenerationally (up to 1000 generations in unicellulars and up to the 3rd generation in mammals are justified). Considering the enormously growing number and amount of faulty imprinters (endocrine disruptors) and the hereditary character of faulty imprinting, this latter is threatening the whole human endocrine system.
Collapse
Affiliation(s)
- György Csaba
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
35
|
|
36
|
Zancan M, Cunha RSR, Schroeder F, Xavier LL, Rasia‐Filho AA. Remodeling of the number and structure of dendritic spines in the medial amygdala: From prepubertal sexual dimorphism to puberty and effect of sexual experience in male rats. Eur J Neurosci 2018; 48:1851-1865. [DOI: 10.1111/ejn.14052] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/18/2018] [Accepted: 06/13/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Mariana Zancan
- Department of Basic Sciences/PhysiologyFederal University of Health Sciences Porto Alegre Brazil
- Graduation Program in NeuroscienceFederal University of Rio Grande do Sul Porto Alegre Brazil
| | - Rick Shandler R. Cunha
- Department of Basic Sciences/PhysiologyFederal University of Health Sciences Porto Alegre Brazil
| | - Francielle Schroeder
- Laboratory of Tissue BiologyFaculty of BiosciencesPontifical Catholic University of Rio Grande do Sul (PUCRS) Porto Alegre Brazil
| | - Léder L. Xavier
- Laboratory of Tissue BiologyFaculty of BiosciencesPontifical Catholic University of Rio Grande do Sul (PUCRS) Porto Alegre Brazil
| | - Alberto A. Rasia‐Filho
- Department of Basic Sciences/PhysiologyFederal University of Health Sciences Porto Alegre Brazil
- Graduation Program in NeuroscienceFederal University of Rio Grande do Sul Porto Alegre Brazil
| |
Collapse
|
37
|
Hippocampus-dependent memory and allele-specific gene expression in adult offspring of alcohol-consuming dams after neonatal treatment with thyroxin or metformin. Mol Psychiatry 2018; 23:1643-1651. [PMID: 28727687 PMCID: PMC5775940 DOI: 10.1038/mp.2017.129] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 02/06/2023]
Abstract
Fetal alcohol spectrum disorder (FASD), the result of fetal alcohol exposure (FAE), affects 2-11% of children worldwide, with no effective treatments. Hippocampus-based learning and memory deficits are key symptoms of FASD. Our previous studies show hypothyroxinemia and hyperglycemia of the alcohol-consuming pregnant rat, which likely affects fetal neurodevelopment. We administered vehicle, thyroxine (T4) or metformin to neonatal rats post FAE and rats were tested in the hippocampus-dependent contextual fear-conditioning paradigm in adulthood. Both T4 and metformin alleviated contextual fear memory deficit induced by FAE, and reversed the hippocampal expression changes in the thyroid hormone-inactivating enzyme, deiodinase-III (Dio3) and insulin-like growth factor 2 (Igf2), genes that are known to modulate memory processes. Neonatal T4 restored maternal allelic expressions of the imprinted Dio3 and Igf2 in the adult male hippocampus, while metformin restored FAE-caused changes in Igf2 expression only. The decreased hippocampal expression of DNA methyltransferase 1 (Dnmt1) that maintains the imprinting of Dio3 and Igf2 during development was normalized by both treatments. Administering Dnmt1 inhibitor to control neonates resulted in FAE-like deficits in fear memory and hippocampal allele-specific expression of Igf2, which were reversed by metformin. We propose that neonatal administration of T4 and metformin post FAE affect memory via elevating Dnmt1 and consequently normalizing hippocampal Dio3 and Igf2 expressions in the adult offspring. The present results indicate that T4 and metformin, administered during the neonatal period that is equivalent to the third trimester of human pregnancy, are potential treatments for FASD and conceivably for other neurodevelopmental disorders with cognitive deficits.
Collapse
|
38
|
Palumbo S, Mariotti V, Iofrida C, Pellegrini S. Genes and Aggressive Behavior: Epigenetic Mechanisms Underlying Individual Susceptibility to Aversive Environments. Front Behav Neurosci 2018; 12:117. [PMID: 29950977 PMCID: PMC6008527 DOI: 10.3389/fnbeh.2018.00117] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/28/2018] [Indexed: 12/14/2022] Open
Abstract
Over the last two decades, the study of the relationship between nature and nurture in shaping human behavior has encountered a renewed interest. Behavioral genetics showed that distinct polymorphisms of genes that code for proteins that control neurotransmitter metabolic and synaptic function are associated with individual vulnerability to aversive experiences, such as stressful and traumatic life events, and may result in an increased risk of developing psychopathologies associated with violence. On the other hand, recent studies indicate that experiencing aversive events modulates gene expression by introducing stable changes to DNA without modifying its sequence, a mechanism known as “epigenetics”. For example, experiencing adversities during periods of maximal sensitivity to the environment, such as prenatal life, infancy and early adolescence, may introduce lasting epigenetic marks in genes that affect maturational processes in brain, thus favoring the emergence of dysfunctional behaviors, including exaggerate aggression in adulthood. The present review discusses data from recent research, both in humans and animals, concerning the epigenetic regulation of four genes belonging to the neuroendocrine, serotonergic and oxytocinergic pathways—Nuclear receptor subfamily 3-group C-member 1 (NR3C1), oxytocin receptor (OXTR), solute carrier-family 6 member 4 (SLC6A4) and monoamine oxidase A (MAOA)—and their role in modulating vulnerability to proactive and reactive aggressive behavior. Behavioral genetics and epigenetics are shedding a new light on the fine interaction between genes and environment, by providing a novel tool to understand the molecular events that underlie aggression. Overall, the findings from these studies carry important implications not only for neuroscience, but also for social sciences, including ethics, philosophy and law.
Collapse
Affiliation(s)
- Sara Palumbo
- Department of Surgical, Medical, Molecular Pathology and Critical Care, University of Pisa, Pisa, Italy
| | - Veronica Mariotti
- Department of Experimental and Clinical Medicine, University of Pisa, Pisa, Italy
| | | | - Silvia Pellegrini
- Department of Experimental and Clinical Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
39
|
Effects of adolescent social stress and antidepressant treatment on cognitive inflexibility and Bdnf epigenetic modifications in the mPFC of adult mice. Psychoneuroendocrinology 2018; 88:92-101. [PMID: 29195162 DOI: 10.1016/j.psyneuen.2017.11.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/22/2017] [Accepted: 11/25/2017] [Indexed: 12/17/2022]
Abstract
Adolescent social stress (ASS) can increase susceptibility to depression in adulthood. However, the underlying psychological and neural mechanisms remain unclear. Cortically mediated cognitive dysfunctions are increasingly recognized as an independent and important risk factor of depression. Using social defeat stress, a classical animal model of depression, our previous studies found that mice subjected to this form of stress during early adolescence displayed cognitive inflexibility (CI) in adulthood. This change was accompanied by a down-regulation of Bdnf gene expression in the medial prefrontal cortex (mPFC); this gene encodes a key molecule involved in depression and antidepressant action. In the present paper, we identified epigenetic modification of Bdnf as a possible mechanism underlying the behavioral and molecular changes. ASS induced a set of depressive phenotypes, including increased social avoidance and CI, as well as reduced levels of total Bdnf and isoform IV but not isoform I or VI transcripts in the mPFC. In parallel with changes in Bdnf gene expression, previously stressed adult mice showed increased levels of dimethylation of histone H3 at lysine K9 (H3K9me2) immediately downstream of the Bdnf IV promoter. On the other hand, no differences were found in trimethylation of histone H3 at lysine K4 (H3K4me3) or in acetylation of histone H3 at lysine K9 (H3K9ac) or at K4 (H3K4ac) in the Bdnf IV promoter. Likewise, no alterations were found in DNA methylation of the Bdnf IV promoter. Additionally, treatment with the chronic antidepressant tranylcypromine reversed Bdnf epigenetic changes and related gene transcription while also reversing CI, but not social avoidance, in previously stressed adult mice. These results suggest that epigenetic changes to the Bdnf gene in the mPFC after adolescent social adversity may be involved in the regulation of cognitive dysfunction in depression and antidepressant action in adulthood.
Collapse
|
40
|
Gałecki P, Talarowska M. Neurodevelopmental theory of depression. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:267-272. [PMID: 28571776 DOI: 10.1016/j.pnpbp.2017.05.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 05/17/2017] [Accepted: 05/27/2017] [Indexed: 01/20/2023]
Abstract
The aim of research studies in the field of psychiatry conducted in recent years is to formulate a consistent theory that would exhaustively explain the aetiology of depression. So far, biochemical, genetic, anatomical and environmental factors, which may play a role in the occurrence of the first symptoms of depressive disorders, have been sought. The authors of this paper present a theory that combines the previously mentioned elements into one whole and links them to one another. We have called our theory "neurodevelopmental" to underline the importance and impact of earlier stages of human life, including the prenatal period, on the occurrence of depressive disorders. We will make an attempt to find an answer to why this time in the life of a human being is so important, what kind of biological mechanisms are activated then, and what aspects of our later functioning are affected by them.
Collapse
Affiliation(s)
- Piotr Gałecki
- Department of Adult Psychiatry, Medical University of Lodz, Lodz, Poland
| | - Monika Talarowska
- Department of Adult Psychiatry, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
41
|
How age, sex and genotype shape the stress response. Neurobiol Stress 2016; 6:44-56. [PMID: 28229108 PMCID: PMC5314441 DOI: 10.1016/j.ynstr.2016.11.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/19/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022] Open
Abstract
Exposure to chronic stress is a leading pre-disposing factor for several neuropsychiatric disorders as it often leads to maladaptive responses. The response to stressful events is heterogeneous, underpinning a wide spectrum of distinct changes amongst stress-exposed individuals'. Several factors can underlie a different perception to stressors and the setting of distinct coping strategies that will lead to individual differences on the susceptibility/resistance to stress. Beyond the factors related to the stressor itself, such as intensity, duration or predictability, there are factors intrinsic to the individuals that are relevant to shape the stress response, such as age, sex and genetics. In this review, we examine the contribution of such intrinsic factors to the modulation of the stress response based on experimental rodent models of response to stress and discuss to what extent that knowledge can be potentially translated to humans. Effect of age in the stress response. Effect of sex in the stress response. Effect of genotype in the stress response.
Collapse
|
42
|
Rao YS, Pak TR. microRNAs and the adolescent brain: Filling the knowledge gap. Neurosci Biobehav Rev 2016; 70:313-322. [PMID: 27328787 PMCID: PMC5074866 DOI: 10.1016/j.neubiorev.2016.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 06/09/2016] [Accepted: 06/11/2016] [Indexed: 12/14/2022]
Abstract
Over two decades ago the discovery of microRNAs (miRNA) broadened our understanding of the diverse molecular pathways mediating post-transcriptional control over gene expression. These small non-coding RNAs dynamically fluctuate, temporally and spatially, throughout the lifespan of all organisms. The fundamental role that miRNAs have in shaping embryonic neurodevelopment provides strong evidence that adolescent brain remodeling could be rooted in the changing miRNA landscape of the cell. Few studies have directly measured miRNA gene expression changes in the brain across pubertal development, and even less is known about the functional impact of those miRNAs on the maturational processes that occur in the developing adolescent brain. This review summarizes miRNA biogenesis and function in the brain in the context of normal (i.e. not diseased) physiology. These landmark studies can guide predictions about the role of miRNAs in facilitating maturation of the adolescent brain. However, there are clear indicators that adolescence/puberty is a unique life stage, suggesting miRNA function during adolescence is distinct from those in any other previously described system.
Collapse
Affiliation(s)
- Yathindar S Rao
- Loyola University Chicago, Stritch School of Medicine, Department of Cell and Molecular Physiology, United States
| | - Toni R Pak
- Loyola University Chicago, Stritch School of Medicine, Department of Cell and Molecular Physiology, United States.
| |
Collapse
|
43
|
Neuroendokrine Regulation der Pubertät und ihre Störungen. GYNAKOLOGISCHE ENDOKRINOLOGIE 2016. [DOI: 10.1007/s10304-016-0091-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
Vigil P, Del Río JP, Carrera BÁ, ArÁnguiz FC, Rioseco H, Cortés ME. Influence of sex steroid hormones on the adolescent brain and behavior: An update. LINACRE QUARTERLY 2016; 83:308-329. [PMID: 27833209 DOI: 10.1080/00243639.2016.1211863] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This review explains the main effects exerted by sex steroids and other hormones on the adolescent brain. During the transition from puberty to adolescence, these hormones participate in the organizational phenomena that structurally shape some brain circuits. In adulthood, this will propitiate some specific behavior as responses to the hormones now activating those neural circuits. Adolescence is, then, a critical "organizational window" for the brain to develop adequately, since steroid hormones perform important functions at this stage. For this reason, the adolescent years are very important for future behaviors in human beings. Changes that occur or fail to occur during adolescence will determine behaviors for the rest of one's lifetime. Consequently, understanding the link between adolescent behavior and brain development as influenced by sex steroids and other hormones and compounds is very important in order to interpret various psycho-affective pathologies. Lay Summary : The effect of steroid hormones on the development of the adolescent brain, and therefore, on adolescent behavior, is noticeable. This review presents their main activational and organizational effects. During the transition from puberty to adolescence, organizational phenomena triggered by steroids structurally affect the remodeling of brain circuits. Later in adulthood, these changes will be reflected in behavioral responses to such hormones. Adolescence can then be seen as a fundamental "organizational window" during which sex steroids and other hormones and compounds play relevant roles. The understanding of the relationship between adolescent behavior and the way hormones influence brain development help understand some psychological disorders.
Collapse
Affiliation(s)
- Pilar Vigil
- Pontificia Universidad Católica de Chile, Vicerrectoría de Comunicaciones, Santiago, Chile; Biomedical Division, Reproductive Health Research Institute, Santiago, Chile
| | - Juan Pablo Del Río
- Biomedical Division, Reproductive Health Research Institute, Santiago, Chile; Universidad de los Andes, Facultad de Medicina, Escuela de Medicina, Santiago, Chile
| | - BÁrbara Carrera
- Biomedical Division, Reproductive Health Research Institute, Santiago, Chile
| | | | - Hernán Rioseco
- Biomedical Division, Reproductive Health Research Institute, Santiago, Chile
| | - Manuel E Cortés
- Biomedical Division, Reproductive Health Research Institute, Santiago, Chile; Universidad Bernardo O Higgins, Facultad de Salud, Departamento de Ciencias Químicas y Biológicas, Santiago, Chile
| |
Collapse
|
45
|
Mychasiuk R, Metz GAS. Epigenetic and gene expression changes in the adolescent brain: What have we learned from animal models? Neurosci Biobehav Rev 2016; 70:189-197. [PMID: 27426956 DOI: 10.1016/j.neubiorev.2016.07.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/27/2016] [Accepted: 07/13/2016] [Indexed: 10/21/2022]
Abstract
Adolescence is defined as the gradual period of transition between childhood and adulthood that is characterized by significant brain maturation, growth spurts, sexual maturation, and heightened social interaction. Although originally believed to be a uniquely human aspect of development, rodent and non-human primates demonstrate maturational patterns that distinctly support an adolescent stage. As epigenetic processes are essential for development and differentiation, but also transpire in mature cells in response to environmental influences, they are an important aspect of adolescent brain maturation. The purpose of this review article was to examine epigenetic programming in animal models of brain maturation during adolescence. The discussion focuses on animal models to examine three main concepts; epigenetic processes involved in normal adolescent brain maturation, the influence of fetal programming on adolescent brain development and the epigenome, and finally, postnatal experiences such as exercise and drugs that modify epigenetic processes important for adolescent brain maturation. This corollary emphasizes the utility of animal models to further our understanding of complex processes such as epigenetic regulation and brain development.
Collapse
Affiliation(s)
- Richelle Mychasiuk
- Alberta Children's Hospital Research Institute, University of Calgary, Department of Psychology, AD030 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada.
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, T1K 3M4, Canada
| |
Collapse
|
46
|
Sex Differences in Nucleus Accumbens Transcriptome Profiles Associated with Susceptibility versus Resilience to Subchronic Variable Stress. J Neurosci 2016; 35:16362-76. [PMID: 26674863 DOI: 10.1523/jneurosci.1392-15.2015] [Citation(s) in RCA: 298] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED Depression and anxiety disorders are more prevalent in females, but the majority of research in animal models, the first step in finding new treatments, has focused predominantly on males. Here we report that exposure to subchronic variable stress (SCVS) induces depression-associated behaviors in female mice, whereas males are resilient as they do not develop these behavioral abnormalities. In concert with these different behavioral responses, transcriptional analysis of nucleus accumbens (NAc), a major brain reward region, by use of RNA sequencing (RNA-seq) revealed markedly different patterns of stress regulation of gene expression between the sexes. Among the genes displaying sex differences was DNA methyltransferase 3a (Dnmt3a), which shows a greater induction in females after SCVS. Interestingly, Dnmt3a expression levels were increased in the NAc of depressed humans, an effect seen in both males and females. Local overexpression of Dnmt3a in NAc rendered male mice more susceptible to SCVS, whereas Dnmt3a knock-out in this region rendered females more resilient, directly implicating this gene in stress responses. Associated with this enhanced resilience of female mice upon NAc knock-out of Dnmt3a was a partial shift of the NAc female transcriptome toward the male pattern after SCVS. These data indicate that males and females undergo different patterns of transcriptional regulation in response to stress and that a DNA methyltransferase in NAc contributes to sex differences in stress vulnerability. SIGNIFICANCE STATEMENT Women have a higher incidence of depression than men. However, preclinical models, the first step in developing new diagnostics and therapeutics, have been performed mainly on male subjects. Using a stress-based animal model of depression that causes behavioral effects in females but not males, we demonstrate a sex-specific transcriptional profile in brain reward circuitry. This transcriptional profile can be altered by removal of an epigenetic mechanism, which normally suppresses DNA transcription, creating a hybrid male/female transcriptional pattern. Removal of this epigenetic mechanism also induces behavioral resilience to stress in females. These findings shed new light onto molecular factors controlling sex differences in stress response.
Collapse
|
47
|
Junien C, Panchenko P, Fneich S, Pirola L, Chriett S, Amarger V, Kaeffer B, Parnet P, Torrisani J, Bolaños Jimenez F, Jammes H, Gabory A. [Epigenetics in transgenerational responses to environmental impacts: from facts and gaps]. Med Sci (Paris) 2016; 32:35-44. [PMID: 26850605 DOI: 10.1051/medsci/20163201007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The existence of non-genetic and non-cultural mechanisms that transfer information on the memory of parental exposures to various environments, determining the reactivity of the following generations to their environments during their life, are of growing interest. Yet fundamental questions remain about the nature, the roles and relative importance of epigenetic marks and processes, non-coding RNAs, or other mechanisms, and their persistence over generations. A model incorporating the various transmission systems, their cross-talks and windows of susceptibility to the environment as a function of sex/gender of parent and offspring, has yet to be built.
Collapse
Affiliation(s)
- Claudine Junien
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| | - Polina Panchenko
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| | - Sara Fneich
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| | | | | | - Valérie Amarger
- Inra, UMR 1280, université de Nantes, Institut des maladies de l'appareil digestif, F-44000 Nantes, France
| | - Bertrand Kaeffer
- Inra, UMR 1280, université de Nantes, Institut des maladies de l'appareil digestif, F-44000 Nantes, France
| | - Patricia Parnet
- Inra, UMR 1280, université de Nantes, Institut des maladies de l'appareil digestif, F-44000 Nantes, France
| | - Jérome Torrisani
- Inserm UMR1037, Centre de recherches en cancérologie de Toulouse, Université de Toulouse III Paul Sabatier, F-31037 Toulouse, France
| | - Francisco Bolaños Jimenez
- Inra, UMR 1280, université de Nantes, Institut des maladies de l'appareil digestif, F-44000 Nantes, France
| | - Hélène Jammes
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| | - Anne Gabory
- Inra, UMR1198, biologie du développement et reproduction, Domaine de Vilvert, Bâtiment 230, F-78350 Jouy-en-Josas, France
| |
Collapse
|
48
|
High Gestational Folic Acid Supplementation Alters Expression of Imprinted and Candidate Autism Susceptibility Genes in a sex-Specific Manner in Mouse Offspring. J Mol Neurosci 2015; 58:277-86. [PMID: 26547318 DOI: 10.1007/s12031-015-0673-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/30/2015] [Indexed: 02/07/2023]
Abstract
Maternal nutrients play critical roles in modulating epigenetic events and exert long-term influences on the progeny's health. Folic acid (FA) supplementation during pregnancy has decreased the incidence of neural tube defects in newborns, but the influence of high doses of maternal FA supplementation on infants' brain development is unclear. The present study was aimed at investigating the effects of a high dose of gestational FA on the expression of genes in the cerebral hemispheres (CHs) of 1-day-old pups. One week prior to mating and throughout the entire period of gestation, female C57BL/6J mice were fed a diet, containing FA at either 2 mg/kg (control diet (CD)) or 20 mg/kg (high maternal folic acid (HMFA)). At postnatal day 1, pups from different dams were sacrificed and CH tissues were collected. Quantitative RT-PCR and Western blot analysis confirmed sex-specific alterations in the expression of several genes that modulate various cellular functions (P < 0.05) in pups from the HMFA group. Genomic DNA methylation analysis showed no difference in the level of overall methylation in pups from the HMFA group. These findings demonstrate that HMFA supplementation alters offsprings' CH gene expression in a sex-specific manner. These changes may influence infants' brain development.
Collapse
|
49
|
Elson AE, Simerly RB. Developmental specification of metabolic circuitry. Front Neuroendocrinol 2015; 39:38-51. [PMID: 26407637 PMCID: PMC4681622 DOI: 10.1016/j.yfrne.2015.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 09/18/2015] [Accepted: 09/21/2015] [Indexed: 01/16/2023]
Abstract
The hypothalamus contains a core circuitry that communicates with the brainstem and spinal cord to regulate energy balance. Because metabolic phenotype is influenced by environmental variables during perinatal development, it is important to understand how these neural pathways form in order to identify key signaling pathways that are responsible for metabolic programming. Recent progress in defining gene expression events that direct early patterning and cellular specification of the hypothalamus, as well as advances in our understanding of hormonal control of central neuroendocrine pathways, suggest several key regulatory nodes that may represent targets for metabolic programming of brain structure and function. This review focuses on components of central circuitry known to regulate various aspects of energy balance and summarizes what is known about their developmental neurobiology within the context of metabolic programming.
Collapse
Affiliation(s)
- Amanda E Elson
- The Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Keck School of Medicine, Los Angeles, CA 90027, USA
| | - Richard B Simerly
- The Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Keck School of Medicine, Los Angeles, CA 90027, USA.
| |
Collapse
|
50
|
Sex differences and stress across the lifespan. Nat Neurosci 2015; 18:1413-20. [PMID: 26404716 DOI: 10.1038/nn.4112] [Citation(s) in RCA: 528] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022]
Abstract
Sex differences in stress responses can be found at all stages of life and are related to both the organizational and activational effects of gonadal hormones and to genes on the sex chromosomes. As stress dysregulation is the most common feature across neuropsychiatric diseases, sex differences in how these pathways develop and mature may predict sex-specific periods of vulnerability to disruption and increased disease risk or resilience across the lifespan. The aging brain is also at risk to the effects of stress, where the rapid decline of gonadal hormones in women combined with cellular aging processes promote sex biases in stress dysregulation. In this Review, we discuss potential underlying mechanisms driving sex differences in stress responses and their relevance to disease. Although stress is involved in a much broader range of diseases than neuropsychiatric ones, we highlight here this area and its examples across the lifespan.
Collapse
|