1
|
Bi H, Jin J, Sun M, Chen M, Li X, Wang Y. Epigenetic changes caused by early life stress in the pathogenesis of depression. Eur J Pharmacol 2025; 999:177671. [PMID: 40288560 DOI: 10.1016/j.ejphar.2025.177671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Major depressive disorder (MDD) is a severe psychiatric disorder with a complex and poorly understood pathogenesis. Epigenetics, a rapidly advancing field of biology, has been implicated in various psychiatric conditions, including schizophrenia, anxiety, substance addiction, and autism. Furthermore, substantial research indicates that epigenetic modifications play a crucial role in the etiology of depression. Early life stress (ELS) refers to adverse experiences occurring during prenatal development (e.g., maternal physical and mental health complications during pregnancy) and/or postnatal life (e.g., abuse, neglect, poverty, parental loss, family conflict, violence, and malnutrition). These early-life adversities can lead to epigenetic modifications, which, in turn, influence key biological processes and contribute to the pathogenesis of MDD. This review provides an overview of the regulatory mechanisms and functions of various epigenetic modifications, including non-coding RNAs, DNA methylation, and histone modifications. We then examine ELS-induced epigenetic alterations and their biological consequences, such as dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, neurogenesis, and neuroplasticity. Finally, we explore their potential implications for both the pathogenesis and treatment of MDD. We hypothesize that ELS-induced epigenetic changes may serve as biomarkers for MDD diagnosis and offer novel therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Hongsheng Bi
- Department of Psychiatry, The First Hospital of China Medical University, China; The Third Hospital of Daqing, Psychiatric Ward No. 2, China
| | - Jingyan Jin
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Mingyuan Sun
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Mingrui Chen
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Xiaobai Li
- Department of Psychiatry, The First Hospital of China Medical University, China.
| | - Yan Wang
- Center for Psychological Development, China Medical University, China.
| |
Collapse
|
2
|
Paes T, Hofland LJ, Iyer AM, Feelders RA. Epigenetic implications in the pathogenesis of corticotroph tumors. Pituitary 2025; 28:51. [PMID: 40257628 PMCID: PMC12011945 DOI: 10.1007/s11102-025-01522-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/02/2025] [Indexed: 04/22/2025]
Abstract
Non-mutational epigenetic reprogramming is considered an important enabling characteristic of neoplasia. Corticotroph tumors and other subtypes of pituitary tumors are characterized by distinct epigenetic profiles. The DNA methylation profile is consistent with disease-specific gene expression, which highlights the importance of epigenetic changes in tumor formation and progression. Elucidating the epigenetic changes underlying tumorigenesis plays an important role in understanding the molecular pathogenesis of corticotroph tumors and may ultimately contribute to improving tumor-specific treatment. Here, we provide an overview of the epigenetic landscape of corticotroph tumors. We also review the role of epigenetics in silencing the expression of tumor suppressor genes and promoting oncogenes expression, which could potentially be involved in the pathogenesis of corticotroph tumors. We briefly discuss microRNAs and epigenetic aspects of POMC regulation. Lastly, since the epigenetic changes are reversible, we discuss drugs that target epigenetic modifiers that could potentially be used in the arsenal of Cushing's disease treatment modalities.
Collapse
Affiliation(s)
- Ticiana Paes
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, Rotterdam, GD 3015, The Netherlands
- Department of Internal Medicine, Roger Williams Medical Center, Providence, RI, USA
| | - Leo J Hofland
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, Rotterdam, GD 3015, The Netherlands
| | - Anand M Iyer
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, Rotterdam, GD 3015, The Netherlands
| | - Richard A Feelders
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, Rotterdam, GD 3015, The Netherlands.
- Division of Endocrinology, Diabetes and Metabolism, New York University Langone Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
3
|
Dixon R, Malave L, Thompson R, Wu S, Li Y, Sadik N, Anacker C. Sex-specific and developmental effects of early life adversity on stress reactivity are rescued by postnatal knockdown of 5-HT 1A autoreceptors. Neuropsychopharmacology 2025; 50:507-518. [PMID: 39396089 PMCID: PMC11736140 DOI: 10.1038/s41386-024-01999-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 10/14/2024]
Abstract
Early Life Adversity (ELA) predisposes to stress hypersensitivity in adulthood, but neurobiological mechanisms that protect from the enduring effects of ELA are poorly understood. Serotonin 1A (5HT1A) autoreceptors in the raphé nuclei regulate adult stress vulnerability, but whether 5HT1A could be targeted to prevent ELA effects on susceptibility to future stressors is unknown. Here, we exposed mice with postnatal knockdown of 5HT1A autoreceptors to the limited bedding and nesting model of ELA from postnatal day (P)3-10 and tested behavioral, neuroendocrine, neurogenic, and neuroinflammatory responses to an acute swim stress in male and female mice in adolescence (P35) and in adulthood (P56). In females, ELA decreased raphé 5HT neuron activity in adulthood and increased passive coping with the acute swim stress, corticosterone levels, neuronal activity, and corticotropin-releasing factor (CRF) levels in the paraventricular nucleus (PVN) of the hypothalamus. ELA also reduced neurogenesis in the ventral dentate gyrus (vDG) of the hippocampus, an important mediator of individual differences in stress susceptibility, and increased microglia activation in the PVN and vDG. These effects of ELA were specific to females and manifested predominantly in adulthood, but not earlier on in adolescence. Postnatal knockdown of 5HT1A autoreceptors prevented these effects of ELA on 5HT neuron activity, stress reactivity, neurogenesis, and neuroinflammation in adult female mice. Our findings demonstrate that ELA induces long-lasting and sex-specific impairments in the serotonin system, stress reactivity, and vDG function, and identify 5HT1A autoreceptors as potential targets to prevent these enduring effects of ELA.
Collapse
Affiliation(s)
- Rushell Dixon
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Lauren Malave
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Rory Thompson
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Serena Wu
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Yifei Li
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Noah Sadik
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Christoph Anacker
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA.
- Columbia University Institute for Developmental Sciences, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA.
- Columbia University Stem Cell Initiative (CSCI), Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA.
| |
Collapse
|
4
|
Mormede E, Mormede P. Genetic Variation of Hypothalamic-Pituitary-Adrenal Axis Activity in Farm Animals and Beyond. Neuroendocrinology 2024; 115:128-137. [PMID: 39626641 DOI: 10.1159/000542831] [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: 04/11/2024] [Accepted: 11/14/2024] [Indexed: 01/21/2025]
Abstract
BACKGROUND Many experimental data in several species clearly demonstrate the important genetic contribution to variations in HPA axis activity. The influence of corticosteroid hormones on adaptive processes and on production traits such as growth rate, feed efficiency, carcass composition, and meat quality is a strong impetus to the search for the molecular bases of these differences for efficient genetic selection. SUMMARY Three main sources of genetic variability have been documented so far in farm animal species, the adrenal cortex sensitivity to ACTH-regulating corticosteroid hormone production, the bioavailability of corticosteroid hormones and especially corticosteroid-binding globulin capacity, and glucocorticoid receptor function. The effect of single mutations may be dependent on the genetic background, and genetic variation of cortisol levels may have different functional consequences depending on the molecular mechanisms responsible for this change. KEY MESSAGES Understanding the genetic basis of HPA axis activity allows the development of genomic tools and breeding technologies aimed at improving adaptive capacity and stress tolerance in farm animals and their use as valuable models for the genetic study of the HPA axis and the correlation with adaptation, metabolism, and other functions regulated by adrenal hormones, and associated pathologies (obesity, cardiovascular, etc.). The next step will be to explore HPA axis variability from a system genetics perspective including the multiple sources of variation and their interactions. This multifactorial approach is a prerequisite to the use of the HPA axis phenotypes in the genetic selection for more productive and robust animals, with a high level of production of quality products.
Collapse
Affiliation(s)
- Elena Mormede
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, Toulouse, France
| | - Pierre Mormede
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, Toulouse, France
| |
Collapse
|
5
|
Dixon R, Malave L, Thompson R, Wu S, Li Y, Sadik N, Anacker C. Sex-specific and Developmental Effects of Early Life Adversity on Stress Reactivity are Rescued by Postnatal Knockdown of 5-HT 1A Autoreceptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576344. [PMID: 38328253 PMCID: PMC10849559 DOI: 10.1101/2024.01.22.576344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Early Life Adversity (ELA) predisposes to stress hypersensitivity in adulthood, but neurobiological mechanisms that protect from the enduring effects of ELA are poorly understood. Serotonin 1A (5HT 1A ) autoreceptors in the raphé nuclei regulate adult stress vulnerability, but whether 5HT 1A could be targeted to prevent ELA effects on susceptibility to future stressors is unknown. Here, we exposed mice with postnatal knockdown of 5HT 1A autoreceptors to the limited bedding and nesting model of ELA from postnatal day (P)3-10 and tested behavioral, neuroendocrine, neurogenic, and neuroinflammatory responses to an acute swim stress in male and female mice in adolescence (P35) and in adulthood (P56). In females, ELA decreased raphé 5HT neuron activity in adulthood and increased passive coping with the acute swim stress, corticosterone levels, neuronal activity, and corticotropin-releasing factor (CRF) levels in the paraventricular nucleus (PVN) of the hypothalamus. ELA also reduced neurogenesis in the ventral dentate gyrus (vDG) of the hippocampus, an important mediator of individual differences in stress susceptibility, and increased microglia activation in the PVN and vDG. These effects of ELA were specific to females and manifested predominantly in adulthood, but not earlier on in adolescence. Postnatal knockdown of 5HT 1A autoreceptors prevented these effects of ELA on 5HT neuron activity, stress reactivity, neurogenesis, and neuroinflammation in adult female mice. Our findings demonstrate that ELA induces long-lasting and sex-specific impairments in the serotonin system, stress reactivity, and vDG function, and identify 5HT 1A autoreceptors as potential targets to prevent these enduring effects of ELA.
Collapse
|
6
|
Coppens G, Vanhorebeek I, Güiza F, Derese I, Wouters PJ, Téblick A, Dulfer K, Joosten KF, Verbruggen SC, Van den Berghe G. Abnormal DNA methylation within HPA-axis genes years after paediatric critical illness. Clin Epigenetics 2024; 16:31. [PMID: 38395991 PMCID: PMC10893716 DOI: 10.1186/s13148-024-01640-y] [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: 11/02/2023] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Critically ill children suffer from impaired physical/neurocognitive development 2 years later. Glucocorticoid treatment alters DNA methylation within the hypothalamus-pituitary-adrenal (HPA) axis which may impair normal brain development, cognition and behaviour. We tested the hypothesis that paediatric-intensive-care-unit (PICU) patients, sex- and age-dependently, show long-term abnormal DNA methylation within the HPA-axis layers, possibly aggravated by glucocorticoid treatment in the PICU, which may contribute to the long-term developmental impairments. RESULTS In a pre-planned secondary analysis of the multicentre PEPaNIC-RCT and its 2-year follow-up, we identified differentially methylated positions and differentially methylated regions within HPA-axis genes in buccal mucosa DNA from 818 former PICU patients 2 years after PICU admission (n = 608 no glucocorticoid treatment; n = 210 glucocorticoid treatment) versus 392 healthy children and assessed interaction with sex and age, role of glucocorticoid treatment in the PICU and associations with long-term developmental impairments. Adjusting for technical variation and baseline risk factors and correcting for multiple testing (false discovery rate < 0.05), former PICU patients showed abnormal DNA methylation of 26 CpG sites (within CRHR1, POMC, MC2R, NR3C1, FKBP5, HSD11B1, SRD5A1, AKR1D1, DUSP1, TSC22D3 and TNF) and three DNA regions (within AVP, TSC22D3 and TNF) that were mostly hypomethylated. These abnormalities were sex-independent and only partially age-dependent. Abnormal methylation of three CpG sites within FKBP5 and one CpG site within SRD5A1 and AKR1D1 was partly attributable to glucocorticoid treatment during PICU stay. Finally, abnormal methylation within FKBP5 and AKR1D1 was most robustly associated with long-term impaired development. CONCLUSIONS Two years after critical illness in children, abnormal methylation within HPA-axis genes was present, predominantly within FKBP5 and AKR1D1, partly attributable to glucocorticoid treatment in the PICU, and explaining part of the long-term developmental impairments. These data call for caution regarding liberal glucocorticoid use in the PICU.
Collapse
Affiliation(s)
- Grégoire Coppens
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Ilse Vanhorebeek
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Fabian Güiza
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Inge Derese
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Pieter J Wouters
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Arno Téblick
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Karolijn Dulfer
- Division of Paediatric Intensive Care Unit, Department of Neonatal and Paediatric ICU, Erasmus Medical Centre, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Koen F Joosten
- Division of Paediatric Intensive Care Unit, Department of Neonatal and Paediatric ICU, Erasmus Medical Centre, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Sascha C Verbruggen
- Division of Paediatric Intensive Care Unit, Department of Neonatal and Paediatric ICU, Erasmus Medical Centre, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
7
|
Park S, Oh S, Kim N, Kim EK. HMBA ameliorates obesity by MYH9- and ACTG1-dependent regulation of hypothalamic neuropeptides. EMBO Mol Med 2023; 15:e18024. [PMID: 37984341 DOI: 10.15252/emmm.202318024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023] Open
Abstract
The global epidemic of obesity remains a daunting problem. Here, we report hexamethylene bisacetamide (HMBA) as a potent anti-obesity compound. Peripheral and central administration of HMBA to diet-induced obese mice regulated the expression of hypothalamic neuropeptides critical for energy balance, leading to beneficial metabolic effects such as anorexia and weight loss. We found that HMBA bound to MYH9 and ACTG1, which were required for the anti-obesity effects of HMBA in both NPY-expressing and POMC-expressing neurons. The binding of HMBA to MYH9 and ACTG1 elevated the expression of HEXIM1 and enhanced its interaction with MDM2, resulting in the dissociation of the HEXIM1-p53 complex in hypothalamic cells. Subsequently, the free HEXIM1 and p53 translocated to the nucleus, where they downregulated the transcription of orexigenic NPY, but p53 and acetylated histone 3 upregulated that of anorexigenic POMC. Our study points to a previously unappreciated efficacy of HMBA and reveals its mechanism of action in metabolic regulation, which may propose HMBA as a potential therapeutic strategy for obesity.
Collapse
Affiliation(s)
- Seokjae Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Sungjoon Oh
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Nayoun Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Eun-Kyoung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| |
Collapse
|
8
|
ElGendy K, Malcomson FC, Afshar S, Bradburn MD, Mathers JC. Effects of obesity, and of weight loss following bariatric surgery, on methylation of DNA from the rectal mucosa and in cell-free DNA from blood. Int J Obes (Lond) 2023; 47:1278-1285. [PMID: 37714902 DOI: 10.1038/s41366-023-01384-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/21/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND DNA methylation is an epigenetic mechanism through which environmental factors including nutrition and inflammation influence health. Obesity is a major modifiable risk factor for many common diseases including cardiovascular diseases and cancer. In particular, obesity-induced inflammation resulting from aberrantly-methylated inflammatory genes may drive risk of several non-communicable diseases including colorectal cancer (CRC). This study is the first to investigate the effects of weight loss induced by bariatric surgery (BS) on DNA methylation in the rectum and in cell-free DNA (cfDNA) from blood. SUBJECTS AND METHODS DNA methylation was quantified in rectal mucosal biopsies and cfDNA from serum of 28 participants with obesity before and 6 months after BS, as well as in 12 participants without obesity (control group) matched for age and sex from the Biomarkers Of Colorectal cancer After Bariatric Surgery (BOCABS) Study. DNA methylation of LEP, IL6, POMC, LINE1, MAPK7 and COX2 was quantified by pyrosequencing. RESULTS BMI decreased significantly from 41.8 kg/m2 pre-surgery to 32.3 kg/m2 at 6 months after BS. Compared with the control group, obesity was associated with lower LEP methylation in both the rectal mucosa and in cfDNA from serum. BS normalised LEP methylation in DNA from the rectal mucosa but not in cfDNA. BS decreased methylation of some CpG sites of LINE1 in the rectal mucosal DNA and in cfDNA to levels comparable with those in participants without obesity. Methylation of POMC in rectal mucosal DNA was normalised at 6 months after BS. CONCLUSION BS reversed LINE1, POMC and LEP methylation in the rectal mucosa of patients with obesity to levels similar to those in individuals without obesity. These findings support current evidence of effects of BS-induced weight loss on reversibility of DNA methylation in other tissues. The DNA methylation changes in the rectal mucosa shows promise as a biomarker for objective assessment of effects of weight loss interventions on risk of cancer and other diseases.
Collapse
Affiliation(s)
- Khalil ElGendy
- Human Nutrition Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, England.
- Surgery Department, Northumbria NHS Foundation Trust, Newcastle upon Tyne, England.
| | - Fiona C Malcomson
- Human Nutrition Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, England
| | - Sorena Afshar
- Human Nutrition Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, England
- Surgery Department, Northumbria NHS Foundation Trust, Newcastle upon Tyne, England
| | - Michael D Bradburn
- Surgery Department, Northumbria NHS Foundation Trust, Newcastle upon Tyne, England
| | - John C Mathers
- Human Nutrition Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, England
| |
Collapse
|
9
|
Meneses-San Juan D, Lamas M, Ramírez-Rodríguez GB. Repetitive Transcranial Magnetic Stimulation Reduces Depressive-like Behaviors, Modifies Dendritic Plasticity, and Generates Global Epigenetic Changes in the Frontal Cortex and Hippocampus in a Rodent Model of Chronic Stress. Cells 2023; 12:2062. [PMID: 37626872 PMCID: PMC10453847 DOI: 10.3390/cells12162062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Depression is the most common affective disorder worldwide, accounting for 4.4% of the global population, a figure that could increase in the coming decades. In depression, there exists a reduction in the availability of dendritic spines in the frontal cortex (FC) and hippocampus (Hp). In addition, histone modification and DNA methylation are also dysregulated epigenetic mechanisms in depression. Repetitive transcranial magnetic stimulation (rTMS) is a technique that is used to treat depression. However, the epigenetic mechanisms of its therapeutic effect are still not known. Therefore, in this study, we evaluated the antidepressant effect of 5 Hz rTMS and examined its effect on dendritic remodeling, immunoreactivity of synapse proteins, histone modification, and DNA methylation in the FC and Hp in a model of chronic mild stress. Our data indicated that stress generated depressive-like behaviors and that rTMS reverses this effect, romotes the formation of dendritic spines, and favors the presynaptic connection in the FC and DG (dentate gyrus), in addition to increasing histone H3 trimethylation and DNA methylation. These results suggest that the antidepressant effect of rTMS is associated with dendritic remodeling, which is probably regulated by epigenetic mechanisms. These data are a first approximation of the impact of rTMS at the epigenetic level in the context of depression. Therefore, it is necessary to analyze in future studies as to which genes are regulated by these mechanisms, and how they are associated with the neuroplastic modifications promoted by rTMS.
Collapse
Affiliation(s)
- David Meneses-San Juan
- National Institute of Psychiatry “Ramón de la Fuente Muñiz”, Mexico City 14370, Mexico;
- Center of Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico;
| | - Mónica Lamas
- Center of Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico;
| | | |
Collapse
|
10
|
Branham EM, McLean SA, Deliwala I, Mauck MC, Zhao Y, McKibben LA, Lee A, Spencer AB, Zannas AS, Lechner M, Danza T, Velilla MA, Hendry PL, Pearson C, Peak DA, Jones J, Rathlev NK, Linnstaedt SD. CpG Methylation Levels in HPA Axis Genes Predict Chronic Pain Outcomes Following Trauma Exposure. THE JOURNAL OF PAIN 2023; 24:1127-1141. [PMID: 36906051 PMCID: PMC10330094 DOI: 10.1016/j.jpain.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/21/2023] [Accepted: 03/01/2023] [Indexed: 03/12/2023]
Abstract
Chronic post-traumatic musculoskeletal pain (CPTP) is a common outcome of traumatic stress exposure. Biological factors that influence the development of CPTP are poorly understood, though current evidence indicates that the hypothalamic-pituitary-adrenal (HPA) axis plays a critical role in its development. Little is known about molecular mechanisms underlying this association, including epigenetic mechanisms. Here, we assessed whether peritraumatic DNA methylation levels at 248 5'-C-phosphate-G-3' (CpG) sites in HPA axis genes (FKBP5, NR3C1, CRH, CRHR1, CRHR2, CRHBP, POMC) predict CPTP and whether identified CPTP-associated methylation levels influence expression of those genes. Using participant samples and data collected from trauma survivors enrolled into longitudinal cohort studies (n = 290), we used linear mixed modeling to assess the relationship between peritraumatic blood-based CpG methylation levels and CPTP. A total of 66 (27%) of the 248 CpG sites assessed in these models statistically significantly predicted CPTP, with the three most significantly associated CpG sites originating from the POMC gene region (ie, cg22900229 [β = .124, P < .001], cg16302441 [β = .443, P < .001], cg01926269 [β = .130, P < .001]). Among the genes analyzed, both POMC (z = 2.36, P = .018) and CRHBP (z = 4.89, P < .001) were enriched in CpG sites significantly associated with CPTP. Further, POMC expression was inversely correlated with methylation levels in a CPTP-dependent manner (6-months NRS<4: r = -.59, P < .001; 6-months NRS ≥ 4: r = -.18, P = .2312). Our results suggest that methylation of HPA axis genes including POMC and CRHBP predict risk for and may contribute to vulnerability to CPTP. PERSPECTIVE: Peritraumatic blood levels of CpG methylation sites in HPA axis genes, particularly CpG sites in the POMC gene, predict CPTP development. This data substantially advances our understanding of epigenetic predictors and potential mediators of CPTP, a highly common, morbid, and hard-to-treat form of chronic pain.
Collapse
Affiliation(s)
- Erica M Branham
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, North Carolina
| | - Samuel A McLean
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina; Department of Emergency Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Ishani Deliwala
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina
| | - Matthew C Mauck
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina
| | - Ying Zhao
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina
| | - Lauren A McKibben
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina
| | - Aaron Lee
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina
| | - Alex B Spencer
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina
| | - Anthony S Zannas
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina; Department of Genetics, University of North Carolina, Chapel Hill, North Carolina; Carolina Stress Initiative, University of North Carolina, Chapel Hill, North Carolina
| | - Megan Lechner
- Forensic Nursing Program, Memorial Health System, Colorado Springs, Colorado
| | - Teresa Danza
- Forensic Nursing Program, Albuquerque SANE Collaborative, Albuquerque, New Mexico
| | | | - Phyllis L Hendry
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville, Florida
| | - Claire Pearson
- Department of Emergency Medicine, Detroit Receiving, Detroit, Michigan
| | - David A Peak
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jeffrey Jones
- Department of Emergency Medicine, Spectrum Health Butterworth Campus, Grand Rapids, Michigan
| | - Niels K Rathlev
- Department of Emergency Medicine, University of Massachusetts Chan Medical School Baystate, Springfield, Massachusetts
| | - Sarah D Linnstaedt
- Institute for Trauma Recovery, University of North Carolina, Chapel Hill, North Carolina; Department of Anesthesiology, University of North Carolina, Chapel Hill, North Carolina; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, North Carolina.
| |
Collapse
|
11
|
Muir RQ, Klocke BJ, Jennings MS, Molina PA, Hsu JS, Kellum CE, Alexander KL, Lee G, Foote JB, Lorenz RG, Pollock JS, Maynard CL. Early Life Stress in Mice Leads to Impaired Colonic Corticosterone Production and Prolonged Inflammation Following Induction of Colitis. Inflamm Bowel Dis 2023; 29:960-972. [PMID: 36661889 PMCID: PMC10233396 DOI: 10.1093/ibd/izac280] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Early life stress (ELS) is an environmental trigger believed to promote increased risk of IBD. Our goal was to identify mechanisms whereby ELS in mice affects susceptibility to and/or severity of gut inflammation. METHODS We utilized 2 published animal models of ELS. In the first model, newborn mice were separated from the dam daily for 4 to 8 hours starting on postnatal day 2 and then weaned early on postnatal day 17. Control mice were left undisturbed with the dams until weaning on postnatal day 21. In the second model, dams were fed dexamethasone or vehicle ad libitum in drinking water on postpartum days 1 to 14. Plasma and colonic corticosterone were measured in juvenile and adult mice. Colitis was induced in 4-week-old mice via intraperitoneal injection of interleukin (IL)-10 receptor blocking antibody every 5 days for 15 days. Five or 15 days later, colitis scores and transcripts for Tnf, glucocorticoid receptors, and steroidogenic enzymes were measured. RESULTS Mice exposed to ELS displayed reduced plasma and colonic corticosterone. Control animals showed improvements in indices of inflammation following cessation of interleukin-10 receptor blockade, whereas ELS-exposed animals maintained high levels of Tnf and histological signs of colitis. In colitic animals, prior exposure to ELS was associated with significantly lower expression of genes associated with corticosterone synthesis and responsiveness. Finally, TNF stimulation of colonic crypt cells from ELS mice led to increased inhibition of corticosterone synthesis. CONCLUSIONS Our study identifies impaired local glucocorticoid production and responsiveness as a potential mechanism whereby ELS predisposes to chronic colitis in susceptible hosts.
Collapse
Affiliation(s)
- Rachel Q Muir
- Department of Pathology, University of Alabama at Birmingham, Birmingham, ALUSA
| | - Barbara J Klocke
- Department of Pathology, University of Alabama at Birmingham, Birmingham, ALUSA
| | - Melissa S Jennings
- Department of Pathology, University of Alabama at Birmingham, Birmingham, ALUSA
| | - Patrick A Molina
- Department of Medicine, University of Alabama at Birmingham, Birmingham, ALUSA
| | - Jung-Shan Hsu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, ALUSA
| | - Cailin E Kellum
- Department of Medicine, University of Alabama at Birmingham, Birmingham, ALUSA
| | - Katie L Alexander
- Department of Medicine, University of Alabama at Birmingham, Birmingham, ALUSA
| | - Goo Lee
- Department of Pathology, University of Alabama at Birmingham, Birmingham, ALUSA
| | - Jeremy B Foote
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robin G Lorenz
- Department of Research Pathology, Genentech, San Francisco, CAUSA
| | - Jennifer S Pollock
- Department of Medicine, University of Alabama at Birmingham, Birmingham, ALUSA
| | - Craig L Maynard
- Department of Pathology, University of Alabama at Birmingham, Birmingham, ALUSA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, ALUSA
| |
Collapse
|
12
|
Malekpour M, Jafari A, Kashkooli M, Salarikia SR, Negahdaripour M. A systems biology approach for discovering the cellular and molecular aspects of psychogenic non-epileptic seizure. Front Psychiatry 2023; 14:1116892. [PMID: 37252132 PMCID: PMC10213457 DOI: 10.3389/fpsyt.2023.1116892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/26/2023] [Indexed: 05/31/2023] Open
Abstract
Objectives Psychogenic non-epileptic seizure (PNES) is the most common non-epileptic disorder in patients referring to epilepsy centers. Contrary to common beliefs about the disease's harmlessness, the death rate of PNES patients is similar to drug-resistant epilepsy. Meanwhile, the molecular pathomechanism of PNES is unknown with very limited related research. Thus, the aim of this in silico study was to find different proteins and hormones associated with PNES via a systems biology approach. Methods Different bioinformatics databases and literature review were used to find proteins associated with PNES. The protein-hormone interaction network of PNES was constructed to discover its most influential compartments. The pathways associated with PNES pathomechanism were found by enrichment analysis of the identified proteins. Besides, the relationship between PNES-related molecules and psychiatric diseases was discovered, and the brain regions that could express altered levels of blood proteins were discovered. Results Eight genes and three hormones were found associated with PNES through the review process. Proopiomelanocortin (POMC), neuropeptide Y (NPY), cortisol, norepinephrine, and brain-derived neurotrophic factor (BDNF) were identified to have a high impact on the disease pathogenesis network. Moreover, activation of Janus kinase-signaling transducer and activator of transcription (JAK-STAT) and JAK, as well as signaling of growth hormone receptor, phosphatidylinositol 3-kinase /protein kinase B (PI3K/AKT), and neurotrophin were found associated with PNES molecular mechanism. Several psychiatric diseases such as depression, schizophrenia, and alcohol-related disorders were shown to be associated with PNES predominantly through signaling molecules. Significance This study was the first to gather the biochemicals associated with PNES. Multiple components and pathways and several psychiatric diseases associated with PNES, and some brain regions that could be altered during PNES were suggested, which should be confirmed in further studies. Altogether, these findings could be used in future molecular research on PNES patients.
Collapse
Affiliation(s)
- Mahdi Malekpour
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Jafari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Kashkooli
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Kazakou P, Nicolaides NC, Chrousos GP. Basic Concepts and Hormonal Regulators of the Stress System. Horm Res Paediatr 2023; 96:8-16. [PMID: 35272295 DOI: 10.1159/000523975] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/02/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Human organisms have to cope with a large number of external or internal stressful stimuli that threaten - or are perceived as threatening - their internal dynamic balance or homeostasis. To face these disturbing forces, or stressors, organisms have developed a complex neuroendocrine system, the stress system, which consists of the hypothalamic-pituitary-adrenal axis and the locus caeruleus/norepinephrine-autonomic nervous system. SUMMARY Upon exposure to stressors beyond a certain threshold, the activation of the stress system leads to a series of physiological and behavioral adaptations that help achieve homeostasis and increase the chances of survival. When, however, the stress response to stressors is inadequate, excessive, or prolonged, the resultant maladaptation may lead to the development of several stress-related pathologic conditions. Adverse environmental events, especially during critical periods of life, such as prenatal life, childhood, and puberty/adolescence, in combination with the underlying genetic background, may leave deep, long-term epigenetic imprints in the human expressed genome. KEY MESSAGES In this review, we describe the components of the stress system and its functional interactions with other homeostatic systems of the organism; we present the hormonal regulators of the stress response, and we discuss the development of stress-related pathologies.
Collapse
Affiliation(s)
- Paraskevi Kazakou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Endocrine Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nicolas C Nicolaides
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, Greece.,Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - George P Chrousos
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, Greece
| |
Collapse
|
14
|
Maier HB, Moschny N, Eberle F, Jahn K, Folsche T, Schülke R, Bleich S, Frieling H, Neyazi A. DNA Methylation of POMC and NR3C1-1F and Its Implication in Major Depressive Disorder and Electroconvulsive Therapy. PHARMACOPSYCHIATRY 2023; 56:64-72. [PMID: 36944329 PMCID: PMC10070046 DOI: 10.1055/a-2034-6536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Precision medicine in psychiatry is still in its infancy. To establish patient-tailored treatment, adequate indicators predicting treatment response are required. Electroconvulsive therapy (ECT) is considered one of the most effective options for pharmacoresistant major depressive disorder (MDD), yet remission rates were reported to be below 50%. METHODS Since epigenetics of the stress response system seem to play a role in MDD, we analyzed the DNA methylation (DNAm) of genes encoding the glucocorticoid receptor (NR3C1) and proopiomelanocortin (POMC) through Sanger Sequencing. For analysis, blood was taken before and after the first and last ECT from MDD patients (n=31), unmedicated depressed controls (UDC; n=19, baseline), and healthy controls (HC; n=20, baseline). RESULTS Baseline DNAm in NR3C1 was significantly lower in UDCs compared to both other groups (UDC: 0.014(±0.002), ECT: 0.031(±0.001), HC: 0.024(±0.002); p<0.001), whereas regarding POMC, ECT patients had the highest DNAm levels (ECT: 0.252(±0.013), UDC: 0.156(±0.015), HC: 0.162(±0.014); p<0.001). NR3C1m and POMCm decreased after the first ECT (NR3C1: p<0.001; POMC: p=0.001), and responders were less methylated compared to non-responders in NR3C1(p<0.001). DISCUSSION Our findings indicate that both genes might play a role in the chronification of depression and NR3C1 may be relevant for ECT response prediction.
Collapse
Affiliation(s)
- Hannah B Maier
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Nicole Moschny
- Laboratory for Molecular Neuroscience, Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Franziska Eberle
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Kirsten Jahn
- Laboratory for Molecular Neuroscience, Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Thorsten Folsche
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Rasmus Schülke
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Stefan Bleich
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
- Laboratory for Molecular Neuroscience, Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
- Laboratory for Molecular Neuroscience, Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Alexandra Neyazi
- Laboratory for Molecular Neuroscience, Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School, Hannover, Germany
- Department of Psychiatry and Psychotherapy, Otto von Guericke University Magdeburg (OVGU), Germany
| |
Collapse
|
15
|
Cosentino L, Zidda F, Dukal H, Witt SH, De Filippis B, Flor H. Low levels of Methyl-CpG binding protein 2 are accompanied by an increased vulnerability to the negative outcomes of stress exposure during childhood in healthy women. Transl Psychiatry 2022; 12:506. [PMID: 36481643 PMCID: PMC9731965 DOI: 10.1038/s41398-022-02259-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
Numerous mental illnesses arise following stressful events in vulnerable individuals, with females being generally more affected than males. Adverse childhood experiences are known to increase the risk of developing psychopathologies and DNA methylation was demonstrated to drive the long-lasting effects of early life stress and promote stress susceptibility. Methyl-CpG binding protein 2 (MECP2), an X-linked reader of the DNA methylome, is altered in many mental disorders of stress origin, suggesting MECP2 as a marker of stress susceptibility; previous works also suggest a link between MECP2 and early stress experiences. The present work explored whether a reduced expression of MECP2 is paralleled by an increased vulnerability to the negative outcomes of stress exposure during childhood. To this aim, blood MECP2 mRNA levels were analyzed in 63 people without history of mental disorders and traits pertaining to depressive and anxiety symptom clusters were assessed as proxies of the vulnerability to develop stress-related disorders; stress exposure during childhood was also evaluated. Using structural equation modeling, we demonstrate that reduced MECP2 expression is accompanied by symptoms of anxiety/depression in association with exposure to stress in early life, selectively in healthy women. These results suggest a gender-specific involvement of MECP2 in the maladaptive outcomes of childhood adversities, and shed new light on the complex biology underlying gender bias in stress susceptibility.
Collapse
Affiliation(s)
- Livia Cosentino
- grid.416651.10000 0000 9120 6856Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Roma, Italy ,grid.7700.00000 0001 2190 4373Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Francesca Zidda
- grid.7700.00000 0001 2190 4373Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Helene Dukal
- grid.7700.00000 0001 2190 4373Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H. Witt
- grid.7700.00000 0001 2190 4373Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Roma, Italy.
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
16
|
Paoli C, Misztak P, Mazzini G, Musazzi L. DNA Methylation in Depression and Depressive-Like Phenotype: Biomarker or Target of Pharmacological Intervention? Curr Neuropharmacol 2022; 20:2267-2291. [PMID: 35105292 PMCID: PMC9890294 DOI: 10.2174/1570159x20666220201084536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/18/2022] [Accepted: 01/26/2022] [Indexed: 12/29/2022] Open
Abstract
Major depressive disorder (MDD) is a debilitating psychiatric disorder, the third leading global cause of disability. Regarding aetiopathogenetic mechanisms involved in the onset of depressive disorders, the interaction between genetic vulnerability traits and environmental factors is believed to play a major role. Although much is still to be elucidated about the mechanisms through which the environment can interact with genetic background shaping the disease risk, there is a general agreement about a key role of epigenetic marking. In this narrative review, we focused on the association between changes in DNA methylation patterns and MDD or depressive-like phenotype in animal models, as well as mechanisms of response to antidepressant drugs. We discussed studies presenting DNA methylation changes at specific genes of interest and profiling analyses in both patients and animal models of depression. Overall, we collected evidence showing that DNA methylation could not only be considered as a promising epigenetic biomarker of pathology but could also help in predicting antidepressant treatment efficacy. Finally, we discussed the hypothesis that specific changes in DNA methylation signature could play a role in aetiopathogenetic processes as well as in the induction of antidepressant effect.
Collapse
Affiliation(s)
- Caterina Paoli
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- School of Pharmacy, Pharmacy Unit, University of Camerino, 62032 Camerino, Italy
| | - Paulina Misztak
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Giulia Mazzini
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Laura Musazzi
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
17
|
Early life adversity shapes neural circuit function during sensitive postnatal developmental periods. Transl Psychiatry 2022; 12:306. [PMID: 35915071 PMCID: PMC9343623 DOI: 10.1038/s41398-022-02092-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
Early life adversity (ELA) is a major risk factor for mental illness, but the neurobiological mechanisms by which ELA increases the risk for future psychopathology are still poorly understood. Brain development is particularly malleable during prenatal and early postnatal life, when complex neural circuits are being formed and refined through an interplay of excitatory and inhibitory neural input, synaptogenesis, synaptic pruning, myelination, and neurogenesis. Adversity that influences these processes during sensitive periods of development can thus have long-lasting and pervasive effects on neural circuit maturation. In this review, we will discuss clinical and preclinical evidence for the impact of ELA on neural circuit formation with a focus on the early postnatal period, and how long-lasting impairments in these circuits can affect future behavior. We provide converging evidence from human and animal studies on how ELA alters the functional development of brain regions, neural circuits, and neurotransmitter systems that are crucial for cognition and affective behavior, including the hippocampus, the hypothalamus-pituitary-adrenal (HPA) axis, neural networks of fear responses and cognition, and the serotonin (5-HT) system. We also discuss how gene-by-environment (GxE) interactions can determine individual differences in susceptibility and resilience to ELA, as well as molecular pathways by which ELA regulates neural circuit development, for which we emphasize epigenetic mechanisms. Understanding the molecular and neurobiological mechanisms underlying ELA effects on brain function and psychopathology during early postnatal sensitive periods may have great potential to advance strategies to better treat or prevent psychiatric disorders that have their origin early in life.
Collapse
|
18
|
Sánchez-Lafuente CL, Kalynchuk LE, Caruncho HJ, Ausió J. The Role of MeCP2 in Regulating Synaptic Plasticity in the Context of Stress and Depression. Cells 2022; 11:cells11040748. [PMID: 35203405 PMCID: PMC8870391 DOI: 10.3390/cells11040748] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Methyl-CpG-binding protein 2 (MeCP2) is a transcriptional regulator that is highly abundant in the brain. It binds to methylated genomic DNA to regulate a range of physiological functions implicated in neuronal development and adult synaptic plasticity. MeCP2 has mainly been studied for its role in neurodevelopmental disorders, but alterations in MeCP2 are also present in stress-related disorders such as major depression. Impairments in both stress regulation and synaptic plasticity are associated with depression, but the specific mechanisms underlying these changes have not been identified. Here, we review the interplay between stress, synaptic plasticity, and MeCP2. We focus our attention on the transcriptional regulation of important neuronal plasticity genes such as BDNF and reelin (RELN). Moreover, we provide evidence from recent studies showing a link between chronic stress-induced depressive symptoms and dysregulation of MeCP2 expression, underscoring the role of this protein in stress-related pathology. We conclude that MeCP2 is a promising target for the development of novel, more efficacious therapeutics for the treatment of stress-related disorders such as depression.
Collapse
Affiliation(s)
- Carla L. Sánchez-Lafuente
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada; (C.L.S.-L.); (L.E.K.); (H.J.C.)
| | - Lisa E. Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada; (C.L.S.-L.); (L.E.K.); (H.J.C.)
| | - Hector J. Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada; (C.L.S.-L.); (L.E.K.); (H.J.C.)
| | - Juan Ausió
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8W 3P6, Canada
- Correspondence:
| |
Collapse
|
19
|
Almeida MM, Dias-Rocha CP, Calviño C, Trevenzoli IH. Lipid endocannabinoids in energy metabolism, stress and developmental programming. Mol Cell Endocrinol 2022; 542:111522. [PMID: 34843899 DOI: 10.1016/j.mce.2021.111522] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022]
Abstract
The endocannabinoid system (ECS) regulates brain development and function, energy metabolism and stress in a sex-, age- and tissue-dependent manner. The ECS comprises mainly the bioactive lipid ligands anandamide (AEA) and 2-aracdonoylglycerol (2-AG), cannabinoid receptors 1 and 2 (CB1 and CB2), and several metabolizing enzymes. The endocannabinoid tonus is increased in obesity, stimulating food intake and a preference for fat, reward, and lipid accumulation in peripheral tissues, as well as favoring a positive energy balance. Energy balance and stress responses share adaptive mechanisms regulated by the ECS that seem to underlie the complex relationship between feeding and emotional behavior. The ECS is also a key regulator of development. Environmental insults (diet, toxicants, and stress) in critical periods of developmental plasticity, such as gestation, lactation and adolescence, alter the ECS and may predispose individuals to the development of chronic diseases and behavioral changes in the long term. This review is focused on the ECS and the developmental origins of health and disease (DOHaD).
Collapse
Affiliation(s)
- Mariana Macedo Almeida
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | | | - Camila Calviño
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, RJ, Brazil
| | - Isis Hara Trevenzoli
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
20
|
Li QS, Galbraith D, Morrison RL, Trivedi MH, Drevets WC. Circulating microRNA associated with future relapse status in major depressive disorder. Front Psychiatry 2022; 13:937360. [PMID: 36061300 PMCID: PMC9428445 DOI: 10.3389/fpsyt.2022.937360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/25/2022] [Indexed: 12/19/2022] Open
Abstract
Major depressive disorder (MDD) is an episodic condition with relapsing and remitting disease course. Elucidating biomarkers that can predict future relapse in individuals responding to an antidepressant treatment holds the potential to identify those patients who are prone to illness recurrence. The current study explored relationships between relapse risk in recurrent MDD and circulating microRNAs (miRNAs) that participate in RNA silencing and post-transcriptional regulation of gene expression. Serum samples were acquired from individuals with a history of recurrent MDD who were followed longitudinally in the observational study, OBSERVEMDD0001 (ClinicalTrials.gov Identifier: NCT02489305). Circulating miRNA data were obtained in 63 participants who relapsed ("relapsers") and 154 participants who did not relapse ("non-relapsers") during follow-up. The miRNA was quantified using the ID3EAL™ miRNA Discovery Platform from MiRXES measuring 575 circulating miRNAs using a patented qPCR technology and normalized with a standard curve from spike-in controls in each plate. The association between miRNAs and subsequent relapse was tested using a linear model, adjusting for age, gender, and plate. Four miRNAs were nominally associated with relapse status during the observational follow-up phase with a false discover rate adjusted p-value < 0.1. Enrichment analysis of experimentally validated targets revealed 112 significantly enriched pathways, including neurogenesis, response to cytokine, neurotrophin signaling, vascular endothelial growth factor signaling, relaxin signaling, and cellular senescence pathways. These data suggest these miRNAs putatively associated with relapse status may have the potential to regulate genes involved in multiple signaling pathways that have previously been associated with MDD. If shown to be significant in a larger, independent sample, these data may hold potential for developing a miRNA signature to identify patients likely to relapse, allowing for earlier intervention.
Collapse
Affiliation(s)
- Qingqin S Li
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, Titusville, NJ, United States.,JRD Data Science, Janssen Research and Development, LLC, Titusville, NJ, United States
| | | | - Randall L Morrison
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, Titusville, NJ, United States
| | - Madhukar H Trivedi
- Department of Psychiatry, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, United States
| | - Wayne C Drevets
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, CA, United States
| |
Collapse
|
21
|
Bobbo VC, Engel DF, Jara CP, Mendes NF, Haddad-Tovolli R, Prado TP, Sidarta-Oliveira D, Morari J, Velloso LA, Araujo EP. Interleukin-6 actions in the hypothalamus protects against obesity and is involved in the regulation of neurogenesis. J Neuroinflammation 2021; 18:192. [PMID: 34465367 PMCID: PMC8408946 DOI: 10.1186/s12974-021-02242-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/18/2021] [Indexed: 01/21/2023] Open
Abstract
Background Interleukin-6 (IL6) produced in the context of exercise acts in the hypothalamus reducing obesity-associated inflammation and restoring the control of food intake and energy expenditure. In the hippocampus, some of the beneficial actions of IL6 are attributed to its neurogenesis-inducing properties. However, in the hypothalamus, the putative neurogenic actions of IL6 have never been explored, and its potential to balance energy intake can be an approach to prevent or attenuate obesity. Methods Wild-type (WT) and IL6 knockout (KO) mice were employed to study the capacity of IL6 to induce neurogenesis. We used cell labeling with Bromodeoxyuridine (BrdU), immunofluorescence, and real-time PCR to determine the expression of markers of neurogenesis and neurotransmitters. We prepared hypothalamic neuroprogenitor cells from KO that were treated with IL6 in order to provide an ex vivo model to further characterizing the neurogenic actions of IL6 through differentiation assays. In addition, we analyzed single-cell RNA sequencing data and determined the expression of IL6 and IL6 receptor in specific cell types of the murine hypothalamus. Results IL6 expression in the hypothalamus is low and restricted to microglia and tanycytes, whereas IL6 receptor is expressed in microglia, ependymocytes, endothelial cells, and astrocytes. Exogenous IL6 reduces diet-induced obesity. In outbred mice, obesity-resistance is accompanied by increased expression of IL6 in the hypothalamus. IL6 induces neurogenesis-related gene expression in the hypothalamus and in neuroprogenitor cells, both from WT as well as from KO mice. Conclusion IL6 induces neurogenesis-related gene expression in the hypothalamus of WT mice. In KO mice, the neurogenic actions of IL6 are preserved; however, the appearance of new fully differentiated proopiomelanocortin (POMC) and neuropeptide Y (NPY) neurons is either delayed or disturbed. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02242-8.
Collapse
Affiliation(s)
- Vanessa C Bobbo
- Nursing School, University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Daiane F Engel
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Carlos Poblete Jara
- Nursing School, University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Natalia F Mendes
- Nursing School, University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Roberta Haddad-Tovolli
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Thais P Prado
- Nursing School, University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Davi Sidarta-Oliveira
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Joseane Morari
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Eliana P Araujo
- Nursing School, University of Campinas, Campinas, Brazil. .,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil.
| |
Collapse
|
22
|
Let's get wild: A review of free-ranging rat assays as context-enriched supplements to traditional laboratory models. J Neurosci Methods 2021; 362:109303. [PMID: 34352335 DOI: 10.1016/j.jneumeth.2021.109303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/21/2021] [Accepted: 07/29/2021] [Indexed: 01/30/2023]
Abstract
More than 24,000 rodent studies are published annually, with the vast majority of these studies focused on genetically undiverse animals in highly-controlled laboratory settings. However, findings from the laboratory have become increasingly unreliable for predicting outcomes in field and clinical settings, leading to a perceived crisis in translational research. One cause of this disparity might be that most human societies, in contrast to laboratory rodents, are genetically diverse and live in super-enriched environments. Methods for importing wild rats into the laboratory, and also exporting laboratory-style chambers into natural environments are not well-known outside their respective disciplines. Therefore, we have reviewed the current status of supplements to the laboratory rodent assay. We progress logically from highly-controlled experiments with natural breeding colonies to purely naturalistic approaches with free-ranging rats. We then highlight a number of approaches that allow genetically-diverse wild rats to be utilized in context-enriched paradigms. While considering the benefits and shortcomings of each available approach, we detail protocols for random sampling, remote-sensing, and deployment of laboratory chambers in the field. As supplements to standardized laboratory trials, some of these assays could offer key insights to help unify outcomes between laboratory and field studies. However, we note several outstanding questions that must be addressed such as: the trade-off between control and context, possible reductions in sample size, ramifications for the 'standardization fallacy', and ethical dilemmas of working with wild animals. Given these challenges, further innovation will be required before supplemental assays can be made broadly-accessible and thus, transferrable across disciplines.
Collapse
|
23
|
Jaimes-Hoy L, Pérez-Maldonado A, Narváez Bahena E, de la Cruz Guarneros N, Rodríguez-Rodríguez A, Charli JL, Soberón X, Joseph-Bravo P. Sex Dimorphic Changes in Trh Gene Methylation and Thyroid-Axis Response to Energy Demands in Maternally Separated Rats. Endocrinology 2021; 162:bqab110. [PMID: 34043769 DOI: 10.1210/endocr/bqab110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 12/18/2022]
Abstract
The hypothalamus-pituitary-thyroid (HPT) axis regulates energy balance through the pleiotropic action of thyroid hormones. HPT basal activity and stimulation by cold or voluntary exercise are repressed by previous chronic stress in adults. Maternal separation (MS) modifies HPT basal activity; we thus studied the response of the axis to energy demands and analyzed possible epigenetic changes on Trh promoter. Nonhandled (NH) or MS male Wistar rats were cold exposed 1 h at adulthood; Trh expression in the hypothalamic paraventricular nucleus (PVN) and serum thyrotropin (TSH) concentration were increased only in NH rats. Two weeks of voluntary exercise decreased fat mass and increased Trh expression, and thyroid hormones concentration changed proportionally to running distance in NH male rats and MS male rats. Although NH females ran more than MS and much more than males, exercise decreased body weight and fat mass only in NH rats with no change on any parameter of the HPT axis but increased Pomc expression in arcuate-nucleus of NH and Npy in MS females. Overall, the methylation pattern of PVN Trh gene promoter was similar in NH males and females; MS modified methylation of specific CpG sites, a thyroid hormone receptor (THR)-binding site present after the initiation site was hypomethylated in MS males; in MS females, the THR binding site of the proximal promoter (site 4) and 2 sites in the first intron were hypermethylated. Our studies showed that, in a sex-dimorphic manner, MS blunted the responses of HPT axis to energy demands in adult animals and caused methylation changes on Trh promoter that could alter T3 feedback.
Collapse
Affiliation(s)
- Lorraine Jaimes-Hoy
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Adrián Pérez-Maldonado
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Elian Narváez Bahena
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Natalia de la Cruz Guarneros
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Adair Rodríguez-Rodríguez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Jean-Louis Charli
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Xavier Soberón
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
- Instituto Nacional de Medicina Genómica, Ciudad de México, México
| | - Patricia Joseph-Bravo
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| |
Collapse
|
24
|
Abstract
Animal and humans exposed to stress early in life are more likely to suffer from long-term behavioral, mental health, metabolic, immune, and cardiovascular health consequences. The hypothalamus plays a nodal role in programming, controlling, and regulating stress responses throughout the life course. Epigenetic reprogramming in the hippocampus and the hypothalamus play an important role in adapting genome function to experiences and exposures during the perinatal and early life periods and setting up stable phenotypic outcomes. Epigenetic programming during development enables one genome to express multiple cell type identities. The most proximal epigenetic mark to DNA is a covalent modification of the DNA itself by enzymatic addition of methyl moieties. Cell-type-specific DNA methylation profiles are generated during gestational development and define cell and tissue specific phenotypes. Programming of neuronal phenotypes and sex differences in the hypothalamus is achieved by developmentally timed rearrangement of DNA methylation profiles. Similarly, other stations in the life trajectory such as puberty and aging involve predictable and scheduled reorganization of DNA methylation profiles. DNA methylation and other epigenetic marks are critical for maintaining cell-type identity in the brain, across the body, and throughout life. Data that have emerged in the last 15 years suggest that like its role in defining cell-specific phenotype during development, DNA methylation might be involved in defining experiential identities, programming similar genes to perform differently in response to diverse experiential histories. Early life stress impact on lifelong phenotypes is proposed to be mediated by DNA methylation and other epigenetic marks. Epigenetic marks, as opposed to genetic mutations, are reversible by either pharmacological or behavioral strategies and therefore offer the potential for reversing or preventing disease including behavioral and mental health disorders. This chapter discusses data testing the hypothesis that DNA methylation modulations of the HPA axis mediate the impact of early life stress on lifelong behavioral and physical phenotypes.
Collapse
Affiliation(s)
- Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
25
|
Lewis CR, Breitenstein RS, Henderson A, Sowards HA, Piras IS, Huentelman MJ, Doane LD, Lemery-Chalfant K. Harsh Parenting Predicts Novel HPA Receptor Gene Methylation and NR3C1 Methylation Predicts Cortisol Daily Slope in Middle Childhood. Cell Mol Neurobiol 2021; 41:783-793. [PMID: 32472381 PMCID: PMC11448560 DOI: 10.1007/s10571-020-00885-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/23/2020] [Indexed: 01/16/2023]
Abstract
Adverse experiences in childhood are associated with altered hypothalamic-pituitary-adrenal (HPA) axis function and negative health outcomes throughout life. It is now commonly accepted that abuse and neglect can alter epigenetic regulation of HPA genes. Accumulated evidence suggests harsh parenting practices such as spanking are also strong predictors of negative health outcomes. We predicted harsh parenting at 2.5 years old would predict HPA gene DNA methylation similarly to abuse and neglect, and cortisol output at 8.5 years old. Saliva samples were collected three times a day across 3 days to estimate cortisol diurnal slopes. Methylation was quantified using the Illumina Infinium MethylationEPIC array BeadChip (850 K) with DNA collected from buccal cells. We used principal components analysis to compute a summary statistic for CpG sites across candidate genes. The first and second components were used as outcome variables in mixed linear regression analyses with harsh parenting as a predictor variable. We found harsh parenting significantly predicted methylation of several HPA axis genes, including novel gene associations with AVPRB1, CRHR1, CRHR2, and MC2R (FDR corrected p < 0.05). Further, we found NR3C1 methylation predicted a steeper diurnal cortisol slope. Our results extend the current literature by demonstrating harsh parenting may influence DNA methylation similarly to more extreme early life experiences such as abuse and neglect. Further, we show NR3C1 methylation is associated with diurnal HPA function. Elucidating the molecular consequences of harsh parenting on health can inform best parenting practices and provide potential treatment targets for common complex disorders.
Collapse
Affiliation(s)
- Candace R Lewis
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA.
- Psychology, Arizona State University, Tempe, AZ, USA.
| | | | - Adrienne Henderson
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | | | - Ignazio S Piras
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | - Matthew J Huentelman
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | - Leah D Doane
- Psychology, Arizona State University, Tempe, AZ, USA
| | | |
Collapse
|
26
|
Detecting the effects of predator-induced stress on the global metabolism of an ungulate prey using fecal metabolomic fingerprinting. Sci Rep 2021; 11:6129. [PMID: 33731769 PMCID: PMC7971053 DOI: 10.1038/s41598-021-85600-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/01/2021] [Indexed: 01/31/2023] Open
Abstract
Few field tests have assessed the effects of predator-induced stress on prey fitness, particularly in large carnivore-ungulate systems. Because traditional measures of stress present limitations when applied to free-ranging animals, new strategies and systemic methodologies are needed. Recent studies have shown that stress and anxiety related behaviors can influence the metabolic activity of the gut microbiome in mammal hosts, and these metabolic alterations may aid in identification of stress. In this study, we used NMR-based fecal metabolomic fingerprinting to compare the fecal metabolome, a functional readout of the gut microbiome, of cattle herds grazing in low vs. high wolf-impacted areas within three wolf pack territories. Additionally, we evaluated if other factors (e.g., cattle nutritional state, climate, landscape) besides wolf presence were related to the variation in cattle metabolism. By collecting longitudinal fecal samples from GPS-collared cattle, we found relevant metabolic differences between cattle herds in areas where the probability of wolf pack interaction was higher. Moreover, cattle distance to GPS-collared wolves was the factor most correlated with this difference in cattle metabolism, potentially reflecting the variation in wolf predation risk. We further validated our results through a regression model that reconstructed cattle distances to GPS-collared wolves based on the metabolic difference between cattle herds. Although further research is needed to explore if similar patterns also hold at a finer scale, our results suggests that fecal metabolomic fingerprinting is a promising tool for assessing the physiological responses of prey to predation risk. This novel approach will help improve our knowledge of the consequences of predators beyond the direct effect of predation.
Collapse
|
27
|
Craig F, Tenuta F, Rizzato V, Costabile A, Trabacca A, Montirosso R. Attachment-related dimensions in the epigenetic era: A systematic review of the human research. Neurosci Biobehav Rev 2021; 125:654-666. [PMID: 33727029 DOI: 10.1016/j.neubiorev.2021.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 03/04/2021] [Accepted: 03/07/2021] [Indexed: 12/16/2022]
Abstract
In recent years, an increasing number of studies documented potential links between parental care and epigenetic mechanisms. The present systematic review focuses on the potential association and interrelationship between attachment-related dimensions and DNA methylation in human studies. We performed a literature review using electronic databases such as PubMed, Scopus, Web of Science, and EBSCOhost. Thirteen papers were included in the review. Findings support significant associations between attachment-related dimensions and epigenetic status in studies which considered different populations, age ranges, attachment measures and peripheral tissues. Although research in this area is still under investigation, available results suggest that DNA methylation associated with attachment-related dimensions might affect the development of stress regulation system and social-emotional capacities, thus contributing to the emerging phenotypic outcomes. However, identifying mediator and moderator effects in the interrelationship between these parameters was problematic owing to heterogeneous methodologies. Finally, we discuss clinical implications, unanswered questions, and future directions for human development in epigenetics research.
Collapse
Affiliation(s)
- Francesco Craig
- Scientific Institute, IRCCS E. Medea, Unit for Severe Disabilities in Developmental Age and Young Adults, Brindisi, Italy
| | - Flaviana Tenuta
- Department of Culture, Education and Society, University of Calabria, Cosenza, Italy
| | - Veronica Rizzato
- Scientific Institute, IRCCS E. Medea, Unit for Severe Disabilities in Developmental Age and Young Adults, Brindisi, Italy
| | - Angela Costabile
- Department of Culture, Education and Society, University of Calabria, Cosenza, Italy
| | - Antonio Trabacca
- Scientific Institute, IRCCS E. Medea, Unit for Severe Disabilities in Developmental Age and Young Adults, Brindisi, Italy.
| | - Rosario Montirosso
- Scientific Institute, IRCCS Eugenio Medea, 0-3 Center for the at-Risk Infant, Bosisio Parini, Italy
| |
Collapse
|
28
|
Cox OH, Song HY, Garrison-Desany HM, Gadiwalla N, Carey JL, Menzies J, Lee RS. Characterization of glucocorticoid-induced loss of DNA methylation of the stress-response gene Fkbp5 in neuronal cells. Epigenetics 2021; 16:1377-1397. [PMID: 33319620 DOI: 10.1080/15592294.2020.1864169] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Exposure to stress or glucocorticoids (GCs) is associated with epigenetic and transcriptional changes in genes that either mediate or are targets of GC signalling. FKBP5 (FK506 binding protein 5) is one such gene that also plays a central role in negative feedback regulation of GC signalling and several stress-related psychiatric disorders. In this study, we sought to examine how the mouse Fkbp5 gene is regulated in a neuronal context and identify requisite factors that can mediate the epigenetic sequelae of excess GC exposure. Mice treated with GCs were used to establish the widespread changes in DNA methylation (DNAm) and expression of Fkbp5 across four brain regions. Then two cell lines were used to test the persistence, decay, and functional significance of GC-induced methylation changes near two GC response elements (GREs) in the fifth intron of Fkbp5. We also tested the involvement of DNMT1, cell proliferation, and MeCP2 in mediating the effect of GCs on DNAm and gene activation. DNAm changes at some CpGs persist while others decay, and reduced methylation states are associated with a more robust transcriptional response. Importantly, the ability to undergo GC-induced DNAm loss is tied to DNMT1 function during cell division. Further, GC-induced DNAm loss is associated with reduced binding of MeCP2 at intron 5 and a physical interaction between the fifth intron and promoter of Fkbp5. Our results highlight several key factors at the Fkbp5 locus that may have important implications for GC- or stress-exposure during early stages of neurodevelopment.
Collapse
Affiliation(s)
- Olivia H Cox
- The Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ha Young Song
- The Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Henri M Garrison-Desany
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nuriya Gadiwalla
- The Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jenny L Carey
- The Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julia Menzies
- The Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard S Lee
- The Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Toorie AM, Vassoler FM, Qu F, Schonhoff CM, Bradburn S, Murgatroyd CA, Slonim DK, Byrnes EM. A history of opioid exposure in females increases the risk of metabolic disorders in their future male offspring. Addict Biol 2021; 26:e12856. [PMID: 31782234 DOI: 10.1111/adb.12856] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/24/2019] [Accepted: 11/03/2019] [Indexed: 12/20/2022]
Abstract
Worldwide consumption of opioids remains at historic levels. Preclinical studies report intergenerational effects on the endogenous opioid system of future progeny following preconception morphine exposure. Given the role of endogenous opioids in energy homeostasis, such effects could impact metabolism in the next generation. Thus, we examined diet-induced modifications in F1 male progeny of morphine-exposed female rats (MORF1). When fed a high fat-sugar diet (FSD) for 6 weeks, MORF1 males display features of emerging metabolic syndrome; they consume more food, gain more weight, and develop fasting-induced hyperglycemia and hyperinsulinemia. In the hypothalamus, proteins involved in energy homeostasis are modified and RNA sequencing revealed down-regulation of genes associated with neuronal plasticity, coupled with up-regulation of genes associated with immune, inflammatory, and metabolic processes that are specific to FSD-maintained MORF1 males. Thus, limited preconception morphine exposure in female rats increases the risk of metabolic syndrome/type 2 diabetes in the next generation.
Collapse
Affiliation(s)
- Anika M. Toorie
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine Tufts University North Grafton Massachusetts
- Department of Biology Rhode Island College North Providence Rhode Island
| | - Fair M. Vassoler
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine Tufts University North Grafton Massachusetts
| | - Fangfang Qu
- Department of Computer Science Tufts University Medford Massachusetts
| | - Christopher M. Schonhoff
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine Tufts University North Grafton Massachusetts
| | - Steven Bradburn
- Division of Biomedical Sciences Manchester Metropolitan University Manchester UK
| | | | - Donna K. Slonim
- Department of Computer Science Tufts University Medford Massachusetts
| | - Elizabeth M. Byrnes
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine Tufts University North Grafton Massachusetts
| |
Collapse
|
30
|
Li M, Fu X, Xie W, Guo W, Li B, Cui R, Yang W. Effect of Early Life Stress on the Epigenetic Profiles in Depression. Front Cell Dev Biol 2020; 8:867. [PMID: 33117794 PMCID: PMC7575685 DOI: 10.3389/fcell.2020.00867] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/11/2020] [Indexed: 12/23/2022] Open
Abstract
Depression is one of the most common mental disorders and has caused an overwhelming burden on world health. Abundant studies have suggested that early life stress may grant depressive-like phenotypes in adults. Childhood adversities that occurred in the developmental period amplified stress events in adulthood. Epigenetic-environment interaction helps to explain the role of early life stress on adulthood depression. Early life stress shaped the epigenetic profiles of the HPA axis, monoamine, and neuropeptides. In the context of early adversities increasing the risk of depression, early life stress decreased the activity of the glucocorticoid receptors, halted the circulation and production of serotonin, and reduced the molecules involved in modulating the neurogenesis and neuroplasticity. Generally, DNA methylation, histone modifications, and the regulation of non-coding RNAs programmed the epigenetic profiles to react to early life stress. However, genetic precondition, subtypes of early life stress, the timing of epigenetic status evaluated, demographic characteristics in humans, and strain traits in animals favored epigenetic outcomes. More research is needed to investigate the direct evidence for how early life stress-induced epigenetic changes contribute to the vulnerability of depression.
Collapse
Affiliation(s)
- Ming Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xiying Fu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wei Xie
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wanxu Guo
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
31
|
Abstract
Epigenetic mechanisms govern the transcription of the genome. Research with model systems reveals that environmental conditions can directly influence epigenetic mechanisms that are associated with interindividual differences in gene expression in brain and neural function. In this review, we provide a brief overview of epigenetic mechanisms and research with relevant rodent models. We emphasize more recent translational research programs in epigenetics as well as the challenges inherent in the integration of epigenetics into developmental and clinical psychology. Our objectives are to present an update with respect to the translational relevance of epigenetics for the study of psychopathology and to consider the state of current research with respect to its potential importance for clinical research and practice in mental health.
Collapse
Affiliation(s)
- Kieran J O'Donnell
- Department of Psychiatry and Sackler Program for Epigenetics and Psychobiology, McGill University, Montreal, Quebec H4H 1R3, Canada; .,Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec H3H 1R4, Canada.,Child and Brain Development Program, CIFAR, Toronto, Ontario M5G 1M1, Canada
| | - Michael J Meaney
- Department of Psychiatry and Sackler Program for Epigenetics and Psychobiology, McGill University, Montreal, Quebec H4H 1R3, Canada; .,Child and Brain Development Program, CIFAR, Toronto, Ontario M5G 1M1, Canada.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), 117609 Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore
| |
Collapse
|
32
|
Abstract
Depression is one of the most common psychiatric disorders affecting public health. Studies over the past years suggest that the methylations of some specific genes such as BDNF, SLC6A4, and NR3C1 play an important role in the development of depression. Recently, epigenetic evidences suggest that the expression levels of DNA methyltransferases differ in several brain areas including the prefrontal cortex, hippocampus, amygdala, and nucleus accumbens in depression patients and animal models, but the potential link between the expression levels of DNA methylatransferases and the methylations of specific genes needs further investigation to clarify the pathogenesis of depression.
Collapse
Affiliation(s)
- Zhenghao Duan
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jie Lu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
33
|
Ahi EP, Brunel M, Tsakoumis E, Schmitz M. Transcriptional study of appetite regulating genes in the brain of zebrafish (Danio rerio) with impaired leptin signalling. Sci Rep 2019; 9:20166. [PMID: 31882937 PMCID: PMC6934527 DOI: 10.1038/s41598-019-56779-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/10/2019] [Indexed: 12/26/2022] Open
Abstract
The hormone leptin is a key regulator of body weight, food intake and metabolism. In mammals, leptin acts as an anorexigen and inhibits food intake centrally by affecting the appetite centres in the hypothalamus. In teleost fish, the regulatory connections between leptin and other appetite-regulating genes are largely unknown. In the present study, we used a zebrafish mutant with a loss of function leptin receptor to investigate brain expression patterns of 12 orexigenic and 24 anorexigenic genes under different feeding conditions (normal feeding, 7-day fasting, 2 and 6-hours refeeding). Expression patterns were compared to wild-type zebrafish, in order to identify leptin-dependent differentially expressed genes under different feeding conditions. We provide evidence that the transcription of certain orexigenic and anorexigenic genes is influenced by leptin signalling in the zebrafish brain. We found that the expression of orexigenic genes was not affected by impaired leptin signalling under normal feeding conditions; however, several orexigenic genes showed increased transcription during fasting and refeeding, including agrp, apln, galr1a and cnr1. This suggests an inhibitory effect of leptin signal on the transcription of these orexigenic genes during short-term fasting and refeeding in functional zebrafish. Most pronounced effects were observed in the group of anorexigenic genes, where the impairment of leptin signalling resulted in reduced gene expression in several genes, including cart family, crhb, gnrh2, mc4r, pomc and spx, in the control group. This suggests a stimulatory effect of leptin signal on the transcription of these anorexigenic genes under normal feeding condition. In addition, we found multiple gain and loss in expression correlations between the appetite-regulating genes, in zebrafish with impaired leptin signal, suggesting the presence of gene regulatory networks downstream of leptin signal in zebrafish brain. The results provide the first evidence for the effects of leptin signal on the transcription of various appetite-regulating genes in zebrafish brain, under different feeding conditions. Altogether, these transcriptional changes suggest an anorexigenic role for leptin signal, which is likely to be mediated through distinct set of appetite-regulating genes under different feeding conditions.
Collapse
Affiliation(s)
- Ehsan Pashay Ahi
- Department of Organismal Biology, Comparative Physiology, Evolutionary Biology Centre, Uppsala University, Norbyvägen 18A, SE-752 36, Uppsala, Sweden
| | - Mathilde Brunel
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, BioCentrum, Allmas Allé 5, SE-750 07 Uppsala, Sweden
| | - Emmanouil Tsakoumis
- Department of Organismal Biology, Comparative Physiology, Evolutionary Biology Centre, Uppsala University, Norbyvägen 18A, SE-752 36, Uppsala, Sweden
| | - Monika Schmitz
- Department of Organismal Biology, Comparative Physiology, Evolutionary Biology Centre, Uppsala University, Norbyvägen 18A, SE-752 36, Uppsala, Sweden.
| |
Collapse
|
34
|
Tomasi J, Lisoway AJ, Zai CC, Harripaul R, Müller DJ, Zai GCM, McCabe RE, Richter MA, Kennedy JL, Tiwari AK. Towards precision medicine in generalized anxiety disorder: Review of genetics and pharmaco(epi)genetics. J Psychiatr Res 2019; 119:33-47. [PMID: 31563039 DOI: 10.1016/j.jpsychires.2019.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023]
Abstract
Generalized anxiety disorder (GAD) is a prevalent and chronic mental disorder that elicits widespread functional impairment. Given the high degree of non-response/partial response among patients with GAD to available pharmacological treatments, there is a strong need for novel approaches that can optimize outcomes, and lead to medications that are safer and more effective. Although investigations have identified interesting targets predicting treatment response through pharmacogenetics (PGx), pharmaco-epigenetics, and neuroimaging methods, these studies are often solitary, not replicated, and carry several limitations. This review provides an overview of the current status of GAD genetics and PGx and presents potential strategies to improve treatment response by combining better phenotyping with PGx and improved analytical methods. These strategies carry the dual benefit of delivering data on biomarkers of treatment response as well as pointing to disease mechanisms through the biology of the markers associated with response. Overall, these efforts can serve to identify clinical, genetic, and epigenetic factors that can be incorporated into a pharmaco(epi)genetic test that may ultimately improve treatment response and reduce the socioeconomic burden of GAD.
Collapse
Affiliation(s)
- Julia Tomasi
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Amanda J Lisoway
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Clement C Zai
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Ricardo Harripaul
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Molecular Neuropsychiatry & Development (MiND) Lab, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Daniel J Müller
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Gwyneth C M Zai
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; General Adult Psychiatry and Health Systems Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Randi E McCabe
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Anxiety Treatment and Research Clinic, St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada
| | - Margaret A Richter
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Frederick W. Thompson Anxiety Disorders Centre, Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - James L Kennedy
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Arun K Tiwari
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
35
|
Kumar A, Kumar P, Pareek V, Faiq MA, Narayan RK, Raza K, Prasoon P, Sharma VK. Neurotrophin mediated HPA axis dysregulation in stress induced genesis of psychiatric disorders: Orchestration by epigenetic modifications. J Chem Neuroanat 2019; 102:101688. [PMID: 31568825 DOI: 10.1016/j.jchemneu.2019.101688] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/22/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022]
Abstract
Apart from their established role in embryonic development, neurotrophins (NTs) have diverse functions in the nervous system. Their role in the integration of physiological and biochemical aspects of the nervous system is currently attracting much attention. Based on a systematic analysis of the literature, we here propose a new paradigm that, by exploiting a novel role of NTs, may help explain the genesis of stress-related psychiatric disorders, opening new avenues for better management of the same. We hypothesize that NTs as an integrated network play a crucial role in maintaining an indivdual's psychological wellbeing. Given the evidence that stress can induce chronic disruption of the hypothalamic-pituitary-adrenal (HPA) axis which, in turn, is causally linked to several psychiatric disorders, this function may be mediated through the homeostatic mechanisms governing regulation of this axis. In fact, NTs, such as nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) are known to participate in neuroendocrine regulation. Recent studies suggest epigenetic modification of NT-HPA axis interplay in the precipitation of psychiatric disorders. Our article highlights why this new knowledge regarding NTs should be considered in the etiogenesis and treatment of stress-induced psychopathology.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India; Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, India; Etiologically Elusive Disorders Research Network (EEDRN), India.
| | - Pavan Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, India; Developmental Neurogenetics Lab, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Vikas Pareek
- Computational Neuroscience and Neuroimaging Division, National Brain Research Centre (NBRC), Manesar, Haryana, India; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Muneeb A Faiq
- Neuroimaging and Visual Science Laboratory, New York University (NYU) Langone Health, NYU School of Medicine, New York, NY 10016, USA; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Ravi K Narayan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Khursheed Raza
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, India; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Pranav Prasoon
- Pittsburgh Centre for Pain Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Vivek K Sharma
- Department of Physiology, Government Institute of Medical Sciences (GIMS), Greater Noida, Kasna, Uttar Pradesh, India; Etiologically Elusive Disorders Research Network (EEDRN), India
| |
Collapse
|
36
|
Holmes L, Shutman E, Chinaka C, Deepika K, Pelaez L, Dabney KW. Aberrant Epigenomic Modulation of Glucocorticoid Receptor Gene (NR3C1) in Early Life Stress and Major Depressive Disorder Correlation: Systematic Review and Quantitative Evidence Synthesis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16214280. [PMID: 31689998 PMCID: PMC6861987 DOI: 10.3390/ijerph16214280] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/27/2019] [Accepted: 10/28/2019] [Indexed: 12/21/2022]
Abstract
Early life stress (ELS) induced by psychological trauma, child maltreatment, maternal separation, and domestic violence predisposes to psycho-behavioral pathologies during adulthood, namely major depressive disorder (MDD), anxiety, and bipolar affective disorder. While environmental data are available in illustrating this association, data remain to be established on the epigenomic underpinning of the nexus between ELS and MDD predisposition. Specifically, despite the observed aberrant epigenomic modulation of the NR3C1, a glucocorticoid receptor gene, in early social adversity and social threats in animal and human models, reliable scientific data for intervention mapping in reducing social adversity and improving human health is required. We sought to synthesize the findings of studies evaluating (a) epigenomic modulations, mainly DNA methylation resulting in MDD following ELS, (b) epigenomic modifications associated with ELS, and (c) epigenomic alterations associated with MDD. A systematic review and quantitative evidence synthesis (QES) were utilized with the random effect meta-analytic procedure. The search strategy involved both the PubMed and hand search of relevant references. Of the 1534 studies identified through electronic search, 592 studies were screened, 11 met the eligibility criteria for inclusion in the QES, and 5 examined ELS and MDD; 4 studies assessed epigenomic modulation and ELS, while 2 studies examined epigenomic modulations and MDD. The dense DNA methylation of the 1F exon of the NR3C1, implying the hypermethylated region of the glucocorticoid receptor gene, was observed in the nexus between ELS and MDD, common effect size (CES) = 14.96, 95%CI, 10.06-19.85. With respect to epigenomic modulation associated with child ELS, hypermethylation was observed, CES = 23.2%, 95%CI, 8.00-38.48. In addition, marginal epigenomic alteration was indicated in MDD, where hypermethylation was associated with increased risk of MDD, CES = 2.12%, 95%CI, -0.63-4.86. Substantial evidence supports the implication of NR3C1 and environmental interaction, mainly DNA methylation, in the predisposition to MDD following ELS. This QES further supports aberrant epigenomic modulation identified in ELS as well as major depressive episodes involving dysfunctional glucocorticoid-mediated negative feedback as a result of allostatic overload. These findings recommend prospective investigation of social adversity and its predisposition to the MDD epidemic via aberrant epigenomic modulation. Such data will facilitate early intervention mapping in reducing MDD in the United States population.
Collapse
Affiliation(s)
- Laurens Holmes
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Biological Sciences Department, University of Delaware, Newark, DE 19716, USA.
- College of Population Health, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Emily Shutman
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Biological Sciences Department, Haverford College, Haverford, PA 19041, USA.
| | - Chinacherem Chinaka
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Department of public health, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
- Community Environmental Health Department, Old Dominion University, Norfolk, VA 23507, USA.
| | - Kerti Deepika
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
| | - Lavisha Pelaez
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
| | - Kirk W Dabney
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
37
|
Holmes L, Chinaka C, Elmi H, Deepika K, Pelaez L, Enwere M, Akinola OT, Dabney KW. Implication of Spiritual Network Support System in Epigenomic Modulation and Health Trajectory. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E4123. [PMID: 31717711 PMCID: PMC6862316 DOI: 10.3390/ijerph16214123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/16/2022]
Abstract
With challenges in understanding the multifactorial etiologies of disease and individual treatment effect heterogeneities over the past four decades, much has been acquired on how physical, chemical and social environments affect human health, predisposing certain subpopulations to adverse health outcomes, especially the socio-environmentally disadvantaged (SED). Current translational data on gene and adverse environment interaction have revealed how adverse gene-environment interaction, termed aberrant epigenomic modulation, translates into impaired gene expression via messenger ribonucleic acid (mRNA) dysregulation, reflecting abnormal protein synthesis and hence dysfunctional cellular differentiation and maturation. The environmental influence on gene expression observed in most literature includes physical, chemical, physicochemical and recently social environment. However, data are limited on spiritual or religious environment network support systems, which reflect human psychosocial conditions and gene interaction. With this limited information, we aimed to examine the available data on spiritual activities characterized by prayers and meditation for a possible explanation of the nexus between the spiritual network support system (SNSS) as a component of psychosocial conditions, implicated in social signal transduction, and the gene expression correlate. With the intent to incorporate SNSS in human psychosocial conditions, we assessed the available data on bereavement, loss of spouse, loneliness, social isolation, low socio-economic status (SES), chronic stress, low social status, social adversity (SA) and early life stress (ELS), as surrogates for spiritual support network connectome. Adverse human psychosocial conditions have the tendency for impaired gene expression through an up-regulated conserved transcriptional response to adversity (CTRA) gene expression via social signal transduction, involving the sympathetic nervous system (SNS), beta-adrenergic receptors, the hypothalamus-pituitary-adrenal (HPA) axis and the glucocorticoid response. This review specifically explored CTRA gene expression and the nuclear receptor subfamily 3 group C member 1 (NR3C1) gene, a glucocorticoid receptor gene, in response to stress and the impaired negative feedback, given allostatic overload as a result of prolonged and sustained stress and social isolation as well as the implied social interaction associated with religiosity. While more remains to be investigated on psychosocial and immune cell response and gene expression, current data on human models do implicate appropriate gene expression via the CTRA and NR3C1 gene in the SNSS as observed in meditation, yoga and thai-chi, implicated in malignant neoplasm remission. However, prospective epigenomic studies in this context are required in the disease causal pathway, prognosis and survival, as well as cautious optimism in the application of these findings in clinical and public health settings, due to unmeasured and potential confoundings implicated in these correlations.
Collapse
Affiliation(s)
- Laurens Holmes
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Biological Sciences Department, University of Delaware, Newark, DE 19716, USA
- College of Population Health, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Chinacherem Chinaka
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Public Health Department, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Community and Environmental Health Department, Old Dominion University, Norfolk, VA 23507, USA
| | - Hikma Elmi
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
| | - Kerti Deepika
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
| | - Lavisha Pelaez
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
| | - Michael Enwere
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Public Health Department, Walden University, Minneapolis, MN 55401, USA
| | - Olumuyiwa T. Akinola
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Public Health Department, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Community and Environmental Health Department, Old Dominion University, Norfolk, VA 23507, USA
| | - Kirk W. Dabney
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
38
|
Molecular programs underlying differences in the expression of mood disorders in males and females. Brain Res 2019; 1719:89-103. [DOI: 10.1016/j.brainres.2019.05.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/20/2019] [Accepted: 05/13/2019] [Indexed: 01/13/2023]
|
39
|
Matthews SG, McGowan PO. Developmental programming of the HPA axis and related behaviours: epigenetic mechanisms. J Endocrinol 2019; 242:T69-T79. [PMID: 30917340 DOI: 10.1530/joe-19-0057] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 12/22/2022]
Abstract
It has been approximately 30 years since the seminal discoveries of David Barker and his colleagues, and research is beginning to unravel the mechanisms that underlie developmental programming. The early environment of the embryo, foetus and newborn have been clearly linked to altered hypothalamic-pituitary-adrenal (HPA) function and related behaviours through the juvenile period and into adulthood. A number of recent studies have shown that these effects can pass across multiple generations. The HPA axis is highly responsive to the environment, impacts both central and peripheral systems and is critical to health in a wide variety of contexts. Mechanistic studies in animals are linking early exposures to adversity with changes in gene regulatory mechanisms, including modifications of DNA methylation and altered levels of miRNA. Similar associations are emerging from recent human studies. These findings suggest that epigenetic mechanisms represent a fundamental link between adverse early environments and developmental programming of later disease. The underlying biological mechanisms that connect the perinatal environment with modified long-term health outcomes represent an intensive area of research. Indeed, opportunities for early interventions must identify the relevant environmental factors and their molecular targets. This new knowledge will likely assist in the identification of individuals who are at risk of developing poor outcomes and for whom early intervention is most effective.
Collapse
Affiliation(s)
- Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Departments of Obstetrics & Gynaecology and Medicine, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Patrick O McGowan
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, University of Toronto, Scarborough, Ontario, Canada
- Department of Cell and Systems Biology, Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Epigenetic regulation of POMC; implications for nutritional programming, obesity and metabolic disease. Front Neuroendocrinol 2019; 54:100773. [PMID: 31344387 DOI: 10.1016/j.yfrne.2019.100773] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 01/07/2023]
Abstract
Proopiomelanocortin (POMC) is a key mediator of satiety. Epigenetic marks such as DNA methylation may modulate POMC expression and provide a biological link between early life exposures and later phenotype. Animal studies suggest epigenetic marks at POMC are influenced by maternal energy excess and restriction, prenatal stress and Triclosan exposure. Postnatal factors including energy excess, folate, vitamin A, conjugated linoleic acid and leptin may also affect POMC methylation. Recent human studies suggest POMC DNA methylation is influenced by maternal nutrition in early pregnancy and associated with childhood and adult obesity. Studies in children propose a link between POMC DNA methylation and elevated lipids and insulin, independent of body habitus. This review brings together evidence from animal and human studies and suggests that POMC is sensitive to nutritional programming and is associated with a wide range of weight-related and metabolic outcomes.
Collapse
|
41
|
Early Developmental Stress Affects Subsequent Gene Expression Response to an Acute Stress in Atlantic Salmon: An Approach for Creating Robust Fish for Aquaculture? G3-GENES GENOMES GENETICS 2019; 9:1597-1611. [PMID: 30885921 PMCID: PMC6505151 DOI: 10.1534/g3.119.400152] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Stress during early life has potential to program and alter the response to stressful events and metabolism in later life. Repeated short exposure of Atlantic salmon to cold water and air during embryonic (E), post-hatch (PH) or both phases of development (EPH) has been shown to alter the methylome and transcriptome and to affect growth performance during later life compared to untreated controls (CO). The aim of this study was to investigate how the transcriptome of these fish responds to subsequent acute stress at the start feeding stage, and to describe methylation differences that might steer these changes. EPH treated fish showed the strongest down-regulation of corticotropin releasing factor 1, up-regulation of glucocorticoid receptor and 3-oxo-5-alpha-steroid 4-dehydrogenase 2 gene expression and a suppressed cortisol response 3 hr after the acute stress, differences that could influence hormesis and be affecting how EPH fish cope and recover from the stress event. Growth hormone 2 and insulin-like growth factor 1 were more strongly down-regulated following acute stress in EPH treated fish relative to E, PH and CO fish. This indicates switching away from growth toward coping with stress following stressful events in EPH fish. Genes implicated in immune function such as major histocompatibility class 1A, T-cell receptor and toll-like receptor also responded to acute stress differently in EPH treated fish, indicating that repeated stresses during early life may affect robustness. Differential DNA methylation was detected in regions mapping <500 bases from genes differentially responding to acute stress suggesting the involvement of epigenetic mechanisms. Stress treatments applied during early development therefore have potential as a husbandry tool for boosting the productivity of aquaculture by affecting how fish respond to stresses at critical stages of production.
Collapse
|
42
|
Armenta TC, Cole SW, Geschwind DH, Blumstein DT, Wayne RK. Gene expression shifts in yellow-bellied marmots prior to natal dispersal. Behav Ecol 2019; 30:267-277. [PMID: 30971856 PMCID: PMC6450206 DOI: 10.1093/beheco/ary175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 11/12/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
The causes and consequences of vertebrate natal dispersal have been studied extensively, yet little is known about the molecular mechanisms involved. We used RNA-seq to quantify transcriptomic gene expression in blood of wild yellow-bellied marmots (Marmota flaviventer) prior to dispersing from or remaining philopatric to their natal colony. We tested 3 predictions. First, we hypothesized dispersers and residents will differentially express genes and gene networks since dispersal is physiologically demanding. Second, we expected differentially expressed genes to be involved in metabolism, circadian processes, and immune function. Finally, in dispersing individuals, we predicted differentially expressed genes would change as a function of sampling date relative to dispersal date. We detected 150 differentially expressed genes, including genes that have critical roles in lipid metabolism and antigen defense. Gene network analysis revealed a module of 126 coexpressed genes associated with dispersal that was enriched for extracellular immune function. Of the dispersal-associated genes, 22 altered expression as a function of days until dispersal, suggesting that dispersal-associated genes do not initiate transcription on the same time scale. Our results provide novel insights into the fundamental molecular changes required for dispersal and suggest evolutionary conservation of functional pathways during this behavioral process.
Collapse
Affiliation(s)
- Tiffany C Armenta
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, Los Angeles, CA, USA
- Rocky Mountain Biological Laboratory, Crested Butte, CO, USA
| | - Steve W Cole
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Daniel H Geschwind
- Department of Psychiatry and Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Daniel T Blumstein
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, Los Angeles, CA, USA
- Rocky Mountain Biological Laboratory, Crested Butte, CO, USA
| | - Robert K Wayne
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
43
|
Jaimes-Hoy L, Romero F, Charli JL, Joseph-Bravo P. Sex Dimorphic Responses of the Hypothalamus-Pituitary-Thyroid Axis to Maternal Separation and Palatable Diet. Front Endocrinol (Lausanne) 2019; 10:445. [PMID: 31354623 PMCID: PMC6637657 DOI: 10.3389/fendo.2019.00445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/20/2019] [Indexed: 12/27/2022] Open
Abstract
Neonatal stress contributes to the development of obesity and has long-lasting effects on elements of the hypothalamus-pituitary-thyroid (HPT) axis. Given the importance of thyroid hormones in metabolic regulation, we studied the effects of maternal separation and a high-fat/high-carbohydrate diet (HFC), offered from puberty or adulthood, on HPT axis activity of adult male and female Wistar rats. Pups were non-handled (NH) or maternally separated (MS) 3 h/day at postnatal days (Pd) 2-21. In a first experiment, at Pd60, rats had access to chow or an HFC diet (cookies, peanuts, chow) for 1 month. Male and female NH and MS rats that consumed the HFC diet increased their caloric intake, body weight, and serum insulin levels; fat weight increased in all groups except in MS males, and serum leptin concentration increased only in females. Mediobasal hypothalamus (MBH) Pomc expression increased in NH-HFC females and Npy decreased in NH-HFC males. MS males showed insulinemia and hypercortisolemia that was attenuated by the HFC diet. The HPT axis activity response to an HFC diet was sex-specific; expression of MBH thyrotropin-releasing hormone-degrading ectoenzyme (Trhde) increased in NH and MS males; serum TSH concentration decreased in NH males, and T4 increased in NH females. In a second experiment, rats were fed chow or an HFC diet from Pd30 or 60 until Pd160 and exposed to 1 h restraint before sacrifice. Regardless of neonatal stress, age of diet exposition, or sex, the HFC diet increased body and fat weight and serum leptin concentration; it induced insulinemia in males, but in females only in Pd30 rats. The HFC diet's capacity to curtail the hypothalamus-pituitary-adrenal axis response to restraint was impaired in MS males. In restrained rats, expression of Trh in the paraventricular nucleus of the hypothalamus, Dio2 and Trhde in MBH, and serum thyroid hormone concentration were altered differently depending on sex, age of diet exposition, and neonatal stress. In conclusion, metabolic alterations associated to an HFC-diet-induced obesity are affected by sex or time of exposition, while various parameters of the HPT axis activity are additionally altered by MS, pointing to the complex interplay that these developmental influences exert on HPT axis activity in adult rats.
Collapse
|
44
|
Kokras N, Sotiropoulos I, Besinis D, Tzouveka EL, Almeida OFX, Sousa N, Dalla C. Neuroplasticity-related correlates of environmental enrichment combined with physical activity differ between the sexes. Eur Neuropsychopharmacol 2019; 29:1-15. [PMID: 30497839 DOI: 10.1016/j.euroneuro.2018.11.1107] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 11/01/2018] [Accepted: 11/09/2018] [Indexed: 01/08/2023]
Abstract
Environmental enrichment (EE), comprising positive physical (exercise) and cognitive stimuli, influences neuronal structure and usually improves brain function. The promise of EE as a preventative strategy against neuropsychiatric disease is especially high during early postnatal development when the brain is still amenable to reorganization. Despite the fact that male and female brains differ in terms of connectivity and function that may reflect early life experiences, knowledge of the neural substrates and mechanisms by which such changes arise remains limited. This study compared the impact of EE combined with physical activity on neuroplasticity and its functional consequences in adult male and female rats; EE was provided during the first 3 months of life and our analysis focused on the hippocampus, an area implicated in cognitive behavior as well as the neuroendocrine response to stress. Both male and female rats reared in EE displayed better object recognition memory than their control counterparts. Interestingly, sex differences were revealed in the effects of EE on time spent exploring the objects during this test. Independently of sex, EE increased hippocampal turnover rates of dopamine and serotonin and reduced expression of 5-HT1A receptors; in addition, EE upregulated expression of synaptophysin, a presynaptic protein, in the hippocampus. As compared to their respective controls, EE-exposed males exhibited parallel increases in phosphorylated Tau and the GluN2B receptor, whereas females responded to EE with reduced hippocampal levels of glutamate and GluN2B. Together, these observations provide further evidence on the differential effects of EE on markers of hippocampal neuroplasticity in males and females.
Collapse
Affiliation(s)
- N Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece; First Department of Psychiatry, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - I Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal; Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | - D Besinis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | - E L Tzouveka
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | | | - N Sousa
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - C Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece.
| |
Collapse
|
45
|
Sahara Y, Matsuzawa D, Ishii D, Fuchida T, Goto T, Sutoh C, Shimizu E. Paternal methyl donor deficient diets during development affect male offspring behavior and memory-related gene expression in mice. Dev Psychobiol 2018; 61:17-28. [DOI: 10.1002/dev.21801] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 09/28/2018] [Accepted: 09/29/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Yuji Sahara
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine; Chiba University; Chiba Japan
| | - Daisuke Matsuzawa
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine; Chiba University; Chiba Japan
- Research Center for Child Mental Development; Chiba University; Chiba Japan
| | - Daisuke Ishii
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine; Chiba University; Chiba Japan
- Center for Medical Sciences; Ibaraki Prefectural University of Health Sciences; Ibaraki Japan
| | - Takahiro Fuchida
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine; Chiba University; Chiba Japan
| | - Takashi Goto
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine; Chiba University; Chiba Japan
| | - Chihiro Sutoh
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine; Chiba University; Chiba Japan
- Research Center for Child Mental Development; Chiba University; Chiba Japan
| | - Eiji Shimizu
- Department of Cognitive Behavioral Physiology, Graduate School of Medicine; Chiba University; Chiba Japan
- Research Center for Child Mental Development; Chiba University; Chiba Japan
| |
Collapse
|
46
|
Abstract
Prenatal adversity shapes child neurodevelopment and risk for later mental health problems. The quality of the early care environment can buffer some of the negative effects of prenatal adversity on child development. Retrospective studies, in adult samples, highlight epigenetic modifications as sentinel markers of the quality of the early care environment; however, comparable data from pediatric cohorts are lacking. Participants were drawn from the Maternal Adversity Vulnerability and Neurodevelopment (MAVAN) study, a longitudinal cohort with measures of infant attachment, infant development, and child mental health. Children provided buccal epithelial samples (mean age = 6.99, SD = 1.33 years, n = 226), which were used for analyses of genome-wide DNA methylation and genetic variation. We used a series of linear models to describe the association between infant attachment and (a) measures of child outcome and (b) DNA methylation across the genome. Paired genetic data was used to determine the genetic contribution to DNA methylation at attachment-associated sites. Infant attachment style was associated with infant cognitive development (Mental Development Index) and behavior (Behavior Rating Scale) assessed with the Bayley Scales of Infant Development at 36 months. Infant attachment style moderated the effects of prenatal adversity on Behavior Rating Scale scores at 36 months. Infant attachment was also significantly associated with a principal component that accounted for 11.9% of the variation in genome-wide DNA methylation. These effects were most apparent when comparing children with a secure versus a disorganized attachment style and most pronounced in females. The availability of paired genetic data revealed that DNA methylation at approximately half of all infant attachment-associated sites was best explained by considering both infant attachment and child genetic variation. This study provides further evidence that infant attachment can buffer some of the negative effects of early adversity on measures of infant behavior. We also highlight the interplay between infant attachment and child genotype in shaping variation in DNA methylation. Such findings provide preliminary evidence for a molecular signature of infant attachment and may help inform attachment-focused early intervention programs.
Collapse
|
47
|
Lacal I, Ventura R. Epigenetic Inheritance: Concepts, Mechanisms and Perspectives. Front Mol Neurosci 2018; 11:292. [PMID: 30323739 PMCID: PMC6172332 DOI: 10.3389/fnmol.2018.00292] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/02/2018] [Indexed: 01/07/2023] Open
Abstract
Parents’ stressful experiences can influence an offspring’s vulnerability to many pathological conditions, including psychopathologies, and their effects may even endure for several generations. Nevertheless, the cause of this phenomenon has not been determined, and only recently have scientists turned to epigenetics to answer this question. There is extensive literature on epigenetics, but no consensus exists with regard to how and what can (and must) be considered to study and define epigenetics processes and their inheritance. In this work, we aimed to clarify and systematize these concepts. To this end, we analyzed the dynamics of epigenetic changes over time in detail and defined three types of epigenetics: a direct form of epigenetics (DE) and two indirect epigenetic processes—within (WIE) and across (AIE). DE refers to changes that occur in the lifespan of an individual, due to direct experiences with his environment. WIE concerns changes that occur inside of the womb, due to events during gestation. Finally, AIE defines changes that affect the individual’s predecessors (parents, grandparents, etc.), due to events that occur even long before conception and that are somehow (e.g., through gametes, the intrauterine environment setting) transmitted across generations. This distinction allows us to organize the main body of epigenetic evidence according to these categories and then focus on the latter (AIE), referring to it as a faster route of informational transmission across generations—compared with genetic inheritance—that guides human evolution in a Lamarckian (i.e., experience-dependent) manner. Of the molecular processes that are implicated in this phenomenon, well-known (methylation) and novel (non-coding RNA, ncRNA) regulatory mechanisms are converging. Our discussion of the chief methods that are used to study epigenetic inheritance highlights the most compelling technical and theoretical problems of this discipline. Experimental suggestions to expand this field are provided, and their practical and ethical implications are discussed extensively.
Collapse
Affiliation(s)
- Irene Lacal
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Rossella Ventura
- Department of Psychology and "Daniel Bovet" Center, Sapienza University of Rome, Rome, Italy.,Fondazione Santa Lucia, IRCCS, Rome, Italy
| |
Collapse
|
48
|
The reduction in sexual behavior induced by neonatal immune stress is not related to androgen levels in male rats. Int J Dev Neurosci 2018; 71:163-171. [DOI: 10.1016/j.ijdevneu.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 08/08/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022] Open
|
49
|
Kvist J, Gonçalves Athanàsio C, Shams Solari O, Brown JB, Colbourne JK, Pfrender ME, Mirbahai L. Pattern of DNA Methylation in Daphnia: Evolutionary Perspective. Genome Biol Evol 2018; 10:1988-2007. [PMID: 30060190 PMCID: PMC6097596 DOI: 10.1093/gbe/evy155] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2018] [Indexed: 02/06/2023] Open
Abstract
DNA methylation is an evolutionary ancient epigenetic modification that is phylogenetically widespread. Comparative studies of the methylome across a diverse range of non-conventional and conventional model organisms is expected to help reveal how the landscape of DNA methylation and its functions have evolved. Here, we explore the DNA methylation profile of two species of the crustacean Daphnia using whole genome bisulfite sequencing. We then compare our data with the methylomes of two insects and two mammals to achieve a better understanding of the function of DNA methylation in Daphnia. Using RNA-sequencing data for all six species, we investigate the correlation between DNA methylation and gene expression. DNA methylation in Daphnia is mainly enriched within the coding regions of genes, with the highest methylation levels observed at exons 2–4. In contrast, vertebrate genomes are globally methylated, and increase towards the highest methylation levels observed at exon 2, and maintained across the rest of the gene body. Although DNA methylation patterns differ among all species, their methylation profiles share a bimodal distribution across the genomes. Genes with low levels of CpG methylation and gene expression are mainly enriched for species specific genes. In contrast, genes associated with high methylated CpG sites are highly transcribed and evolutionary conserved across all species. Finally, the positive correlation between internal exons and gene expression potentially points to an evolutionary conserved mechanism, whereas the negative regulation of gene expression via methylation of promoters and exon 1 is potentially a secondary mechanism that has been evolved in vertebrates.
Collapse
Affiliation(s)
- Jouni Kvist
- School of Biosciences, University of Birmingham, United Kingdom
| | | | | | - James B Brown
- Department of Statistics, University of California, Berkeley.,Centre for Computational Biology (CCB), University of Birmingham, United Kingdom
| | | | - Michael E Pfrender
- Department of Biological Sciences and Environmental Change Initiative, University of Notre Dame
| | - Leda Mirbahai
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
50
|
Alvarez-Monjaras M, Mayes LC, Potenza MN, Rutherford HJ. A developmental model of addictions: integrating neurobiological and psychodynamic theories through the lens of attachment. Attach Hum Dev 2018; 21:616-637. [PMID: 30021489 DOI: 10.1080/14616734.2018.1498113] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although substance use and abuse may impact brain and behavior, it is still unclear why some people become addicted while others do not. Neuroscientific theories explain addiction as a series of between- and within-system neuroadaptations that lead to an increasingly dysregulating cycle, affecting reward, motivation, and executive control systems. In contrast, psychoanalysis understands addiction through a relational perspective wherein there is an underlying failure in affect regulation, a capacity shaped early developmentally. Considering recent findings suggesting the neurobiological overlap of addiction and attachment, it may be possible to integrate both perspectives into a developmental model through the lens of attachment. The goal of the present review is to evaluate the value of neurobiological and psychodynamic perspectives to inform our understanding of addiction, particularly substance-use disorders.
Collapse
Affiliation(s)
- Mauricio Alvarez-Monjaras
- Yale Child Study Center, Yale University School of Medicine , New Haven , CT.,Department of Clinical, Educational, and Health Psychology, University College London , London , UK
| | - Linda C Mayes
- Yale Child Study Center, Yale University School of Medicine , New Haven , CT
| | - Marc N Potenza
- Yale Child Study Center, Yale University School of Medicine , New Haven , CT.,Departments of Psychiatry and Neuroscience, Yale University School of Medicine , New Haven , CT.,Connecticut Council on Problem Gambling , Wethersfield , CT.,Connecticut Mental Health Center , New Haven , CT
| | | |
Collapse
|