1
|
Torres DB, Lopes A, Rodrigues AJ, Lopes MG, Ventura-Silva AP, Sousa N, Gontijo JAR, Boer PA. Gestational protein restriction alters early amygdala neurochemistry in male offspring. Nutr Neurosci 2023; 26:1103-1119. [PMID: 36331123 DOI: 10.1080/1028415x.2022.2131064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Gestational protein intake restriction-induced long-lasting harmful outcomes in the offspring's organs and systems. However, few studies have focused on this event's impact on the brain's structures and neurochemical compounds. AIM The present study investigated the effects on the amygdala neurochemical composition and neuronal structure in gestational protein-restricted male rats' offspring. METHODS Dams were maintained on isocaloric standard rodent laboratory chow with regular protein [NP, 17%] or low protein content [LP, 6%]. Total cells were quantified using the Isotropic fractionator method, Neuronal 3D reconstruction, and dendritic tree analysis using the Golgi-Cox technique. Western blot and high-performance liquid chromatography performed neurochemical studies. RESULTS The gestational low-protein feeding offspring showed a significant decrease in birth weight up to day 14, associated with unaltered brain weight in youth or adult progenies. The amygdala cell numbers were unchanged, and the dendrites length and dendritic ramifications 3D analysis in LP compared to age-matched NP progeny. However, the current study shows reduced amygdala content of norepinephrine, epinephrine, and dopamine in LP progeny. These offspring observed a significant reduction in the amygdala glucocorticoid (GR) and mineralocorticoid (MR) receptor protein levels. Also corticotrophin-releasing factor (CRF) amygdala protein content was reduced in 7 and 14-day-old LP rats. CONCLUSION The observed amygdala neurochemical changes may represent adaptation during embryonic development in response to elevated fetal exposure to maternal corticosteroid levels. In this way, gestational malnutrition stress can alter the amygdala's neurochemical content and may contribute to known behavioral changes induced by gestational protein restriction.
Collapse
Affiliation(s)
- Daniele B Torres
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| | - Agnes Lopes
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| | - Ana J Rodrigues
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Marcelo G Lopes
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| | - Ana P Ventura-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - José A R Gontijo
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| | - Patricia A Boer
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
2
|
Wei M, Gao Q, Liu J, Yang Y, Yang J, Fan J, Lv S, Yang S. Development programming: Stress during gestation alters offspring development in sheep. Reprod Domest Anim 2023; 58:1497-1511. [PMID: 37697713 DOI: 10.1111/rda.14465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/05/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023]
Abstract
Inappropriate management practices of domestic animals during pregnancy can be potential stressors, resulting in complex behavioural, physiological and neurological consequences in the developing offspring. Some of these consequences can last into adulthood or propagate to subsequent generations. We systematically summarized the results of different experimental patterns using artificially increased maternal glucocorticoid levels or prenatal maternal physiological stress paradigms, mediators between prenatal maternal stress (PMS) and programming effects in the offspring and the effects of PMS on offspring phenotypes in sheep. PMS can impair birthweight, regulate the development of the hypothalamic-pituitary-adrenal axis, modify behavioural patterns and cognitive abilities and alter gene expression and brain morphology in offspring. Further research should focus on the effects of programming on gene expression, immune function, gut microbiome, sex-specific effects and maternal behaviour of offspring, especially comparative studies of gestational periods when PMS is applied, continual studies of programming effects on offspring and treatment strategies that effectively reverse the detrimental programming effects of prenatal stress.
Collapse
Affiliation(s)
- Mingji Wei
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, China
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Qian Gao
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Junjun Liu
- Hebei Agriculture University, Baoding, China
| | - Yan Yang
- Linyi Academy of Agricultural Sciences, Linyi, China
| | - Jinyan Yang
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Jingchang Fan
- Jiaxiang County Sheep Breeding Farm, Jiaxiang, China
| | - Shenjin Lv
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Shengmei Yang
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, China
| |
Collapse
|
3
|
Sato S, Watanabe S, Saito Y, Takanashi A, Ikeda H, Sakurai Y, Koshinami S, Kumagai Y, Usuda H, Hanita T, Kikuchi A, Saito M. High Expression of Adrenal Cortisol Synthases Is Acquired After Intrauterine Inflammation in Periviable Sheep Fetuses. J Endocr Soc 2023; 7:bvad100. [PMID: 37564887 PMCID: PMC10410294 DOI: 10.1210/jendso/bvad100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Indexed: 08/12/2023] Open
Abstract
Context Intrauterine inflammation, a representative stressor for the fetus, has been shown to alter the hypothalamus-pituitary-adrenal (HPA) axis reactivity in preterm fetuses and increase postnatal cortisol production. However, the mechanism of this alteration has not yet been elucidated. Objective We aimed to clarify the effects of endotoxin-induced intrauterine inflammation on the HPA axis of periviable sheep fetuses. Methods Fetal sheep (0.63 term) were divided into 2 groups: (1) the endotoxin group, in which the endotoxin was injected into the amniotic fluid; and (2) the control group, in which the saline solution was injected instead. A corticotropin-releasing hormone (CRH) challenge test was performed on the third day after injection to evaluate the cortisol-producing capacity of each group. Gene expression levels in the fetal adrenal glands of each group were analyzed by RNA-seq. Results The cortisol levels were significantly higher in the endotoxin group than in the control group after CRH challenge (P = .02). There were no significant differences in the responsiveness of adrenocorticotropin and cortisone between the 2 groups. Gene expression levels of the following enzymes involved in cortisol synthesis were significantly elevated in the endotoxin group: cytochrome P450 family (CYP) 11 subfamily A member 1 (log2FC 1.75), CYP 17 subfamily A member 1 (log2FC 3.41), 3β-hydroxysteroid dehydrogenase type I (log2FC 1.13), steroidogenic acute regulatory protein (log2FC 1.09), and CYP 21 (log2FC 0.89). Conclusion Periviable fetuses exposed to inflammation in utero have altered the responsiveness of the HPA axis with increased expression of enzymes involved in cortisol synthesis in the adrenal gland.
Collapse
Affiliation(s)
- Shinichi Sato
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
- Department of Pediatrics, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Shimpei Watanabe
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
- Department of Pediatrics, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Yuya Saito
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Aika Takanashi
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
- Department of Pediatrics, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Hideyuki Ikeda
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
- Department of Pediatrics, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Yoshie Sakurai
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
- Department of Pediatrics, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Shouta Koshinami
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
- Department of Pediatrics, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Yusaku Kumagai
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Haruo Usuda
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
- Department of Pediatrics, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
- School of Women's and Infants’ Health, University of Western Australia, Perth, Western Australia, Australia
| | - Takushi Hanita
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
- Department of Pediatrics, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Masatoshi Saito
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| |
Collapse
|
4
|
Deer LK, Su C, Thwaites NA, Davis EP, Doom JR. A framework for testing pathways from prenatal stress-responsive hormones to cardiovascular disease risk. Front Endocrinol (Lausanne) 2023; 14:1111474. [PMID: 37223037 PMCID: PMC10200937 DOI: 10.3389/fendo.2023.1111474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/10/2023] [Indexed: 05/25/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death globally, with the prevalence projected to keep rising. Risk factors for adult CVD emerge at least as early as the prenatal period. Alterations in stress-responsive hormones in the prenatal period are hypothesized to contribute to CVD in adulthood, but little is known about relations between prenatal stress-responsive hormones and early precursors of CVD, such as cardiometabolic risk and health behaviors. The current review presents a theoretical model of the relation between prenatal stress-responsive hormones and adult CVD through cardiometabolic risk markers (e.g., rapid catch-up growth, high BMI/adiposity, high blood pressure, and altered blood glucose, lipids, and metabolic hormones) and health behaviors (e.g., substance use, poor sleep, poor diet and eating behaviors, and low physical activity levels). Emerging evidence in human and non-human animal literatures suggest that altered stress-responsive hormones during gestation predict higher cardiometabolic risk and poorer health behaviors in offspring. This review additionally highlights limitations of the current literature (e.g., lack of racial/ethnic diversity, lack of examination of sex differences), and discusses future directions for this promising area of research.
Collapse
Affiliation(s)
- LillyBelle K. Deer
- Department of Psychology, University of Denver, Denver, CO, United States
| | - Chen Su
- Department of Psychology, University of Denver, Denver, CO, United States
| | | | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, United States
- Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA, United States
| | - Jenalee R. Doom
- Department of Psychology, University of Denver, Denver, CO, United States
| |
Collapse
|
5
|
Gultig KD, de Rooij SR, Hilberdink CE, Olff M, Roseboom TJ, van Zuiden M. Effects of prenatal exposure to the 1944-45 Dutch famine and glucocorticoid receptor polymorphisms on later life PTSD susceptibility. Eur J Psychotraumatol 2023; 14:2219075. [PMID: 37335018 PMCID: PMC10281402 DOI: 10.1080/20008066.2023.2219075] [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: 11/24/2022] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 06/21/2023] Open
Abstract
Background: Exposure to adversity in utero is thought to increase susceptibility to develop posttraumatic stress disorder (PTSD) following later life trauma, due to neurobiological programming effects during critical developmental periods. It remains unknown whether effects of prenatal adversity on PTSD susceptibility are modulated by genetic variations in neurobiological pathways implicated in PTSD susceptibility.Objective: We investigated whether genetic variation in the glucocorticoid receptor (GR) modulated effects of prenatal famine exposure on late adulthood PTSD symptom severity after trauma exposure in childhood and mid-to-late adulthood.Method: We included N = 439 term-born singleton adults (mean age: 72 years, 54.2% women) from the Dutch Famine Birth Cohort, born around the time of the Dutch Famine of 1944/1945, divided into exposure and control groups based on timing of the famine during gestation. Participants filled out self-report questionnaires on childhood (Childhood Trauma Questionnaire) and mid-to-late adulthood (Life Events Checklist for DSM-5) trauma, and current PTSD symptom severity (PTSD Checklist for DSM-5). GR haplotypes were determined from four functional GR single nucleotide polymorphisms (ER22/23EK, N363S, BclI and exon 9β) in previously collected DNA. Linear regression analyses were performed to investigate associations of GR haplotype and prenatal famine exposure in conjunction with later life trauma on PTSD symptom severity.Results: We observed a significant three-way interaction between the GR Bcll haplotype, famine exposure during early gestation, and adulthood trauma exposure on PTSD symptom severity in late adulthood. Only participants exposed to famine during early gestation without the GR Bcll haplotype showed a significantly stronger positive association between adulthood trauma and PTSD symptom severity than non-exposed participants, indicating increased PTSD susceptibility.Conclusions: Our results illustrate the importance of integrated approaches considering genetics and environmental contexts throughout various life periods, including the rarely investigated prenatal environment, to elucidate how PTSD susceptibility evolves throughout life.HIGHLIGHTS Adversity during pregnancy is thought to increase offspring's PTSD risk following later life trauma, but exact neurobiological mechanisms underlying this process remain unknown.We found that effects of prenatal famine exposure on PTSD symptom severity were influenced by genetic variation in the glucocorticoid receptor, which signals effects of the stress hormone cortisol.Integrated approaches considering genetics and environmental contexts throughout both early and later life are important to understand how PTSD risk evolves throughout life.
Collapse
Affiliation(s)
- Kayleigh D. Gultig
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Public Health Research Institute and Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
| | - Susanne R. de Rooij
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Public Health Research Institute and Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Charlotte E. Hilberdink
- Departments of Psychiatry and Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
| | - Miranda Olff
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Public Health Research Institute and Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
- ARQ National Psychotrauma Centre, Diemen, the Netherlands
| | - Tessa J. Roseboom
- Departments of Epidemiology and Data Science and Obstetrics and Gynaecology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Mirjam van Zuiden
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Public Health Research Institute and Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
- Department of Clinical Psychology, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
6
|
Trans- and Multigenerational Maternal Social Isolation Stress Programs the Blood Plasma Metabolome in the F3 Generation. Metabolites 2022; 12:metabo12070572. [PMID: 35888696 PMCID: PMC9320469 DOI: 10.3390/metabo12070572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/12/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022] Open
Abstract
Metabolic risk factors are among the most common causes of noncommunicable diseases, and stress critically contributes to metabolic risk. In particular, social isolation during pregnancy may represent a salient stressor that affects offspring metabolic health, with potentially adverse consequences for future generations. Here, we used proton nuclear magnetic resonance (1H NMR) spectroscopy to analyze the blood plasma metabolomes of the third filial (F3) generation of rats born to lineages that experienced either transgenerational or multigenerational maternal social isolation stress. We show that maternal social isolation induces distinct and robust metabolic profiles in the blood plasma of adult F3 offspring, which are characterized by critical switches in energy metabolism, such as upregulated formate and creatine phosphate metabolisms and downregulated glucose metabolism. Both trans- and multigenerational stress altered plasma metabolomic profiles in adult offspring when compared to controls. Social isolation stress increasingly affected pathways involved in energy metabolism and protein biosynthesis, particularly in branched-chain amino acid synthesis, the tricarboxylic acid cycle (lactate, citrate), muscle performance (alanine, creatine phosphate), and immunoregulation (serine, threonine). Levels of creatine phosphate, leucine, and isoleucine were associated with changes in anxiety-like behaviours in open field exploration. The findings reveal the metabolic underpinnings of epigenetically heritable diseases and suggest that even remote maternal social stress may become a risk factor for metabolic diseases, such as diabetes, and adverse mental health outcomes. Metabolomic signatures of transgenerational stress may aid in the risk prediction and early diagnosis of non-communicable diseases in precision medicine approaches.
Collapse
|
7
|
The Molecular Biology of Susceptibility to Post-Traumatic Stress Disorder: Highlights of Epigenetics and Epigenomics. Int J Mol Sci 2021; 22:ijms221910743. [PMID: 34639084 PMCID: PMC8509551 DOI: 10.3390/ijms221910743] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022] Open
Abstract
Exposure to trauma is one of the most important and prevalent risk factors for mental and physical ill-health. Excessive or prolonged stress exposure increases the risk of a wide variety of mental and physical symptoms. However, people differ strikingly in their susceptibility to develop signs and symptoms of mental illness after traumatic stress. Post-traumatic stress disorder (PTSD) is a debilitating disorder affecting approximately 8% of the world’s population during their lifetime, and typically develops after exposure to a traumatic event. Despite that exposure to potentially traumatizing events occurs in a large proportion of the general population, about 80–90% of trauma-exposed individuals do not develop PTSD, suggesting an inter-individual difference in vulnerability to PTSD. While the biological mechanisms underlying this differential susceptibility are unknown, epigenetic changes have been proposed to underlie the relationship between exposure to traumatic stress and the susceptibility to develop PTSD. Epigenetic mechanisms refer to environmentally sensitive modifications to DNA and RNA molecules that regulate gene transcription without altering the genetic sequence itself. In this review, we provide an overview of various molecular biological, biochemical and physiological alterations in PTSD, focusing on changes at the genomic and epigenomic level. Finally, we will discuss how current knowledge may aid us in early detection and improved management of PTSD patients.
Collapse
|
8
|
Crawford AA, Bankier S, Altmaier E, Barnes CLK, Clark DW, Ermel R, Friedrich N, van der Harst P, Joshi PK, Karhunen V, Lahti J, Mahajan A, Mangino M, Nethander M, Neumann A, Pietzner M, Sukhavasi K, Wang CA, Bakker SJL, Bjorkegren JLM, Campbell H, Eriksson J, Gieger C, Hayward C, Jarvelin MR, McLachlan S, Morris AP, Ohlsson C, Pennell CE, Price J, Rudan I, Ruusalepp A, Spector T, Tiemeier H, Völzke H, Wilson JF, Michoel T, Timpson NJ, Smith GD, Walker BR. Variation in the SERPINA6/SERPINA1 locus alters morning plasma cortisol, hepatic corticosteroid binding globulin expression, gene expression in peripheral tissues, and risk of cardiovascular disease. J Hum Genet 2021; 66:625-636. [PMID: 33469137 PMCID: PMC8144017 DOI: 10.1038/s10038-020-00895-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 01/30/2023]
Abstract
The stress hormone cortisol modulates fuel metabolism, cardiovascular homoeostasis, mood, inflammation and cognition. The CORtisol NETwork (CORNET) consortium previously identified a single locus associated with morning plasma cortisol. Identifying additional genetic variants that explain more of the variance in cortisol could provide new insights into cortisol biology and provide statistical power to test the causative role of cortisol in common diseases. The CORNET consortium extended its genome-wide association meta-analysis for morning plasma cortisol from 12,597 to 25,314 subjects and from ~2.2 M to ~7 M SNPs, in 17 population-based cohorts of European ancestries. We confirmed the genetic association with SERPINA6/SERPINA1. This locus contains genes encoding corticosteroid binding globulin (CBG) and α1-antitrypsin. Expression quantitative trait loci (eQTL) analyses undertaken in the STARNET cohort of 600 individuals showed that specific genetic variants within the SERPINA6/SERPINA1 locus influence expression of SERPINA6 rather than SERPINA1 in the liver. Moreover, trans-eQTL analysis demonstrated effects on adipose tissue gene expression, suggesting that variations in CBG levels have an effect on delivery of cortisol to peripheral tissues. Two-sample Mendelian randomisation analyses provided evidence that each genetically-determined standard deviation (SD) increase in morning plasma cortisol was associated with increased odds of chronic ischaemic heart disease (0.32, 95% CI 0.06-0.59) and myocardial infarction (0.21, 95% CI 0.00-0.43) in UK Biobank and similarly in CARDIoGRAMplusC4D. These findings reveal a causative pathway for CBG in determining cortisol action in peripheral tissues and thereby contributing to the aetiology of cardiovascular disease.
Collapse
Affiliation(s)
- Andrew A Crawford
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sean Bankier
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Division of Genetics and Genomics, The Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Elisabeth Altmaier
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Catriona L K Barnes
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - David W Clark
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Raili Ermel
- Department of Cardiac Surgery, Tartu University Hospital, Tartu, Estonia
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475, Greifswald, Germany
- German Center for Cardiovascular Disease (DZHK e.V.), partner site Greifswald, 17475, Greifswald, Germany
| | - Pim van der Harst
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, PO box 30.001, 9700 RB, The Netherlands
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Ville Karhunen
- Department of Epidemiology and Biostatistics, Medical Research Council-Public Health England Centre for Environment and Health, Imperial College London, London, UK
- Centre for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Turku Institute of Advanced Studies, University of Turku, Turku, Finland
| | - Anubha Mahajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College, Lambeth Palace Road, London, SE1 7EH, UK
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, UK
| | - Maria Nethander
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alexander Neumann
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Maik Pietzner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475, Greifswald, Germany
- German Center for Cardiovascular Disease (DZHK e.V.), partner site Greifswald, 17475, Greifswald, Germany
| | | | - Carol A Wang
- School of Medicine and Public Health, Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Johan L M Bjorkegren
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Clinical Gene Networks AB, Stockholm, Sweden
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Johan Eriksson
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Helsinki, Singapore
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital University of Edinburgh, Edinburgh, EH4 2XU, Scotland
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, Medical Research Council-Public Health England Centre for Environment and Health, Imperial College London, London, UK
- Centre for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care and Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Stela McLachlan
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Andrew P Morris
- Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Wellcome Centre for Human genetics, University of Oxford, Oxford, UK
| | - Claes Ohlsson
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Craig E Pennell
- School of Medicine and Public Health, Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Jackie Price
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
| | - Igor Rudan
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Arno Ruusalepp
- Department of Cardiac Surgery, Tartu University Hospital, Tartu, Estonia
- Clinical Gene Networks AB, Stockholm, Sweden
| | - Tim Spector
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, UK
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Social and Behavioural Science, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Walther-Rathenau-Str. 48, 17489, Greifswald, Germany
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, Scotland
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital University of Edinburgh, Edinburgh, EH4 2XU, Scotland
| | - Tom Michoel
- Division of Genetics and Genomics, The Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
- Computational Biology Unit, Department of Informatics, University of Bergen, PO Box 7803, 5020, Bergen, Norway
| | - Nicolas J Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Brian R Walker
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Clinical and Translational Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
9
|
Lalonde C, Grandbois J, Khurana S, Murray A, Tharmalingam S, Tai TC. Late gestational exposure to dexamethasone and fetal programming of abnormal behavior in Wistar Kyoto rats. Brain Behav 2021; 11:e02049. [PMID: 33528889 PMCID: PMC8035474 DOI: 10.1002/brb3.2049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/16/2020] [Accepted: 01/08/2021] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Fetal programming was characterized a few decades ago, explaining the correlation of physiological phenotypes of offspring exposed to early-life stress. High acute or chronic prenatal stress can overwhelm the enzymatic placental barrier, inducing transcriptional changes in the fetus that can result in different adverse behavioral and physiological phenotypes. The current study investigates the impact of exposure to the synthetic glucocorticoid, dexamethasone, during late gestation on behavioral outcomes. METHODS Pregnant Wistar Kyoto rats were given daily subcutaneous injections from gestational days 15-21 of either dexamethasone (0.9% NaCl, 4% EtOH, 100 µg kg-1 day-1 ) or were physically manipulated as naïve controls. Pups were raised normally until 17 weeks of age and underwent the Porsolt swim task and elevated plus maze for depressive and anxiety-like behaviors, respectively. Neural tissue was preserved for genetic analysis using quantitative real-time polymerase chain reaction. RESULTS Statistical analyses show significant disruption of behavior and genetic profiles of offspring exposed to dexamethasone in-utero. Exposed animals spent more time immobile on the swim task and entered open arms of the elevated plus maze more often than their naïve counterparts. In the prefrontal cortex, there was a sex by treatment interaction on gene expression relevant to neural transmission in ryanodine receptor 2, as well as increased gene expression in SNAP25, COMT, and LSAMP in males prenatally exposed to dexamethasone compared with controls. Both dysregulated genes and behavior are linked to decreased anxiety and fear inhibition. CONCLUSION Our results indicate adult offspring exposed to dexamethasone in-utero have a tendency toward passive stress-coping strategies and an inhibition of anxiety on behavioral tasks. Methyltransferase activity, synaptic activity, and cellular processes were disrupted in the prefrontal cortices of these animals. Specifically, genes involved in emotional response pathways were overexpressed, supporting the link between the behavioral and genetic profiles. Combined, we determine that dexamethasone offspring have adaptive predispositions when faced with novel situations, with increased immobility in the swim task and increased exploration on the elevated plus maze.
Collapse
Affiliation(s)
- Christine Lalonde
- Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada.,Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Julie Grandbois
- Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.,Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Sandhya Khurana
- Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada
| | - Alyssa Murray
- Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.,Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Sujeenthar Tharmalingam
- Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada.,Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.,Department of Biology, Laurentian University, Sudbury, ON, Canada.,Department of Chem/Biochem, Laurentian University, Sudbury, ON, Canada
| | - T C Tai
- Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada.,Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.,Department of Biology, Laurentian University, Sudbury, ON, Canada.,Department of Chem/Biochem, Laurentian University, Sudbury, ON, Canada
| |
Collapse
|
10
|
Vaughan G, Kompanijec K, Malik S, Bechard AR. Childhood trauma and post-trauma environment affect fear memory and alcohol use differently in male and female mice. Drug Alcohol Depend 2021; 219:108471. [PMID: 33385691 DOI: 10.1016/j.drugalcdep.2020.108471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Childhood trauma is associated with the development of adult mental health and substance use disorders, with females generally being more at risk. Alcohol is commonly used for coping with trauma, and alcohol use disorder (AUD) affects ∼14.4 million adult Americans annually. Research investigating sex differences in the environmental modification of anxiety and alcohol use following childhood trauma will extend our understanding of the etiology of AUD. Here, we sought to model the interacting effects of a single-episode late childhood trauma with post-trauma environment on adult alcohol use using male and female mice. METHODS C57Bl6/J mice (d22) exposed to predator odor (TMT) or water were reared in standard environments (SE) or environmental enrichment (EE). Mice were assessed for adolescent anxiety and conditioned fear, and for adult alcohol use in a limited access, response non-contingent, alcohol exposure paradigm. RESULTS A single exposure to predator odor was an effective stressor, inducing long-term sex-dependent changes in conditioned fear and alcohol behaviors that interacted with post-trauma environment. Adolescent EE females showed more conditioned freezing to the trauma-associated context. Adult EE mice consumed less total alcohol than SE mice. However, alcohol use across time differed for males and females. Exposure to a childhood stressor increased alcohol use significantly in females, but not males. EE males, but not EE females, drank less than SE counterparts. CONCLUSIONS Findings from this model recapitulate greater vulnerability to childhood trauma in females and support sex differences in post-trauma development of conditioned fear and alcohol use that are modified by environment.
Collapse
Affiliation(s)
- Gavin Vaughan
- Department of Psychology and Neuroscience, SUNY Geneseo, 1 College Circle, Geneseo, NY, 14454, United States.
| | - Katherine Kompanijec
- Department of Psychology and Neuroscience, SUNY Geneseo, 1 College Circle, Geneseo, NY, 14454, United States.
| | - Shreyya Malik
- Department of Psychology and Neuroscience, SUNY Geneseo, 1 College Circle, Geneseo, NY, 14454, United States.
| | - Allison R Bechard
- Department of Psychology and Neuroscience, SUNY Geneseo, 1 College Circle, Geneseo, NY, 14454, United States.
| |
Collapse
|
11
|
Kurek M, Borowska B, Lubowiedzka-Gontarek B, Rosset I, Żądzińska E. Disturbances in primary dental enamel in Polish autistic children. Sci Rep 2020; 10:12751. [PMID: 32728144 PMCID: PMC7391627 DOI: 10.1038/s41598-020-69642-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/15/2020] [Indexed: 01/21/2023] Open
Abstract
Dental enamel is a structure that is formed as a result of the regular functioning of ameloblasts. The knowledge of the patterns of enamel secretion allows an analysis of their disruptions manifested in pronounced additional accentuated lines. These lines represent a physiological response to stress experienced during enamel development. The aim of this study was to assess the occurrence of accentuated lines in the tooth enamel of autistic boys. The width of the neonatal line and the periodicity of the striae of Retzius were also assessed. The study material consisted of longitudinal ground sections of 56 primary teeth (incisors and molars): 22 teeth from autistic children and 34 teeth from the control group. The Mann-Whitney U test indicates that the accentuated lines were found significantly more often in autistic children (Z = 3.03; p = 0.002). No differentiation in the rate of enamel formation and in the rate of regaining homeostasis after childbirth were found. The obtained results may indicate a higher sensitivity of autistic children to stress factors, manifested in more frequent disturbances in the functioning of ameloblasts or may be a reflection of differences in the occurrence of stress factors in the first years of life in both analyzed groups.
Collapse
Affiliation(s)
- Marta Kurek
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland.
| | - Beata Borowska
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland
| | | | - Iwona Rosset
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland
| | - Elżbieta Żądzińska
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland
- Visiting Research Fellow in the School of Medical Sciences, Faculty of Health Sciences, The University of Adelaide, South Australia, 5005, Australia
| |
Collapse
|
12
|
Wilson AE, Michaud SA, Jackson AM, Stenhouse G, Coops NC, Janz DM. Development and validation of protein biomarkers of health in grizzly bears. CONSERVATION PHYSIOLOGY 2020; 8:coaa056. [PMID: 32607241 PMCID: PMC7311831 DOI: 10.1093/conphys/coaa056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/09/2020] [Accepted: 05/31/2020] [Indexed: 06/11/2023]
Abstract
Large carnivores play critical roles in the maintenance and function of natural ecosystems; however, the populations of many of these species are in decline across the globe. Therefore, there is an urgent need to develop novel techniques that can be used as sensitive conservation tools to detect new threats to the health of individual animals well in advance of population-level effects. Our study aimed to determine the expression of proteins related to energetics, reproduction and stress in the skin of grizzly bears (Ursus arctos) using a liquid chromatography and multiple reaction monitoring mass spectrometry assay. We hypothesized that a suite of target proteins could be measured using this technique and that the expression of these proteins would be associated with biological (sex, age, sample location on body) and environmental (geographic area, season, sample year) variables. Small skin biopsies were collected from free-ranging grizzly bears in Alberta, Canada, from 2013 to 2019 (n = 136 samples from 111 individuals). Over 700 proteins were detected in the skin of grizzly bears, 19 of which were chosen as targets because of their established roles in physiological function. Generalized linear mixed model analysis was used for each target protein. Results indicate that sample year influenced the majority of proteins, suggesting that physiological changes may be driven in part by responses to changes in the environment. Season influenced the expression of proteins related to energetics, reproduction and stress, all of which were lower during fall compared to early spring. The expression of proteins related to energetics and stress varied by geographic area, while the majority of proteins that were affected by biological attributes (age class, sex and age class by sex interaction) were related to reproduction and stress. This study provides a novel method by which scientists and managers can further assess and monitor physiological function in wildlife.
Collapse
Affiliation(s)
- Abbey E Wilson
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, 44 Campus Drive, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Sarah A Michaud
- The University of Victoria Genome BC Proteomics Centre, 4464 Markham St #3101, Victoria, British Columbia V8Z 7X8, Canada
| | - Angela M Jackson
- The University of Victoria Genome BC Proteomics Centre, 4464 Markham St #3101, Victoria, British Columbia V8Z 7X8, Canada
| | - Gordon Stenhouse
- Foothills Research Institute, Grizzly Bear Program, 1176 Switzer Drive, Hinton, Alberta T7V 1V3, Canada
| | - Nicholas C Coops
- Department of Forest Resource Management, University of British Columbia, 2424 Main Mall, Vancouver, British Columbia V6T 1Z4, Canada
| | - David M Janz
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, 44 Campus Drive, Saskatoon, Saskatchewan S7N 5B3, Canada
| |
Collapse
|
13
|
Kowalczyk M, Szemraj J, Bliźniewska K, Maes M, Berk M, Su KP, Gałecki P. An immune gate of depression - Early neuroimmune development in the formation of the underlying depressive disorder. Pharmacol Rep 2019; 71:1299-1307. [PMID: 31706254 DOI: 10.1016/j.pharep.2019.05.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 12/14/2022]
Abstract
The prevalence of depression worldwide is increasing from year to year and constitutes a serious medical, economic and social problem. Currently, despite multifactorial risk factors and pathways contributing to depression development, a significant aspect is attributed to the inflammatory process. Cytokines are considered a factor activating the kynurenine pathway, which leads to the exhaustion of tryptophan in the tryptophan catabolite (TRYCAT) pathway. This results in the activation of potentially neuroprogressive processes and also affects the metabolism of many neurotransmitters. The immune system plays a coordinating role in mediating inflammatory process. Beginning from foetal life, dendritic cells have the ability to react to bacterial and viral antigens, stimulating T lymphocytes in a similar way to adult cells. Cytotoxicity in the prenatal period shapes the predisposition to the development of depression in adult life. Allostasis, i.e. the ability to maintain the body's balance in the face of environmental adversity through changes in its behaviour or physiology, allows the organism to survive but its consequences may be unfavourable if it lasts too long. As a result, Th lymphocytes, in particular T helper 17 cells, which play a central role in the immunity of the whole body, contribute to the development of both autoimmune diseases and psychiatric disorders including depression, as well as have an impact on the differentiation of T CD4+ cells into Th17 cells in the later development of the child's organism, which confirms the importance of the foetal period for the progression of depressive disorders.
Collapse
Affiliation(s)
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Łódź, Poland
| | | | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Berk
- IMPACT Strategic Research Centre, Deakin University School of Medicine, and Barwon Health, Geelong, VIC, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Kuan-Pin Su
- Department of Psychiatry and Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
| | - Piotr Gałecki
- Department of Adult Psychiatry, Medical University of Lodz, Łódź, Poland
| |
Collapse
|
14
|
Yehuda R, Lehrner A. Intergenerational transmission of trauma effects: putative role of epigenetic mechanisms. World Psychiatry 2018; 17:243-257. [PMID: 30192087 PMCID: PMC6127768 DOI: 10.1002/wps.20568] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 12/18/2022] Open
Abstract
This paper reviews the research evidence concerning the intergenerational transmission of trauma effects and the possible role of epigenetic mechanisms in this transmission. Two broad categories of epigenetically mediated effects are highlighted. The first involves developmentally programmed effects. These can result from the influence of the offspring's early environmental exposures, including postnatal maternal care as well as in utero exposure reflecting maternal stress during pregnancy. The second includes epigenetic changes associated with a preconception trauma in parents that may affect the germline, and impact fetoplacental interactions. Several factors, such as sex-specific epigenetic effects following trauma exposure and parental developmental stage at the time of exposure, explain different effects of maternal and paternal trauma. The most compelling work to date has been done in animal models, where the opportunity for controlled designs enables clear interpretations of transmissible effects. Given the paucity of human studies and the methodological challenges in conducting such studies, it is not possible to attribute intergenerational effects in humans to a single set of biological or other determinants at this time. Elucidating the role of epigenetic mechanisms in intergenerational effects through prospective, multi-generational studies may ultimately yield a cogent understanding of how individual, cultural and societal experiences permeate our biology.
Collapse
Affiliation(s)
- Rachel Yehuda
- James J. Peters Bronx Veterans Affairs Hospital, Bronx, NY, USA
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amy Lehrner
- James J. Peters Bronx Veterans Affairs Hospital, Bronx, NY, USA
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
15
|
The effect of gestational period on the association between maternal prenatal salivary cortisol and birth weight: A systematic review and meta-analysis. Psychoneuroendocrinology 2018; 94:49-62. [PMID: 29754005 DOI: 10.1016/j.psyneuen.2018.04.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/21/2018] [Accepted: 04/23/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Studies exploring the relations between maternal stress and fetal development show an association between increased maternal stress and adverse birth outcomes. A frequently proposed mechanism linking maternal prenatal stress and adverse birth outcomes is heightened concentrations of maternal cortisol. To date, studies exploring this association have reported conflicting results because of the diverse approaches taken to measuring cortisol and the wide variety of possible birth outcomes explored. To add clarity to the growing body of literature, this systematic review and meta-analysis reports empirical findings on the association between maternal prenatal salivary cortisol and newborn birth weight. METHODS Searches for relevant papers published up until November 2017 were run in MEDLINE, EMBASE, PsycINFO, and CINAHL. Non-English language papers were included and experts were contacted when necessary. We included data from human observational studies that were designed or had an underlying intention to measure maternal prenatal salivary cortisol and newborn birth weight. We only included data from measurements of salivary cortisol to prevent rendering of the review unsuitable for meta-analysis. Two independent reviewers assessed study eligibility and quality. For every maternal-fetal dyad, an area under the curve with respect to ground (AUCg) of maternal cortisol was calculated to determine a Pearson's correlation coefficient with a continuous measure of newborn birth weight. Correlation coefficients were then pooled across all stages of gestation. To examine if there are critical gestational periods in which the fetus may be more susceptible to elevated concentration of maternal salivary cortisol, a meta-analysis was performed on separate correlations calculated from gestational trimesters. RESULTS Nine studies with a total of 1606 maternal-fetal dyads demonstrated a negative correlation between pooled maternal salivary cortisol and birth weight (-0.24, 95% CI -0.28 to -0.20), but there was a high degree of heterogeneity between studies (I2 = 88.9%). To investigate heterogeneity, subgroup analysis by trimester of the pooled correlation between salivary cortisol and birth weight was performed with the following correlations found: first trimester, -0.18 (95% CI -0.32 to -0.03, I2 = 97.3%); second trimester, -0.20 (95% CI -0.28 to -0.12, I2 = 98.3%); and third trimester, -0.30 (95% CI -0.33 to -0.26, I2 = 85.4%). DISCUSSION A consistently negative association was observed between maternal cortisol and infant birth weight. The review highlights specific gaps in the literature on the relationship between maternal prenatal salivary cortisol and newborn birth weight. Although a significant negative correlation was found, substantial heterogeneity of effects and the likelihood of publication bias exist. The third trimester was revealed as a possible critical gestational period for heightened maternal cortisol concentration to affect birth weight. Challenges faced in this body of research and recommendations for future research are discussed.
Collapse
|
16
|
Lotero Osorio HD, Villa González IC, Torres Trujillo LE. Afectividad y Apoyo Social Percibido en Mujeres Gestantes: un Análisis Comparativo. REVISTA COLOMBIANA DE PSICOLOGÍA 2018. [DOI: 10.15446/rcp.v27n2.65584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
El propósito de esta investigación es comparar las manifestaciones afectivas en sus dimensiones positivas y negativas, a modo de rasgo y de estado, con variables ginecobstétricas y de funcionamiento familiar en mujeres gestantes. Se seleccionaron a conveniencia 229 mujeres que ingresaron a una institución prestadora de servicios de salud en Medellín. Se utilizaron los cuestionarios PANAS (Robles & Páez, 2003), APGAR-familiar (Smilkstein, 1978) y una encuesta de variables sociodemográficas. Se encontró que las gestantes con mayor rasgo afectivo positivo presentan menor riesgo obstétrico, mientras que las mujeres con mayor rasgo afectivo negativo demuestran menor planeación del embarazo y mayor percepción de disfunción familiar. Se concluye que la percepción de apoyo familiar y los rasgos emocionales positivos son componentes que favorecen la salud gestacional, mientras que el afecto negativo es un obstáculo para la adaptación a la maternidad y las relaciones familiares percibidas.
Collapse
|
17
|
Xu YJ, Sheng H, Wu TW, Bao QY, Zheng Y, Zhang YM, Gong YX, Lu JQ, You ZD, Xia Y, Ni X. CRH/CRHR1 mediates prenatal synthetic glucocorticoid programming of depression-like behavior across 2 generations. FASEB J 2018. [PMID: 29543532 DOI: 10.1096/fj.201700948rr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pregnant women at risk of preterm labor usually receive synthetic glucocorticoids (sGCs) to promote fetal lung development. Emerging evidence indicates that antenatal sGC increases the risk of affective disorders in offspring. Data from animal studies show that such disorders can be transmitted to the second generation. However, the molecular mechanisms underlying the intergenerational effects of prenatal sGC remain largely unknown. Here we show that prenatal dexamethasone (Dex) administration in late pregnancy induced depression-like behavior in first-generation (F1) offspring, which could be transmitted to second-generation (F2) offspring with maternal dependence. Moreover, corticotropin-releasing hormone (CRH) and CRH receptor type 1 (CRHR1) expression in the hippocampus was increased in F1 Dex offspring and F2 offspring from F1 Dex female rats. Administration of a CRHR1 antagonist to newborn F1 Dex offspring alleviated depression-like behavior in these rats at adult. Furthermore, we demonstrated that increased CRHR1 expression in F1 and F2 offspring was associated with hypomethylation of CpG islands in Crhr1 promoter. Our results revealed that prenatal sGC exposure could program Crh and Crhr1 gene expression in hippocampus across 2 generations, thereby leading to depression-like behavior. Our study indicates that prenatal sGC can cause epigenetic instability, which increases the risk of disease development in the offspring's later life.-Xu, Y.-J., Sheng, H., Wu, T.-W., Bao, Q.-Y., Zheng, Y., Zhang, Y.-M., Gong, Y.-X., Lu, J.-Q., You, Z.-D., Xia, Y., Ni, X. CRH/CRHR1 mediates prenatal synthetic glucocorticoid programming of depression-like behavior across 2 generations.
Collapse
Affiliation(s)
- Yong-Jun Xu
- Department of Physiology, Second Military Medical University, Shanghai, China.,Department of Clinical Genetics and Experimental Medicine, Fuzhou General Hospital, Xiamen University School of Medicine, Fuzhou, China
| | - Hui Sheng
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Tian-Wen Wu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Qing-Yue Bao
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - You Zheng
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Yan-Min Zhang
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Yu-Xiang Gong
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences, Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Jian-Qiang Lu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences, Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Zhen-Dong You
- Department of Neurobiology, Second Military Medical University, Shanghai, China
| | - Yang Xia
- Department of Physiology, Second Military Medical University, Shanghai, China.,Department of Biochemistry and Molecular Biology, The University of Texas Medical School at Houston, Houston, Texas, USA.,Institute of Molecular Metabolomics, Xiangya Hospital, Changsha, China
| | - Xin Ni
- Department of Physiology, Second Military Medical University, Shanghai, China.,Institute of Molecular Metabolomics, Xiangya Hospital, Changsha, China
| |
Collapse
|
18
|
Jacobs S, Moxley K, Womersley JS, Spies G, Hemmings SM, Seedat S. HPA-axis genes as potential risk variants for neurocognitive decline in trauma-exposed, HIV-positive females. Neuropsychiatr Dis Treat 2018; 14:2497-2504. [PMID: 30319260 PMCID: PMC6167976 DOI: 10.2147/ndt.s166992] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
PURPOSE Previous studies have independently provided evidence for the effects of HIV infection, hypothalamic-pituitary-adrenal (HPA) axis dysfunction and early life trauma on neurocognitive impairment (NCI). This study examined the interaction between single-nucleotide polymorphisms (SNPs) of two HPA axis genes, corticotrophin-releasing hormone receptor 1 (CRHR1; rs110402, rs242924, rs7209436, and rs4792888) and corticotrophin-releasing hormone-binding protein (CRHBP; rs32897, rs10062367, and rs1053989), childhood trauma, and HIV-associated NCI. PATIENTS AND METHODS The sample comprised 128 HIV-positive Xhosa females of whom 88 (69%) had a history of childhood trauma. NCI was assessed using a battery of 17 measures sensitive to the effects of HIV, and the history of childhood trauma was assessed using the validated retrospective Childhood Trauma Questionnaire-Short Form. Generalized linear regression models were used to compare allelic distribution by trauma status and global NCI. The association between genotype, childhood trauma, and cognitive scores was also evaluated using generalized linear regression models, assuming additive models for the SNPs, and ANOVA. RESULTS Of the seven polymorphisms assessed, only the rs10062367 variant of CRHBP was significantly associated with global NCI (P=0.034), independent of childhood trauma. This polymorphism was not significantly associated with z-scores on any specific cognitive domain. The interaction of childhood trauma and variants of CRHR1 was associated with poorer learning (rs110402) and/or recall (rs110402 and rs4792888). CONCLUSION These findings suggest that CRHBP rs10062367 A allele is a possible risk variant for NCI in HIV, independent of childhood trauma. Furthermore, results show that the interaction of childhood trauma with variants of CRHR1, rs110402 and rs4792888, confer added vulnerability to NCI in HIV-infected individuals in cognitive domains that are known to be impacted by HIV. While these findings need independent replication in larger samples, it adds CRHBP and CRHR1 to the list of known genes linked to HIV- and childhood trauma-associated neurocognitive phenotypes.
Collapse
Affiliation(s)
- Sean Jacobs
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa,
| | - Karis Moxley
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa,
| | - Jacqueline S Womersley
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa,
| | - Georgina Spies
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa,
| | - Sian Mj Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa,
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa,
| |
Collapse
|
19
|
Thayer ZM. Dark shadow of the long white cloud: Neighborhood safety is associated with self-rated health and cortisol during pregnancy in Auckland, Aotearoa/New Zealand. SSM Popul Health 2017; 3:75-80. [PMID: 29349206 PMCID: PMC5768991 DOI: 10.1016/j.ssmph.2016.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 11/04/2016] [Accepted: 11/21/2016] [Indexed: 12/25/2022] Open
Abstract
Auckland, Aotearoa/New Zealand is a culturally and ethnically diverse city. Despite popular global conceptions regarding its utopian nature, the lived experience for many individuals in Auckland attests to the substantial social, economic, and health inequalities that exist there. In particular, rapidly rising home prices constrain housing decisions and force individuals to live in less desirable neighborhoods, with potential impacts on individual health. One of the pathways through which adverse neighborhood conditions could impact health is through alterations in the functioning of the hypothalamic pituitary adrenal (HPA)-axis, which regulates the physiological stress response. This paper evaluates the relationship between perceived neighborhood safety, self-rated health, and cortisol, an end product of HPA-axis activation, among women in late pregnancy. Pregnant women living in neighborhoods where they were concerned about safety of their property had poorer self-rated health and elevated morning cortisol, even after adjusting for maternal age, material deprivation, and ethnicity. However, fear of personal safety was unrelated to self-rated health and cortisol. These results suggest that maternal health in pregnancy is sensitive to perceptions regarding neighborhood safety. Such findings are important since higher cortisol levels in pregnancy could not only influence maternal health, but also the health and development of women's children.
Collapse
|
20
|
Paternal transmission of early life traumatization through epigenetics: Do fathers play a role? Med Hypotheses 2017; 109:59-64. [DOI: 10.1016/j.mehy.2017.09.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/23/2017] [Accepted: 09/15/2017] [Indexed: 12/12/2022]
|
21
|
Howland MA, Sandman CA, Glynn LM. Developmental origins of the human hypothalamic-pituitary-adrenal axis. Expert Rev Endocrinol Metab 2017; 12:321-339. [PMID: 30058893 PMCID: PMC6334849 DOI: 10.1080/17446651.2017.1356222] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The developmental origins of disease or fetal programming model predicts that intrauterine exposures have life long consequences for physical and psychological health. Prenatal programming of the fetal hypothalamic-pituitary-adrenal (HPA) axis is proposed as a primary mechanism by which early experiences are linked to later disease risk. Areas covered: This review describes the development of the fetal HPA axis, which is determined by an intricately timed cascade of endocrine events during gestation and is regulated by an integrated maternal-placental-fetal steroidogenic unit. Mechanisms by which stress-induced elevations in hormones of maternal, fetal, or placental origin influence the structure and function of the emerging fetal HPA axis are discussed. Recent prospective studies documenting persisting associations between prenatal stress exposures and altered postnatal HPA axis function are summarized, with effects observed beginning in infancy into adulthood. Expert commentary: The results of these studies are synthesized, and potential moderating factors are discussed. Promising areas of further research highlighted include epigenetic mechanisms and interactions between pre and postnatal influences.
Collapse
Affiliation(s)
- Mariann A. Howland
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA
| | - Curt A. Sandman
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA
| | - Laura M. Glynn
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA
- Department of Psychology, Chapman University, Orange, CA, USA
| |
Collapse
|
22
|
Crittenden PM, Heller MB. The Roots of Chronic Posttraumatic Stress Disorder: Childhood Trauma, Information Processing, and Self-protective Strategies. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2017; 1:2470547016682965. [PMID: 32440576 PMCID: PMC7219921 DOI: 10.1177/2470547016682965] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/28/2016] [Accepted: 11/07/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Although childhood endangerment often precedes adult posttraumatic stress disorder (PTSD), the mechanism from danger to disorder is unclear. We proposed a developmental process in which unprotected and uncomforted danger in childhood would be associated with "shortcuts" in information processing that, in adulthood, could result in PTSD if the adult experienced additional exposure to danger. Information processing was defined as the basic associative, dissociative, and integrative processes used by all humans. Individual differences in parents' (or primary caregivers') protective and comforting behavior were expected to force unprotected children to use psychological shortcuts that linked early trauma to later vulnerability for PTSD. METHOD We compared 22 adults with chronic PTSD to (a) 22 adults with other psychiatric diagnoses and (b) 22 normative adults without any diagnosis, in terms of information processing around childhood danger. The Adult Attachment Interview was used to derive information processing variables, including self-protective strategies, childhood traumas, and depression. RESULTS The two patient groups differed from the normative group on all variables. Adults with chronic PTSD differed from other psychiatric patients in having more childhood traumas and using more transformations of associative and dissociative processes. Within the PTSD group, there were three psychologically different subgroups. CONCLUSION Our findings suggest that (1) prediction of risk for adult PTSD may be possible, (2) treatment might be facilitated by provision of a protective and supportive therapist, (3) who included a focus on correction of information processing errors and use of more adaptive strategies, and (4) subgroups of adults with PTSD may require different forms of treatment.
Collapse
|
23
|
Koenen KC, Sumner JA, Gilsanz P, Glymour MM, Ratanatharathorn A, Rimm EB, Roberts AL, Winning A, Kubzansky LD. Post-traumatic stress disorder and cardiometabolic disease: improving causal inference to inform practice. Psychol Med 2017; 47:209-225. [PMID: 27697083 PMCID: PMC5214599 DOI: 10.1017/s0033291716002294] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Post-traumatic stress disorder (PTSD) has been declared 'a life sentence' based on evidence that the disorder leads to a host of physical health problems. Some of the strongest empirical research - in terms of methodology and findings - has shown that PTSD predicts higher risk of cardiometabolic diseases, specifically cardiovascular disease (CVD) and type 2 diabetes (T2D). Despite mounting evidence, PTSD is not currently acknowledged as a risk factor by cardiovascular or endocrinological medicine. This view is unlikely to change absent compelling evidence that PTSD causally contributes to cardiometabolic disease. This review suggests that with developments in methods for epidemiological research and the rapidly expanding knowledge of the behavioral and biological effects of PTSD the field is poised to provide more definitive answers to questions of causality. First, we discuss methods to improve causal inference using the observational data most often used in studies of PTSD and health, with particular reference to issues of temporality and confounding. Second, we consider recent work linking PTSD with specific behaviors and biological processes, and evaluate whether these may plausibly serve as mechanisms by which PTSD leads to cardiometabolic disease. Third, we evaluate how looking more comprehensively into the PTSD phenotype provides insight into whether specific aspects of PTSD phenomenology are particularly relevant to cardiometabolic disease. Finally, we discuss new areas of research that are feasible and could enhance understanding of the PTSD-cardiometabolic relationship, such as testing whether treatment of PTSD can halt or even reverse the cardiometabolic risk factors causally related to CVD and T2D.
Collapse
Affiliation(s)
- K C Koenen
- Department of Epidemiology,Harvard T.H. Chan School of Public Health,Boston, MA,USA
| | - J A Sumner
- Department of Epidemiology,Harvard T.H. Chan School of Public Health,Boston, MA,USA
| | - P Gilsanz
- Department of Social and Behavioral Sciences,Harvard T.H. Chan School of Public Health,Boston, MA,USA
| | - M M Glymour
- Department of Social and Behavioral Sciences,Harvard T.H. Chan School of Public Health,Boston, MA,USA
| | - A Ratanatharathorn
- Department of Epidemiology,Harvard T.H. Chan School of Public Health,Boston, MA,USA
| | - E B Rimm
- Channing Division of Network Medicine,Brigham and Women's Hospital,Harvard Medical School and Departments of Epidemiology and Nutrition,Harvard T.H. Chan School of Public Health,Boston, MA,USA
| | - A L Roberts
- Department of Social and Behavioral Sciences,Harvard T.H. Chan School of Public Health,Boston, MA,USA
| | - A Winning
- Department of Social and Behavioral Sciences,Harvard T.H. Chan School of Public Health,Boston, MA,USA
| | - L D Kubzansky
- Department of Social and Behavioral Sciences,Harvard T.H. Chan School of Public Health,Boston, MA,USA
| |
Collapse
|
24
|
Prokai D, Berga SL. Neuroprotection via Reduction in Stress: Altered Menstrual Patterns as a Marker for Stress and Implications for Long-Term Neurologic Health in Women. Int J Mol Sci 2016; 17:ijms17122147. [PMID: 27999413 PMCID: PMC5187947 DOI: 10.3390/ijms17122147] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/07/2016] [Accepted: 12/13/2016] [Indexed: 01/30/2023] Open
Abstract
Individuals under chronic psychological stress can be difficult to identify clinically. There is often no outwardly visible phenotype. Chronic stress of sufficient magnitude not only impacts reproductive function, but also concomitantly elicits a constellation of neuroendocrine changes that may accelerate aging in general and brain aging in particular. Functional hypothalamic amenorrhea, a phenotypically recognizable form of stress, is due to stress-induced suppression of endogenous gonadotropin-releasing hormone secretion. Reversal of functional hypothalamic amenorrhea includes restoration of ovulatory ovarian function and fertility and amelioration of hypercortisolism and hypothyroidism. Taken together, recovery from functional hypothalamic amenorrhea putatively offers neuroprotection and ameliorates stress-induced premature brain aging and possibly syndromic Alzheimer’s disease. Amenorrhea may be viewed as a sentinel indicator of stress. Hypothalamic hypogonadism is less clinically evident in men and the diagnosis is difficult to establish. Whether there are other sex differences in the impact of stress on brain aging remains to be better investigated, but it is likely that both low estradiol from stress-induced anovulation and low testosterone from stress-induced hypogonadism compromise brain health.
Collapse
Affiliation(s)
- David Prokai
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | - Sarah L Berga
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
25
|
Affiliation(s)
- Connie J. Mulligan
- Department of Anthropology, Genetics Institute, University of Florida, Gainesville, Florida 32610-3610;
| |
Collapse
|
26
|
Jin ZL, Liu JX, Liu X, Zhang LM, Ran YH, Zheng YY, Tang Y, Li YF, Xiong J. Anxiolytic effects of GLYX-13 in animal models of posttraumatic stress disorder-like behavior. J Psychopharmacol 2016; 30:913-21. [PMID: 27147594 DOI: 10.1177/0269881116645298] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the present study, we investigated the effectiveness of GLYX-13, an NMDA receptor glycine site functional partial agonist, to alleviate the enhanced anxiety and fear response in both a mouse and rat model of stress-induced behavioral changes that might be relevant to posttraumatic stress disorder (PTSD). Studies over the last decades have suggested that the hyperactivity of hypothalamic-pituitary-adrenal (HPA) axis is one of the most consistent findings in stress-related disease. Herein, we used these animal models to further investigate the effect of GLYX-13 on the stress hormone levels and glucocorticoid receptor (GR) expression. We found that exposure to foot shock induced long-lasting behavioral deficiencies in mice, including freezing and anxiety-like behaviors, that were significantly ameliorated by the long-term administration of GLYX-13 (5 or 10 mg/kg). Our enzyme-linked immunosorbent assay results showed that long-term administration of GLYX-13 at behaviorally effective doses (5 or 10 mg/kg) significantly decreased the elevated serum levels of both corticosterone and its upstream stress hormone adrenocorticotropic hormone in rats subjected to the TDS procedure. These results suggest that GLYX-13 exerts a therapeutic effect on PTSD-like stress responding that is accompanied by (or associated with) modulation of the HPA axis, including inhibition of stress hormone levels and upregulation of hippocampal GR expression.
Collapse
Affiliation(s)
- Zeng-Liang Jin
- Department of Pharmacology, Capital Medical University, Beijing, P.R. China
| | - Jin-Xu Liu
- Pingdu People's Hospital, Qingdao, P.R. China
| | - Xu Liu
- Department of Pharmacy, General Hospital of Chinese People's Armed Police Forces, Beijing, P.R. China
| | - Li-Ming Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing, P.R. China
| | - Yu-Hua Ran
- Beijing Institute of Pharmacology and Toxicology, Beijing, P.R. China
| | - Yuan-Yuan Zheng
- Department of Pharmacology, Capital Medical University, Beijing, P.R. China
| | - Yu Tang
- Department of Pharmacology, Capital Medical University, Beijing, P.R. China
| | - Yun-Feng Li
- Beijing Institute of Pharmacology and Toxicology, Beijing, P.R. China
| | - Jie Xiong
- Department of Pharmacology, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
27
|
Hiroi R, Carbone DL, Zuloaga DG, Bimonte-Nelson HA, Handa RJ. Sex-dependent programming effects of prenatal glucocorticoid treatment on the developing serotonin system and stress-related behaviors in adulthood. Neuroscience 2016; 320:43-56. [PMID: 26844389 PMCID: PMC4840233 DOI: 10.1016/j.neuroscience.2016.01.055] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 01/12/2016] [Accepted: 01/26/2016] [Indexed: 12/25/2022]
Abstract
Prenatal stress and overexposure to glucocorticoids (GC) during development may be associated with an increased susceptibility to a number of diseases in adulthood including neuropsychiatric disorders, such as depression and anxiety. In animal models, prenatal overexposure to GC results in hyper-responsiveness to stress in adulthood, and females appear to be more susceptible than males. Here, we tested the hypothesis that overexposure to GC during fetal development has sex-specific programming effects on the brain, resulting in altered behaviors in adulthood. We examined the effects of dexamethasone (DEX; a synthetic GC) during prenatal life on stress-related behaviors in adulthood and on the tryptophan hydroxylase-2 (TpH2) gene expression in the adult dorsal raphe nucleus (DRN). TpH2 is the rate-limiting enzyme for serotonin (5-HT) synthesis and has been implicated in the etiology of human affective disorders. Timed-pregnant rats were treated with DEX from gestational days 18-22. Male and female offspring were sacrificed on the day of birth (postnatal day 0; P0), P7, and in adulthood (P80-84) and brains were examined for changes in TpH2 mRNA expression. Adult animals were also tested for anxiety- and depressive- like behaviors. In adulthood, prenatal DEX increased anxiety- and depressive- like behaviors selectively in females, as measured by decreased time spent in the center of the open field and increased time spent immobile in the forced swim test, respectively. Prenatal DEX increased TpH2 mRNA selectively in the female caudal DRN at P7, whereas it decreased TpH2 mRNA selectively in the female caudal DRN in adulthood. In animals challenged with restraint stress in adulthood, TpH2 mRNA was significantly lower in rostral DRN of prenatal DEX-treated females compared to vehicle-treated females. These data demonstrated that prenatal overexposure to GC alters the development of TpH2 gene expression and these alterations correlated with lasting behavioral changes found in adult female offspring.
Collapse
Affiliation(s)
- R Hiroi
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA; Department of Psychology, Arizona State University, 950 S. McAllister Avenue, Tempe, AZ 85287, USA.
| | - D L Carbone
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA.
| | - D G Zuloaga
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA.
| | - H A Bimonte-Nelson
- Department of Psychology, Arizona State University, 950 S. McAllister Avenue, Tempe, AZ 85287, USA.
| | - R J Handa
- Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, 550 E. Van Buren Street, Phoenix, AZ 85004, USA.
| |
Collapse
|
28
|
Klengel T, Dias BG, Ressler KJ. Models of Intergenerational and Transgenerational Transmission of Risk for Psychopathology in Mice. Neuropsychopharmacology 2016; 41:219-31. [PMID: 26283147 PMCID: PMC4677139 DOI: 10.1038/npp.2015.249] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 08/03/2015] [Accepted: 08/12/2015] [Indexed: 01/03/2023]
Abstract
Trajectories toward risk or resilience in psychiatric disorders are influenced by acquired and inherited factors. More recently, evidence from rodent studies suggest that acquired risk factors can be transmitted through non-genomic, epigenetic mechanisms to subsequent generations, potentially contributing to a cycle of disease and disease risk. Here, we review examples of transmission of environmental factors across generations and illustrate the difference between behavioral transmission and epigenetic inheritance. We highlight essential definitions of intergenerational and transgenerational transmission of disease risk with corresponding examples. We then explore how these phenomena may influence our understanding of psychiatric disorders leading toward new prevention and therapeutic approaches.
Collapse
Affiliation(s)
- Torsten Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA,Department of Psychiatry and Behavioral Sciences and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Brian G Dias
- Department of Psychiatry and Behavioral Sciences and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA,Department of Psychiatry and Behavioral Sciences and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA,Howard Hughes Medical Institute, Chevy Chase, MD, USA,Department of Psychiatry, Harvard Medical School, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA, Tel: +1 404 727 7739, Fax: +1 404 727 8070, E-mail:
| |
Collapse
|
29
|
Moloney RD, Stilling RM, Dinan TG, Cryan JF. Early-life stress-induced visceral hypersensitivity and anxiety behavior is reversed by histone deacetylase inhibition. Neurogastroenterol Motil 2015; 27:1831-6. [PMID: 26403543 DOI: 10.1111/nmo.12675] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 08/20/2015] [Indexed: 02/08/2023]
Abstract
Stressful life events, especially in childhood, can have detrimental effects on health and are associated with a host of psychiatric and gastrointestinal disorders including irritable bowel syndrome (IBS). Early-life stress can be recapitulated in animals using the maternal separation (MS) model, exhibiting many key phenotypic outcomes including visceral hypersensitivity and anxiety-like behaviors. The molecular mechanisms of MS are unclear, but recent studies point to a role for epigenetics. Histone acetylation is a key epigenetic mark that is altered in numerous stress-related disease states. Here, we investigated the role of histone acetylation in early-life stress-induced visceral hypersensitivity. Interestingly, increased number of pain behaviors and reduced threshold of visceral sensation were associated with alterations in histone acetylation in the lumbosacral spinal cord, a key region in visceral pain processing. Moreover, we also investigated whether the histone deacetylase (HDAC) inhibitor, suberoylanilide hydroxamic acid (SAHA), could reverse early-life stress-induced visceral hypersensitivity and stress-induced fecal pellet output in the MS model. Significantly, SAHA reversed both of these parameters. Taken together, these data describe, for the first time, a key role of histone acetylation in the pathophysiology of early-life stress-induced visceral hypersensitivity in a well-established model of IBS. These findings will inform new research aimed at the development of novel pharmaceutical approaches targeting the epigenetic machinery for novel anti-IBS drugs.
Collapse
Affiliation(s)
- R D Moloney
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - R M Stilling
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - T G Dinan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - J F Cryan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
30
|
The impact of early postnatal environmental enrichment on maternal care and offspring behaviour following weaning. Behav Processes 2015; 122:51-8. [PMID: 26562657 DOI: 10.1016/j.beproc.2015.11.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/27/2015] [Accepted: 11/05/2015] [Indexed: 11/20/2022]
Abstract
The early postnatal period is a sensitive period in rodents as behavioural systems are developing and maturing during this time. However, relatively little information is available about the impact of environmental enrichment on offspring behaviour if enrichment is implemented only during this period. Here, environmental enrichment was provided from postnatal day 1 until weaning. On post-natal day 9, maternal behaviour and nonmaternal behaviour of the dam was observed. Nursing time in the enriched group was reduced but dams showed more non-maternal appetitive behaviours. Offspring were exposed to either the open field or the elevated plus maze (EPM) after weaning. In the open field, rats from the enriched group approached the more aversive inner zone of the open field later than control rats. Offspring from the enriched group made fewer entries into the inner zone and spent less time in this part of the arena. Enrichment had no impact on behaviour in the EPM. The present study provides evidence that postnatal enrichment can interfere with maternal behaviour in rats and can possibly lead to increased anxiety in the offspring. The findings suggest that enrichment procedures can have potentially unintended effects, interfering with the development of emotional behaviours in rats.
Collapse
|
31
|
Rotstein M, Stolar O, Uliel S, Mandel D, Mani A, Dollberg S, Reifen R, Steiner JE, Harel S, Leitner Y. Facial Expression in Response to Smell and Taste Stimuli in Small and Appropriate for Gestational Age Newborns. J Child Neurol 2015; 30:1466-71. [PMID: 25694467 DOI: 10.1177/0883073815570153] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 12/11/2014] [Indexed: 11/17/2022]
Abstract
Small for gestational age newborns can later suffer from eating difficulties and slow growth. Nutritional preferences can be influenced by changes in sensory perception of smell and taste. To determine whether these could be detected at birth, the authors examined the different recognition pattern of smell and taste in small for gestational age newborns compared to appropriate for gestational age controls, as expressed by gusto-facial and naso-facial reflexes. The authors performed video analysis of facial expressions of 10 small for gestational age and 12 control newborns exposed to various tastes and smells. No difference in the facial recognition patterns for taste or smell was demonstrated between small for gestational age and controls, except for perception of distilled water. Newborns show recognizable patterns of facial expression in response to taste and smell stimuli. Perception of taste and smell in small for gestational age newborns is not different from controls, as measured by the method of facial recognition.
Collapse
Affiliation(s)
- Michael Rotstein
- Institute for Child Development and Pediatric Neurology Unit, Tel Aviv Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Orit Stolar
- Institute for Child Development and Pediatric Neurology Unit, Tel Aviv Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Shimrit Uliel
- Institute for Child Development and Pediatric Neurology Unit, Tel Aviv Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Dror Mandel
- Lis Maternity Hospital, Tel Aviv Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Ariel Mani
- Lis Maternity Hospital, Tel Aviv Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Shaul Dollberg
- Lis Maternity Hospital, Tel Aviv Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Ram Reifen
- School of Nutritional Sciences, Hebrew University of Jerusalem, Rehovot, Israel
| | - Jacob E Steiner
- School of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shaul Harel
- Institute for Child Development and Pediatric Neurology Unit, Tel Aviv Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Yael Leitner
- Institute for Child Development and Pediatric Neurology Unit, Tel Aviv Medical Center and Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
32
|
Ramo-Fernández L, Schneider A, Wilker S, Kolassa IT. Epigenetic Alterations Associated with War Trauma and Childhood Maltreatment. BEHAVIORAL SCIENCES & THE LAW 2015; 33:701-721. [PMID: 26358541 DOI: 10.1002/bsl.2200] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Survivors of war trauma or childhood maltreatment are at increased risk for trauma-spectrum disorders such as post-traumatic stress disorder (PTSD). In addition, traumatic stress has been associated with alterations in the neuroendocrine and the immune system, enhancing the risk for physical diseases. Traumatic experiences might even affect psychological as well as biological parameters in the next generation, i.e. traumatic stress might have transgenerational effects. This article outlines how epigenetic processes, which represent a pivotal biological mechanism for dynamic adaptation to environmental challenges, might contribute to the explanation of the long-lasting and transgenerational effects of trauma. In particular, epigenetic alterations in genes regulating the hypothalamus-pituitary-adrenal axis as well as the immune system have been observed in survivors of childhood and adult trauma. These changes could result in enduring alterations of the stress response as well as the physical health risk. Furthermore, the effects of parental trauma could be transmitted to the next generation by parental distress and the pre- and postnatal environment, as well as by epigenetic marks transmitted via the germline. While epigenetic research has a high potential of advancing our understanding of the consequences of trauma, the findings have to be interpreted with caution, as epigenetics only represent one piece of a complex puzzle of interacting biological and environmental factors. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | - Anna Schneider
- Clinical and Biological Psychology, Ulm University, Germany
| | - Sarah Wilker
- Clinical and Biological Psychology, Ulm University, Germany
| | | |
Collapse
|
33
|
Sexually dimorphic effects of maternal dietary protein restriction on fetal growth and placental expression of 11β-HSD2 in the pig. Anim Reprod Sci 2015. [PMID: 26205550 DOI: 10.1016/j.anireprosci.2015.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Placental 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2) inactivates glucocorticoids (GCs) to protect fetuses from over-exposure to maternal GCs, yet how maternal malnutrition affects placental 11β-HSD2 expression is unknown. In this study, Meishan sows were fed standard-protein (SP) or low-protein (LP, 50% of SP) diets and fetuses/newborn piglets were weighed and the corresponding placenta and umbilical cord blood were collected on gestational day 70 and the day of parturition. Significant growth retardation was observed in female, but not male, fetuses (P < 0.05) and the newborns (P < 0.01) of the LP group, which was accompanied by sexually dimorphic expression of 11β-HSD2 in placentas. Female fetuses in LP group showed significant decrease in placental 11β-HSD2 protein content (P < 0.05) and enzyme activity (P < 0.05), whereas male fetuses demonstrated significantly enhanced placental 11β-HSD2 activity (P < 0.05). Serum cortisol levels were significantly higher (P < 0.05) in male piglets compared to females, and the effects of maternal protein restriction on thyroid hormones (T3 and T4) in the umbilical cord blood were also sex dimorphic. Male piglets in LP group had significantly higher T3 (P < 0.01) and lower T4 (P < 0.01), whereas female piglets showed significantly lower T4 (P < 0.01) with no change in T3. As a result, male piglets in LP group exhibited significantly higher T3/T4 ratio compared to female counterparts. These results indicate that the effects of maternal protein restriction on placental 11β-HSD2 expression are gender-dependent in the pig, and thyroid hormones may be involved in such effects.
Collapse
|
34
|
Hoeijmakers L, Lucassen PJ, Korosi A. The interplay of early-life stress, nutrition, and immune activation programs adult hippocampal structure and function. Front Mol Neurosci 2015; 7:103. [PMID: 25620909 PMCID: PMC4288131 DOI: 10.3389/fnmol.2014.00103] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/16/2014] [Indexed: 01/08/2023] Open
Abstract
Early-life adversity increases the vulnerability to develop psychopathologies and cognitive decline later in life. This association is supported by clinical and preclinical studies. Remarkably, experiences of stress during this sensitive period, in the form of abuse or neglect but also early malnutrition or an early immune challenge elicit very similar long-term effects on brain structure and function. During early-life, both exogenous factors like nutrition and maternal care, as well as endogenous modulators, including stress hormones and mediator of immunological activity affect brain development. The interplay of these key elements and their underlying molecular mechanisms are not fully understood. We discuss here the hypothesis that exposure to early-life adversity (specifically stress, under/malnutrition and infection) leads to life-long alterations in hippocampal-related cognitive functions, at least partly via changes in hippocampal neurogenesis. We further discuss how these different key elements of the early-life environment interact and affect one another and suggest that it is a synergistic action of these elements that shapes cognition throughout life. Finally, we consider different intervention studies aiming to prevent these early-life adversity induced consequences. The emerging evidence for the intriguing interplay of stress, nutrition, and immune activity in the early-life programming calls for a more in depth understanding of the interaction of these elements and the underlying mechanisms. This knowledge will help to develop intervention strategies that will converge on a more complete set of changes induced by early-life adversity.
Collapse
Affiliation(s)
- Lianne Hoeijmakers
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| | - Paul J Lucassen
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
35
|
Schmidt U, Willmund GD, Holsboer F, Wotjak CT, Gallinat J, Kowalski JT, Zimmermann P. Searching for non-genetic molecular and imaging PTSD risk and resilience markers: Systematic review of literature and design of the German Armed Forces PTSD biomarker study. Psychoneuroendocrinology 2015; 51:444-58. [PMID: 25236294 DOI: 10.1016/j.psyneuen.2014.08.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 08/16/2014] [Accepted: 08/17/2014] [Indexed: 12/13/2022]
Abstract
Biomarkers allowing the identification of individuals with an above average vulnerability or resilience for posttraumatic stress disorder (PTSD) would especially serve populations at high risk for trauma exposure like firefighters, police officers and combat soldiers. Aiming to identify the most promising putative PTSD vulnerability markers, we conducted the first systematic review on potential imaging and non-genetic molecular markers for PTSD risk and resilience. Following the PRISMA guidelines, we systematically screened the PubMed database for prospective longitudinal clinical studies and twin studies reporting on pre-trauma and post-trauma PTSD risk and resilience biomarkers. Using 25 different combinations of search terms, we retrieved 8151 articles of which we finally included and evaluated 9 imaging and 27 molecular studies. In addition, we briefly illustrate the design of the ongoing prospective German Armed Forces (Bundeswehr) PTSD biomarker study (Bw-BioPTSD) which not only aims to validate these previous findings but also to identify novel and clinically applicable molecular, psychological and imaging risk, resilience and disease markers for deployment-related psychopathology in a cohort of German soldiers who served in Afghanistan.
Collapse
Affiliation(s)
- Ulrike Schmidt
- Max Planck Institute of Psychiatry, Kraepelinstrasse 10, 80804 München, Germany.
| | - Gerd-Dieter Willmund
- German Armed Forces Center of Military Mental Health, Scharnhorststrasse 13, 10115 Berlin, Germany
| | - Florian Holsboer
- Max Planck Institute of Psychiatry, Kraepelinstrasse 10, 80804 München, Germany
| | - Carsten T Wotjak
- Max Planck Institute of Psychiatry, Kraepelinstrasse 10, 80804 München, Germany
| | - Jürgen Gallinat
- Clinic for Psychiatry and Psychotherapy, University of Hamburg, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jens T Kowalski
- German Armed Forces Center of Military Mental Health, Scharnhorststrasse 13, 10115 Berlin, Germany
| | - Peter Zimmermann
- German Armed Forces Center of Military Mental Health, Scharnhorststrasse 13, 10115 Berlin, Germany
| |
Collapse
|
36
|
Analysis of extinction acquisition to attenuated tones in prenatally stressed and non-stressed offspring following auditory fear conditioning. Physiol Behav 2014; 139:157-66. [PMID: 25449394 DOI: 10.1016/j.physbeh.2014.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 07/04/2014] [Accepted: 11/07/2014] [Indexed: 11/24/2022]
Abstract
Stimulus generalization occurs when stimuli with characteristics similar to a previously conditioned stimulus (CS) become able to evoke a previously conditioned response. Experimental data (Lissek et al., 2005) indicate that patients with post-traumatic stress disorder (PTSD), more often show stimulus generalization following fear conditioning when tested under laboratory conditions. Factors surrounding this observation may contribute to two common features of PTSD: 1) hyper-responsiveness to sensory stimuli reminiscent of those associated with the original trauma, and 2) resistance of PTSD to extinction-based therapies. Adverse early experience is considered a risk factor for the later development of PTSD and in the present experiments we hypothesized that stimulus generalization would occur in an animal model of adverse early experience, the prenatally stressed (PS) rat. Adult PS and control (CON) rats underwent extensive pre-habituation to a conditioning chamber followed by conventional auditory fear conditioning. The next day both groups began an extinction regimen where a series of quieter (attenuated), CSs were administered prior to the full 75 dB training CS. When tested in this manner, PS rats froze at significantly lower tone amplitudes than did CON offspring on the first day of extinction training. This suggests that the PS rats had stimulus-generalized the CS to lower decibel tones. In addition to this finding, we also observed that PS rats froze more often and longer during three ensuing days of extinction training to attenuated tones. Group differences vanished when PS and CON rats were extinguished under conventional conditions. Thus, it appears that the two extinction regimens differed in their aversive cue saliency for the PS vs. CON rats. Follow-up prefrontal cortex transcriptome probing suggests that cholinergic and dopaminergic alterations may be involved.
Collapse
|
37
|
Jones NC, O'Brien TJ, Carmant L. Interaction between sex and early-life stress: influence on epileptogenesis and epilepsy comorbidities. Neurobiol Dis 2014; 72 Pt B:233-41. [PMID: 25266701 DOI: 10.1016/j.nbd.2014.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/08/2014] [Accepted: 09/12/2014] [Indexed: 12/22/2022] Open
Abstract
Epilepsy is a common brain disorder which is characterised by recurring seizures. In addition to suffering from the constant stress of living with this neurological condition, patients also frequently experience comorbid psychiatric and cognitive disorders which significantly impact their quality of life. There is growing appreciation that stress, in particular occurring in early life, can negatively impact brain development, creating an enduring vulnerability to develop epilepsy. This aligns with the solid connections between early life environments and the development of psychiatric conditions, promoting the possibility that adverse early life events could represent a common risk factor for the later development of both epilepsy and comorbid psychiatric disorders. The influence of sex has been little studied, but recent research points to potential important interactions, particularly with regard to effects mediated by HPA axis programming. Understanding these interactions, and the underlying molecular mechanisms, will provide important new insights into the causation of both epilepsy and of psychiatric disorders, and potentially open up novel avenues for treatment.
Collapse
Affiliation(s)
- Nigel C Jones
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.
| | - Terence J O'Brien
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Lionel Carmant
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
38
|
Genome wide association identifies common variants at the SERPINA6/SERPINA1 locus influencing plasma cortisol and corticosteroid binding globulin. PLoS Genet 2014; 10:e1004474. [PMID: 25010111 PMCID: PMC4091794 DOI: 10.1371/journal.pgen.1004474] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 05/15/2014] [Indexed: 11/30/2022] Open
Abstract
Variation in plasma levels of cortisol, an essential hormone in the stress response, is associated in population-based studies with cardio-metabolic, inflammatory and neuro-cognitive traits and diseases. Heritability of plasma cortisol is estimated at 30–60% but no common genetic contribution has been identified. The CORtisol NETwork (CORNET) consortium undertook genome wide association meta-analysis for plasma cortisol in 12,597 Caucasian participants, replicated in 2,795 participants. The results indicate that <1% of variance in plasma cortisol is accounted for by genetic variation in a single region of chromosome 14. This locus spans SERPINA6, encoding corticosteroid binding globulin (CBG, the major cortisol-binding protein in plasma), and SERPINA1, encoding α1-antitrypsin (which inhibits cleavage of the reactive centre loop that releases cortisol from CBG). Three partially independent signals were identified within the region, represented by common SNPs; detailed biochemical investigation in a nested sub-cohort showed all these SNPs were associated with variation in total cortisol binding activity in plasma, but some variants influenced total CBG concentrations while the top hit (rs12589136) influenced the immunoreactivity of the reactive centre loop of CBG. Exome chip and 1000 Genomes imputation analysis of this locus in the CROATIA-Korcula cohort identified missense mutations in SERPINA6 and SERPINA1 that did not account for the effects of common variants. These findings reveal a novel common genetic source of variation in binding of cortisol by CBG, and reinforce the key role of CBG in determining plasma cortisol levels. In turn this genetic variation may contribute to cortisol-associated degenerative diseases. Cortisol is a steroid hormone from the adrenal glands that is essential in the response to stress. Most cortisol in blood is bound to corticosteroid binding globulin (CBG). Diseases causing cortisol deficiency (Addison's disease) or excess (Cushing's syndrome) are life-threatening. Variations in plasma cortisol have been associated with cardiovascular and psychiatric diseases and their risk factors. To dissect the genetic contribution to variation in plasma cortisol, we formed the CORtisol NETwork (CORNET) consortium and recruited collaborators with suitable samples from more than 15,000 people. The results reveal that the major genetic influence on plasma cortisol is mediated by variations in the binding capacity of CBG. This is determined by differences in the circulating concentrations of CBG and also in the immunoreactivity of its ‘reactive centre loop’, potentially influencing not only binding affinity for cortisol but also the stability of CBG and hence the tissue delivery of cortisol. These findings provide the first evidence for a common genetic effect on levels of this clinically important hormone, suggest that differences in CBG between individuals are biologically important, and pave the way for further research to dissect causality in the associations of plasma cortisol with common diseases.
Collapse
|
39
|
Abstract
The primary adrenal cortical steroid hormones, aldosterone, and the glucocorticoids cortisol and corticosterone, act through the structurally similar mineralocorticoid (MR) and glucocorticoid receptors (GRs). Aldosterone is crucial for fluid, electrolyte, and hemodynamic homeostasis and tissue repair; the significantly more abundant glucocorticoids are indispensable for energy homeostasis, appropriate responses to stress, and limiting inflammation. Steroid receptors initiate gene transcription for proteins that effect their actions as well as rapid non-genomic effects through classical cell signaling pathways. GR and MR are expressed in many tissues types, often in the same cells, where they interact at molecular and functional levels, at times in synergy, others in opposition. Thus the appropriate balance of MR and GR activation is crucial for homeostasis. MR has the same binding affinity for aldosterone, cortisol, and corticosterone. Glucocorticoids activate MR in most tissues at basal levels and GR at stress levels. Inactivation of cortisol and corticosterone by 11β-HSD2 allows aldosterone to activate MR within aldosterone target cells and limits activation of the GR. Under most conditions, 11β-HSD1 acts as a reductase and activates cortisol/corticosterone, amplifying circulating levels. 11β-HSD1 and MR antagonists mitigate inappropriate activation of MR under conditions of oxidative stress that contributes to the pathophysiology of the cardiometabolic syndrome; however, MR antagonists decrease normal MR/GR functional interactions, a particular concern for neurons mediating cognition, memory, and affect.
Collapse
Affiliation(s)
- Elise Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Celso E. Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
40
|
|
41
|
Devakumar D, Birch M, Osrin D, Sondorp E, Wells JCK. The intergenerational effects of war on the health of children. BMC Med 2014; 12:57. [PMID: 24694212 PMCID: PMC3997818 DOI: 10.1186/1741-7015-12-57] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 02/26/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The short- and medium-term effects of conflict on population health are reasonably well documented. Less considered are its consequences across generations and potential harms to the health of children yet to be born. DISCUSSION Looking first at the nature and effects of exposures during conflict, and then at the potential routes through which harm may propagate within families, we consider the intergenerational effects of four features of conflict: violence, challenges to mental health, infection and malnutrition. Conflict-driven harms are transmitted through a complex permissive environment that includes biological, cultural and economic factors, and feedback loops between sources of harm and weaknesses in individual and societal resilience to them. We discuss the multiplicative effects of ongoing conflict when hostilities are prolonged. SUMMARY We summarize many instances in which the effects of war can propagate across generations. We hope that the evidence laid out in the article will stimulate research and--more importantly--contribute to the discussion of the costs of war; particularly in the longer-term in post-conflict situations in which interventions need to be sustained and adapted over many years.
Collapse
Affiliation(s)
- Delan Devakumar
- Institute for Global Health, University College London, London, UK
| | | | - David Osrin
- Institute for Global Health, University College London, London, UK
| | | | - Jonathan CK Wells
- Childhood Nutrition Research Centre, Institute of Child Health, University College London, London, UK
| |
Collapse
|
42
|
Zucchi FCR, Yao Y, Ilnytskyy Y, Robbins JC, Soltanpour N, Kovalchuk I, Kovalchuk O, Metz GAS. Lifetime stress cumulatively programs brain transcriptome and impedes stroke recovery: benefit of sensory stimulation. PLoS One 2014; 9:e92130. [PMID: 24651125 PMCID: PMC3961295 DOI: 10.1371/journal.pone.0092130] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/18/2014] [Indexed: 12/24/2022] Open
Abstract
Prenatal stress (PS) represents a critical variable affecting lifetime health trajectories, metabolic and vascular functions. Beneficial experiences may attenuate the effects of PS and its programming of health outcomes in later life. Here we investigated in a rat model (1) if PS modulates recovery following cortical ischemia in adulthood; (2) if a second hit by adult stress (AS) exaggerates stress responses and ischemic damage; and (3) if tactile stimulation (TS) attenuates the cumulative effects of PS and AS. Prenatally stressed and non-stressed adult male rats underwent focal ischemic motor cortex lesion and were tested in skilled reaching and skilled walking tasks. Two groups of rats experienced recurrent restraint stress in adulthood and one of these groups also underwent daily TS therapy. Animals that experienced both PS and AS displayed the most severe motor disabilities after lesion. By contrast, TS promoted recovery from ischemic lesion and reduced hypothalamic-pituitary-adrenal axis activity. The data also showed that cumulative effects of adverse and beneficial lifespan experiences interact with disease outcomes and brain plasticity through the modulation of gene expression. Microarray analysis of the lesion motor cortex revealed that cumulative PS and AS interact with genes related to growth factors and transcription factors, which were not affected by PS or lesion alone. TS in PS+AS animals reverted these changes, suggesting a critical role for these factors in activity-dependent motor cortical reorganization after ischemic lesion. These findings suggest that beneficial experience later in life can moderate adverse consequences of early programming to improve cerebrovascular health.
Collapse
Affiliation(s)
- Fabíola C. R. Zucchi
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
- Departments of Medicine and Biological Sciences, University of Mato Grosso State, Cáceres, MT, Brazil
| | - Youli Yao
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Jerrah C. Robbins
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Nasrin Soltanpour
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Gerlinde A. S. Metz
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
- * E-mail:
| |
Collapse
|
43
|
|
44
|
Abstract
INTRODUCTION The success of antidepressant research has long been challenged by a limited mechanistic understanding of depression pathogenesis and antidepressant treatment response. Progress in this field has thereby consistently been hindered by a lack of novel conceptual approaches and sophisticated experimental techniques to dissect the highly intricate neurobiology of depression. Using fresh approaches to investigate the cellular and molecular mechanisms underlying depression will thus be vital for discovery of novel antidepressant targets. AREAS COVERED This article provides an overview of some fundamental problems that depression research is currently facing and critically evaluates how these issues could be addressed by future research. It also discusses novel conceptual and technological advances in the field of neuroscience, particularly in regard to how they may help in providing unprecedented insight into the molecular mechanisms of depression pathogenesis. EXPERT OPINION Although progress in antidepressant drug discovery has been limited over recent years, modern innovations in neuroscience, molecular biology, genetics and bioinformatics are just beginning to revolutionize depression research and to reveal novel and promising treatment targets. Integrating findings from a range of relevant experimental models and using the most advanced technology will be vital for the future success of antidepressant drug discovery.
Collapse
Affiliation(s)
- Christoph Anacker
- McGill University, Douglas Mental Health University Institute , 6875 Boulevard La Salle, Montreal, Quebec, H4H 1R3 , Canada +1 514 761 6131 - 2503 ;
| |
Collapse
|
45
|
Yahyavi ST, Zarghami M, Marwah U. A review on the evidence of transgenerational transmission of posttraumatic stress disorder vulnerability. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2014; 36:89-94. [PMID: 24402183 DOI: 10.1590/1516-4446-2012-0995] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Accepted: 05/16/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To understand the risks of posttraumatic stress disorder (PTSD) development in the next generation of PTSD patients, we conducted a review on the biological, but not genetic, evidence of transgenerational transmission of PTSD vulnerability. METHODS Pertinent articles published from 1985 to September 2011 were searched using online academic search engines, including MEDLINE, EMBASE, ScienceDirect, OVID, PsycLIT, and SCOPUS, and a non-systematic review was conducted. RESULTS There is paradoxical evidence that hypothalamic-pituitary-adrenal axis changes in PTSD patients may also be evident in their offspring. This effect and biological vulnerability to PTSD may be transmitted across generations through maternal epigenetic programming during pregnancy. The samples of most studies, which were not large enough and represented the outcome of few research groups, consisted of a specific type of patients with a particular trauma. CONCLUSIONS There is still a need to conduct studies in other geographical areas with different genetic background and larger samples considering different types of trauma other than those specified in the current literature, so as to strengthen the evidence of transgenerational transmission of PTSD vulnerability.
Collapse
|
46
|
Braun T, Challis JR, Newnham JP, Sloboda DM. Early-life glucocorticoid exposure: the hypothalamic-pituitary-adrenal axis, placental function, and long-term disease risk. Endocr Rev 2013; 34:885-916. [PMID: 23970762 DOI: 10.1210/er.2013-1012] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
An adverse early-life environment is associated with long-term disease consequences. Adversity early in life is hypothesized to elicit developmental adaptations that serve to improve fetal and postnatal survival and prepare the organism for a particular range of postnatal environments. These processes, although adaptive in their nature, may later prove to be maladaptive or disadvantageous if the prenatal and postnatal environments are widely discrepant. The exposure of the fetus to elevated levels of either endogenous or synthetic glucocorticoids is one model of early-life adversity that contributes substantially to the propensity of developing disease. Moreover, early-life glucocorticoid exposure has direct clinical relevance because synthetic glucocorticoids are routinely used in the management of women at risk of early preterm birth. In this regard, reports of adverse events in human newborns have raised concerns about the safety of glucocorticoid treatment; synthetic glucocorticoids have detrimental effects on fetal growth and development, childhood cognition, and long-term behavioral outcomes. Experimental evidence supports a link between prenatal exposure to synthetic glucocorticoids and alterations in fetal development and changes in placental function, and many of these alterations appear to be permanent. Because the placenta is the conduit between the maternal and fetal environments, it is likely that placental function plays a key role in mediating effects of fetal glucocorticoid exposure on hypothalamic-pituitary-adrenal axis development and long-term disease risk. Here we review recent insights into how the placenta responds to changes in the intrauterine glucocorticoid environment and discuss possible mechanisms by which the placenta mediates fetal hypothalamic-pituitary-adrenal development, metabolism, cardiovascular function, and reproduction.
Collapse
Affiliation(s)
- Thorsten Braun
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, 1280 Main Street West, HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1.
| | | | | | | |
Collapse
|
47
|
Kalinina TS, Dygalo NN. Development of the noradrenergic system of the rat brain after prenatal exposure to corticosterone. BIOL BULL+ 2013. [DOI: 10.1134/s1062359013040043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
48
|
Bromer C, Marsit CJ, Armstrong DA, Padbury JF, Lester B. Genetic and epigenetic variation of the glucocorticoid receptor (NR3C1) in placenta and infant neurobehavior. Dev Psychobiol 2013; 55:673-83. [PMID: 22714792 PMCID: PMC3458180 DOI: 10.1002/dev.21061] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 05/26/2012] [Indexed: 01/21/2023]
Abstract
The intrauterine environment can impact the developing infant by altering the function of the placenta through changes to the epigenetic regulatory features of this tissue. Genetic variation, too, may impact infant development or may modify the relationship between epigenetic alterations and infant outcomes. To examine the associations of these variations with early life infant neurodevelopment, we examined the extent of DNA methylation of the glucocorticoid receptor gene (NR3C1) promoter and a common single nucleotide polymorphism in the promoter region in a series of 186 placentas from healthy newborn infants. We associated these molecular features with specific summary measures from the NICU Network Neurobehavioral Scales. After controlling for genotype and confounders, we identified significant associations of NR3C1 methylation with infant quality of movement (p = .05) and with infant attention (p = .05), and a potential interaction between methylation and genotype on infant attention score. These results suggest that epigenetic alteration of the NR3C1 gene in the placentas of genetically susceptible infants can have impacts on neurodevelopment which may have lifelong impact on neurobehavioral and mental health outcomes. Further research is needed to more precisely define these relationships and the interaction between epigenetic alterations and genetic variations on infant health.
Collapse
Affiliation(s)
- Cailey Bromer
- The Brown Center for the Study of Children at Risk, Providence, RI 02903 USA
| | - Carmen J. Marsit
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI 02903 USA
- Departments of Pharmacology and Toxicology and Community and Family Medicine Section Epidemiology and Biostatistics, Dartmouth Medical School, Hanover, NH 03755
| | - David A. Armstrong
- Departments of Pharmacology and Toxicology and Community and Family Medicine Section Epidemiology and Biostatistics, Dartmouth Medical School, Hanover, NH 03755
| | - James F. Padbury
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI 02903 USA
- Brown Alpert Medical School, Providence, RI 02903 USA
| | - Barry Lester
- The Brown Center for the Study of Children at Risk, Providence, RI 02903 USA
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI 02903 USA
- Brown Alpert Medical School, Providence, RI 02903 USA
| |
Collapse
|
49
|
McGowan PO. Epigenomic Mechanisms of Early Adversity and HPA Dysfunction: Considerations for PTSD Research. Front Psychiatry 2013; 4:110. [PMID: 24133457 PMCID: PMC3783846 DOI: 10.3389/fpsyt.2013.00110] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/03/2013] [Indexed: 01/22/2023] Open
Abstract
Childhood adversity can have life-long consequences for the response to stressful events later in life. Abuse or severe neglect are well-known risk factors for post-traumatic stress disorder (PTSD), at least in part via changes in neural systems mediating the endocrine response to stress. Determining the biological signatures of risk for stress-related mental disorders such as PTSD is important for identifying homogenous subgroups and improving treatment options. This review will focus on epigenetic regulation in early life by adversity and parental care - prime mediators of offspring neurodevelopment - in order to address several questions: (1) what have studies of humans and analogous animal models taught us about molecular mechanisms underlying changes in stress-sensitive physiological systems in response to early life trauma? (2) What are the considerations for studies relating early adversity and PTSD risk, going forward? I will summarize studies in animals and humans that address the epigenetic response to early adversity in the brain and in peripheral tissues. In so doing, I will describe work on the glucocorticoid receptor and other well-characterized genes within the stress response pathway and then turn to genomic studies to illustrate the use of increasingly powerful high-throughput approaches to the study of epigenomic mechanisms.
Collapse
Affiliation(s)
- Patrick O McGowan
- Centre for Environmental Epigenetics and Development , Toronto, ON , Canada ; Department of Biological Sciences, University of Toronto , Scarborough, ON , Canada ; Cell and Systems Biology, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
50
|
Grigoryan G, Segal M. Prenatal stress alters noradrenergic modulation of LTP in hippocampal slices. J Neurophysiol 2013; 110:279-85. [DOI: 10.1152/jn.00834.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Long-term effects of stress during pregnancy on brain and behavior have been analyzed extensively in recent years. These effects include changes in emotional behavior, a reduction in learning capacity, and ability to generate long-term potentiation (LTP) in the offspring. In earlier studies, we and others have described a difference in ability to express LTP in dorsal and ventral sectors of the hippocampus (DH and VH, respectively) and its modification by prior stress. We now found that norepinephrine (NE) facilitated conversion of short-term potentiation to LTP in the normal DH but not in VH. Prenatal stress (PS) switched the locus of the facilitating action of NE from the DH to the VH. The effects of NE are likely to be mediated by activation of calcium stores. PS also facilitated ( S)-3,5-dihydroxyphenylglycine hydrate (DHPG)-induced LTD in the VH, assumed to be mediated by release of calcium from stores. These observations have important implications for the role of the hippocampus in cognitive and emotional memories.
Collapse
Affiliation(s)
- Gayane Grigoryan
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
| | - Menahem Segal
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|