1
|
Sălcudean A, Popovici RA, Pitic DE, Sârbu D, Boroghina A, Jomaa M, Salehi MA, Kher AAM, Lica MM, Bodo CR, Enatescu VR. Unraveling the Complex Interplay Between Neuroinflammation and Depression: A Comprehensive Review. Int J Mol Sci 2025; 26:1645. [PMID: 40004109 PMCID: PMC11855341 DOI: 10.3390/ijms26041645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
The relationship between neuroinflammation and depression is a complex area of research that has garnered significant attention in recent years. Neuroinflammation, characterized by the activation of glial cells and the release of pro-inflammatory cytokines, has been implicated in the pathophysiology of depression. The relationship between neuroinflammation and depression is bidirectional; not only can inflammation contribute to the onset of depressive symptoms, but depression itself can also exacerbate inflammatory responses, creating a vicious cycle that complicates treatment and recovery. The present comprehensive review aimed to explore the current findings on the interplay between neuroinflammation and depression, as well as the mechanisms, risk factors, and therapeutic implications. The mechanisms by which neuroinflammation induces depressive-like behaviors are diverse. Neuroinflammation can increase pro-inflammatory cytokines, activate the hypothalamus-pituitary-adrenal (HPA) axis, and impair serotonin synthesis, all of which contribute to depressive symptoms. Furthermore, the activation of microglia has been linked to the release of inflammatory mediators that can disrupt neuronal function and contribute to mood disorders. Stress-induced neuroinflammatory responses can lead to the release of pro-inflammatory cytokines that not only affect brain function but also influence behavior and mood. Understanding these mechanisms is crucial for developing targeted therapies that can mitigate the effects of neuroinflammation on mood disorders.
Collapse
Affiliation(s)
- Andreea Sălcudean
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.S.); (M.M.L.); (C.R.B.)
| | - Ramona-Amina Popovici
- Department of Management and Communication in Dental Medicine, Department I, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania;
| | - Dana Emanuela Pitic
- Department of Management and Communication in Dental Medicine, Department I, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania;
| | - Diana Sârbu
- Doctoral School of Pharmacy, Victor Babes University of Medicine and Pharmacy of Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Adela Boroghina
- Doctoral School of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania; (A.B.); (M.J.); (M.A.S.); (A.A.M.K.)
| | - Mohammad Jomaa
- Doctoral School of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania; (A.B.); (M.J.); (M.A.S.); (A.A.M.K.)
| | - Matin Asad Salehi
- Doctoral School of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania; (A.B.); (M.J.); (M.A.S.); (A.A.M.K.)
| | - Alsayed Ahmad Mhd Kher
- Doctoral School of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania; (A.B.); (M.J.); (M.A.S.); (A.A.M.K.)
| | - Maria Melania Lica
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.S.); (M.M.L.); (C.R.B.)
| | - Cristina Raluca Bodo
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.S.); (M.M.L.); (C.R.B.)
| | - Virgil Radu Enatescu
- Department of Psychiatry, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania;
| |
Collapse
|
2
|
Kremer TL, Chen J, Buhl A, Berhe O, Bilek E, Geiger LS, Ma R, Moessnang C, Reichert M, Reinhard I, Schwarz K, Schweiger JI, Streit F, Witt SH, Zang Z, Zhang X, Nöthen MM, Rietschel M, Ebner-Priemer UW, Schwarz E, Meyer-Lindenberg A, Braun U, Tost H. Multimodal Associations of FKBP5 Methylation With Emotion-Regulatory Brain Circuits. Biol Psychiatry 2024; 96:858-867. [PMID: 38460581 DOI: 10.1016/j.biopsych.2024.03.003] [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: 08/16/2023] [Revised: 02/02/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND Understanding the biological processes that underlie individual differences in emotion regulation and stress responsivity is a key challenge for translational neuroscience. The gene FKBP5 is a core regulator in molecular stress signaling that is implicated in the development of psychiatric disorders. However, it remains unclear how FKBP5 DNA methylation in peripheral blood is related to individual differences in measures of neural structure and function and their relevance to daily-life stress responsivity. METHODS Here, we characterized multimodal correlates of FKBP5 DNA methylation by combining epigenetic data with neuroimaging and ambulatory assessment in a sample of 395 healthy individuals. RESULTS First, we showed that FKBP5 demethylation as a psychiatric risk factor was related to an anxiety-associated reduction of gray matter volume in the ventromedial prefrontal cortex, a brain area that is involved in emotion regulation and mental health risk and resilience. This effect of epigenetic upregulation of FKBP5 on neuronal structure is more pronounced where FKBP5 is epigenetically downregulated at baseline. Leveraging 208 functional magnetic resonance imaging scans during a well-established emotion-processing task, we found that FKBP5 DNA methylation in peripheral blood was associated with functional differences in prefrontal-limbic circuits that modulate affective responsivity to daily stressors, which we measured using ecological momentary assessment in daily life. CONCLUSIONS Overall, we demonstrated how FKBP5 contributes to interindividual differences in neural and real-life affect regulation via structural and functional changes in prefrontal-limbic brain circuits.
Collapse
Affiliation(s)
- Thomas L Kremer
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Junfang Chen
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anais Buhl
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Oksana Berhe
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Edda Bilek
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Lena S Geiger
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ren Ma
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carolin Moessnang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Markus Reichert
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mental mHealth Lab, Chair of Applied Psychology, Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany; Department of eHealth and Sports Analytics, Ruhr University Bochum, Bochum, Germany
| | - Iris Reinhard
- Department of Biostatistics, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kristina Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Janina I Schweiger
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Fabian Streit
- DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H Witt
- DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Zhenxiang Zang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Xiaolong Zhang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany; Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Marcella Rietschel
- DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ulrich W Ebner-Priemer
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Mental mHealth Lab, Chair of Applied Psychology, Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Urs Braun
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Heike Tost
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany.
| |
Collapse
|
3
|
Clarke SA, Eng PC, Comninos AN, Lazarus K, Choudhury S, Tsang C, Meeran K, Tan TM, Dhillo WS, Abbara A. Current Challenges and Future Directions in the Assessment of Glucocorticoid Status. Endocr Rev 2024; 45:795-817. [PMID: 38795365 PMCID: PMC11581704 DOI: 10.1210/endrev/bnae016] [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: 08/04/2023] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 05/27/2024]
Abstract
Glucocorticoid (GC) hormones are secreted in a circadian and ultradian rhythm and play a critical role in maintaining physiological homeostasis, with both excess and insufficient GC associated with adverse effects on health. Current assessment of GC status is primarily clinical, often in conjunction with serum cortisol values, which may be stimulated or suppressed depending on the GC disturbance being assessed. In the setting of extreme perturbations in cortisol levels ie, markedly low or high levels, symptoms and signs of GC dysfunction may be overt. However, when disturbances in cortisol GC status values are less extreme, such as when assessing optimization of a GC replacement regimen, signs and symptoms can be more subtle or nonspecific. Current tools for assessing GC status are best suited to identifying profound disturbances but may lack sensitivity for confirming optimal GC status. Moreover, single cortisol values do not necessarily reflect an individual's GC status, as they are subject to inter- and intraindividual variation and do not take into account the pulsatile nature of cortisol secretion, variation in binding proteins, or local tissue concentrations as dictated by 11beta-hydroxysteroid dehydrogenase activity, as well as GC receptor sensitivity. In the present review, we evaluate possible alternative methods for the assessment of GC status that do not solely rely on the measurement of circulating cortisol levels. We discuss the potential of changes in metabolomic profiles, micro RNA, gene expression, and epigenetic and other novel biomarkers such as growth differentiating factor 15 and osteocalcin, which could in the future aid in the objective classification of GC status.
Collapse
Affiliation(s)
- Sophie A Clarke
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Pei Chia Eng
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
- Department of Endocrinology, National University of Singapore, Singapore
| | - Alexander N Comninos
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Katharine Lazarus
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Sirazum Choudhury
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Christie Tsang
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
| | - Karim Meeran
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Tricia M Tan
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Waljit S Dhillo
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Ali Abbara
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| |
Collapse
|
4
|
Lee A, Thuras P, Baller J, Jiao C, Guo B, Erbes CR, Polusny MA, Liu C, Wu B, Lim KO, Bishop JR. Serotonin Transporter (SLC6A4) and FK506-Binding Protein 5 (FKBP5) Genotype and Methylation Relationships with Response to Meditation in Veterans with PTSD. Mol Neurobiol 2024; 61:9608-9622. [PMID: 38671329 DOI: 10.1007/s12035-024-04096-6] [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: 11/21/2022] [Accepted: 03/04/2024] [Indexed: 04/28/2024]
Abstract
Meditation-based interventions are novel and effective non-pharmacologic treatments for veterans with PTSD. We examined relationships between treatment response, early life trauma exposure, DNA polymorphisms, and methylation in the serotonin transporter (SLC6A4) and FK506-binding protein 5 (FKBP5) genes. DNA samples and clinical outcomes were examined in 72 veterans with PTSD who received meditation-based therapy in two separate studies of mindfulness-based stress reduction (MBSR) and Transcendental Meditation (TM). The PTSD Checklist was administered to assess symptoms at baseline and after 9 weeks of meditation intervention. We examined the SLC6A4 promoter (5HTTLPR_L/S insertion/deletion + rs25531_A/G) polymorphisms according to previously defined gene expression groups, and the FKBP5 variant rs1360780 previously associated with PTSD disease risk. Methylation for CpG sites of SLC6A4 (28 sites) and FKBP5 (45 sites) genes was quantified in DNA samples collected before and after treatment. The 5HTTLPR LALA high expression genotype was associated with greater symptom improvement in participants exposed to early life trauma (p = 0.015). Separately, pre to post-treatment change of DNA methylation in a group of nine FKBP5 CpG sites was associated with greater symptom improvement (OR = 2.8, 95% CI 1.1-7.1, p = 0.027). These findings build on a wealth of existing knowledge regarding epigenetic and genetic relationships with PTSD disease risk to highlight the potential importance of SLC6A4 and FKBP5 for treatment mechanisms and as biomarkers of symptom improvement.
Collapse
Affiliation(s)
- Adam Lee
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Room 7-115 Weaver-Densford Hall, 308 Harvard St SE, Minneapolis, MN, 55455, USA
| | - Paul Thuras
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
| | - Joshua Baller
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Chuan Jiao
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team Krebs, Université Paris Cité, 75014, Paris, France
| | - Bin Guo
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Christopher R Erbes
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Center for Care Delivery and Outcomes Research, Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
| | - Melissa A Polusny
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Center for Care Delivery and Outcomes Research, Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
| | - Chunyu Liu
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Baolin Wu
- Department of Epidemiology and Biostatistics, Program in Public Health, University of California-Irvine, Irvine, CA, USA
| | - Kelvin O Lim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Geriatric Research, Education, and Clinical Center, Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Room 7-115 Weaver-Densford Hall, 308 Harvard St SE, Minneapolis, MN, 55455, USA.
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Bolouki A. Role of Epigenetic Modification in the Intergeneration Transmission of War Trauma. Indian J Clin Biochem 2024; 39:312-321. [PMID: 39005862 PMCID: PMC11239641 DOI: 10.1007/s12291-023-01136-1] [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: 09/13/2022] [Accepted: 04/25/2023] [Indexed: 07/16/2024]
Abstract
War trauma has been linked to changes in the neuroendocrine and immunological systems and increases the risk of physical disorders. Traumatic events during the war may have long-term repercussions on psychological and biological parameters in future generations, implying that traumatic stress may have transgenerational consequences. This article addresses how epigenetic mechanisms, which are a key biological mechanism for dynamic adaptation to environmental stressors, may help explain the long-term and transgenerational consequences of trauma. In war survivors, epigenetic changes in genes mediating the hypothalamus-pituitary-adrenal axis, as well as the immune system, have been reported. These genetic modifications may cause long-term changes in the stress response as well as physical health risks. Also, the finding of biomarkers for diagnosing the possibility of psychiatric illnesses in people exposed to stressful conditions such as war necessitates extensive research. While epigenetic research has the potential to further our understanding of the effects of trauma, the findings must be interpreted with caution because epigenetic molecular mechanisms is only one piece of a complicated puzzle of interwoven biological and environmental components.
Collapse
Affiliation(s)
- Ayeh Bolouki
- Clinical Biochemistry Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Research Unit on Cellular Biology (URBC), University of Namur, Namur, Belgium
| |
Collapse
|
6
|
Paes T, Feelders RA, Hofland LJ. Epigenetic Mechanisms Modulated by Glucocorticoids With a Focus on Cushing Syndrome. J Clin Endocrinol Metab 2024; 109:e1424-e1433. [PMID: 38517306 PMCID: PMC11099489 DOI: 10.1210/clinem/dgae151] [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: 08/22/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024]
Abstract
In Cushing syndrome (CS), prolonged exposure to high cortisol levels results in a wide range of devastating effects causing multisystem morbidity. Despite the efficacy of treatment leading to disease remission and clinical improvement, hypercortisolism-induced complications may persist. Since glucocorticoids use the epigenetic machinery as a mechanism of action to modulate gene expression, the persistence of some comorbidities may be mediated by hypercortisolism-induced long-lasting epigenetic changes. Additionally, glucocorticoids influence microRNA expression, which is an important epigenetic regulator as it modulates gene expression without changing the DNA sequence. Evidence suggests that chronically elevated glucocorticoid levels may induce aberrant microRNA expression which may impact several cellular processes resulting in cardiometabolic disorders. The present article reviews the evidence on epigenetic changes induced by (long-term) glucocorticoid exposure. Key aspects of some glucocorticoid-target genes and their implications in the context of CS are described. Lastly, the effects of epigenetic drugs influencing glucocorticoid effects are discussed for their ability to be potentially used as adjunctive therapy in CS.
Collapse
Affiliation(s)
- Ticiana Paes
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston 02115, MA, USA
| | - Richard A Feelders
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Leo J Hofland
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
7
|
Krumpolec P, Kodada D, Hadžega D, Petrovič O, Babišová K, Dosedla E, Turcsányiová Z, Minárik G. Changes in DNA methylation associated with a specific mode of delivery: a pilot study. Front Med (Lausanne) 2024; 11:1291429. [PMID: 38314203 PMCID: PMC10835804 DOI: 10.3389/fmed.2024.1291429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/05/2024] [Indexed: 02/06/2024] Open
Abstract
Background The mode of delivery represents an epigenetic factor with potential to affect further development of the individual by multiple mechanisms. DNA methylation may be one of them, representing a major epigenetic mechanism involving direct chemical modification of the individual's DNA. This pilot study aims to examine whether a specific mode of delivery induces changes of DNA methylation by comparing the umbilical cord blood and peripheral blood of the newborns. Methods Blood samples from infants born by vaginal delivery and caesarean section were analysed to prepare the Methylseq library according to NEBNext enzymatic Methyl-seq Methylation Library Preparation Kit with further generation of target-enriched DNA libraries using the Twist Human Methylome Panel. DNA methylation status was determined using Illumina next-generation sequencing (NGS). Results We identified 168 differentially methylated regions in umbilical cord blood samples and 157 regions in peripheral blood samples. These were associated with 59 common biological, metabolic and signalling pathways for umbilical cord and peripheral blood samples. Conclusion Caesarean section is likely to represent an important epigenetic factor with the potential to induce changes in the genome that could play an important role in development of a broad spectrum of disorders. Our results could contribute to the elucidation of how epigenetic factors, such as a specific mode of delivery, could have adverse impact on health of an individual later in their life.
Collapse
Affiliation(s)
| | - Dominik Kodada
- Medirex Group Academy n.o., Nitra, Slovakia
- Department of Clinical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | | | | | | | - Erik Dosedla
- Department of Gynaecology and Obstetrics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Zuzana Turcsányiová
- Department of Gynaecology and Obstetrics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | | |
Collapse
|
8
|
Yu Y, Chen M, Shen ZG. Molecular biological, physiological, cytological, and epigenetic mechanisms of environmental sex differentiation in teleosts: A systematic review. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 267:115654. [PMID: 37918334 DOI: 10.1016/j.ecoenv.2023.115654] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/04/2023]
Abstract
Human activities have been exerting widespread stress and environmental risks in aquatic ecosystems. Environmental stress, including temperature rise, acidification, hypoxia, light pollution, and crowding, had a considerable negative impact on the life histology of aquatic animals, especially on sex differentiation (SDi) and the resulting sex ratios. Understanding how the sex of fish responds to stressful environments is of great importance for understanding the origin and maintenance of sex, the dynamics of the natural population in the changing world, and the precise application of sex control in aquaculture. This review conducted an exhaustive search of the available literature on the influence of environmental stress (ES) on SDi. Evidence has shown that all types of ES can affect SDi and universally result in an increase in males or masculinization, which has been reported in 100 fish species and 121 cases. Then, this comprehensive review aimed to summarize the molecular biology, physiology, cytology, and epigenetic mechanisms through which ES contributes to male development or masculinization. The relationship between ES and fish SDi from multiple aspects was analyzed, and it was found that environmental sex differentiation (ESDi) is the result of the combined effects of genetic and epigenetic factors, self-physiological regulation, and response to environmental signals, which involves a sophisticated network of various hormones and numerous genes at multiple levels and multiple gradations in bipotential gonads. In both normal male differentiation and ES-induced masculinization, the stress pathway and epigenetic regulation play important roles; however, how they co-regulate SDi is unclear. Evidence suggests that the universal emergence or increase in males in aquatic animals is an adaptation to moderate ES. ES-induced sex reversal should be fully investigated in more fish species and extensively in the wild. The potential aquaculture applications and difficulties associated with ESDi have also been addressed. Finally, the knowledge gaps in the ESDi are presented, which will guide the priorities of future research.
Collapse
Affiliation(s)
- Yue Yu
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Huazhong Agricultural University, Wuhan, PR China
| | - Min Chen
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Huazhong Agricultural University, Wuhan, PR China
| | - Zhi-Gang Shen
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Huazhong Agricultural University, Wuhan, PR China.
| |
Collapse
|
9
|
Vidovič E, Pelikan S, Atanasova M, Kouter K, Pileckyte I, Oblak A, Novak Šarotar B, Videtič Paska A, Bon J. DNA Methylation Patterns in Relation to Acute Severity and Duration of Anxiety and Depression. Curr Issues Mol Biol 2023; 45:7286-7303. [PMID: 37754245 PMCID: PMC10527760 DOI: 10.3390/cimb45090461] [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: 08/04/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Depression and anxiety are common mental disorders that often occur together. Stress is an important risk factor for both disorders, affecting pathophysiological processes through epigenetic changes that mediate gene-environment interactions. In this study, we explored two proposed models about the dynamic nature of DNA methylation in anxiety and depression: a stable change, in which DNA methylation accumulates over time as a function of the duration of clinical symptoms of anxiety and depression, or a flexible change, in which DNA methylation correlates with the acute severity of clinical symptoms. Symptom severity was assessed using clinical questionnaires for anxiety and depression (BDI-II, IDS-C, and HAM-A), and the current episode and the total lifetime symptom duration was obtained from patients' medical records. Peripheral blood DNA methylation levels were determined for the BDNF, COMT, and SLC6A4 genes. We found a significant negative correlation between COMT_1 amplicon methylation and acute symptom scores, with BDI-II (R(22) = 0.190, p = 0.033), IDS-C (R(22) = 0.199, p = 0.029), and HAM-A (R(22) = 0.231, p = 0.018) all showing a similar degree of correlation. Our results suggest that DNA methylation follows flexible dynamics, with methylation levels closely associated with acute clinical presentation rather than with the duration of anxiety and depression. These results provide important insights into the dynamic nature of DNA methylation in anxiety and affective disorders and contribute to our understanding of the complex interplay between stress, epigenetics, and individual phenotype.
Collapse
Affiliation(s)
- Eva Vidovič
- University Psychiatric Clinic Ljubljana, 1260 Ljubljana, Slovenia (J.B.)
| | - Sebastian Pelikan
- University Psychiatric Clinic Ljubljana, 1260 Ljubljana, Slovenia (J.B.)
| | - Marija Atanasova
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Katarina Kouter
- Institute for Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Indre Pileckyte
- Center for Brain and Cognition, Pompeu Fabra University, 08018 Barcelona, Spain
| | - Aleš Oblak
- University Psychiatric Clinic Ljubljana, 1260 Ljubljana, Slovenia (J.B.)
| | - Brigita Novak Šarotar
- University Psychiatric Clinic Ljubljana, 1260 Ljubljana, Slovenia (J.B.)
- Department of Psychiatry, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Alja Videtič Paska
- Institute for Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Jurij Bon
- University Psychiatric Clinic Ljubljana, 1260 Ljubljana, Slovenia (J.B.)
- Department of Psychiatry, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
10
|
Zannas AS. Molecular integrators of stress and aging: the example of FKBP5. Acta Neuropathol 2023; 145:713-715. [PMID: 37020089 DOI: 10.1007/s00401-023-02572-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/02/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023]
Affiliation(s)
- Anthony S Zannas
- Department of Psychiatry, University of North Carolina, 438 Taylor Hall, 109 Mason Farm Rd, Chapel Hill, NC, 27599-7096, USA.
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Stress Initiative, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
11
|
Schreyer L, Reilly J, McConkey H, Kerkhof J, Levy MA, Hu J, Hnaini M, Sadikovic B, Campbell C. The discovery of the DNA methylation episignature for Duchenne muscular dystrophy. Neuromuscul Disord 2023; 33:5-14. [PMID: 36572586 DOI: 10.1016/j.nmd.2022.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/09/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) is an X-linked recessive neuromuscular disorder characterized by progressive muscle weakness due to loss of function mutations in the dystrophin gene. Variation in clinical presentation, the rate of disease progression, and treatment responsiveness have been observed amongst DMD patients, suggesting that factors beyond the loss of dystrophin may contribute to DMD pathophysiology. Epigenetic mechanisms are becoming recognized as important factors implicated in the etiology and progression of various diseases. A growing number of genetic syndromes have been associated with unique genomic DNA methylation patterns (called "episignatures") that can be used for diagnostic testing and as disease biomarkers. To further investigate DMD pathophysiology, we assessed the genome-wide DNA methylation profiles of peripheral blood from 36 patients with DMD using the combination of Illumina Infinium Methylation EPIC bead chip array and EpiSign technology. We identified a unique episignature for DMD that whose specificity was confirmed in relation other neurodevelopmental disorders with known episignatures. By modeling the DMD episignature, we developed a new DMD episignature biomarker and provided novel insights into the molecular pathogenesis of this disorder, which have the potential to advance more effective, personalized approaches to DMD care.
Collapse
Affiliation(s)
- Leighton Schreyer
- Department of Neuroscience, Western University, London, ON N6A 3K7, Canada
| | - Jack Reilly
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada; Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Haley McConkey
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Jennifer Kerkhof
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Michael A Levy
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Jonathan Hu
- Department of Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Mona Hnaini
- Department of Pediatrics, Clinical Neurological Sciences, Western University, London, ON N6A 3K7, Canada
| | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada; Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada.
| | - Craig Campbell
- Department of Pediatrics, Clinical Neurological Sciences and Epidemiology, Western University, London, ON N6A 3K7, Canada.
| |
Collapse
|
12
|
Tronson NC, Schuh KM. Hormonal contraceptives, stress, and the brain: The critical need for animal models. Front Neuroendocrinol 2022; 67:101035. [PMID: 36075276 DOI: 10.1016/j.yfrne.2022.101035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/26/2022] [Accepted: 09/01/2022] [Indexed: 01/10/2023]
Abstract
Hormonal contraceptives are among the most important health and economic developments in the 20thCentury, providing unprecedented reproductive control and a range of health benefits including decreased premenstrual symptoms and protections against various cancers. Hormonal contraceptives modulate neural function and stress responsivity. These changes are usually innocuous or even beneficial, including their effects onmood. However, in approximately 4-10% of users, or up to 30 million people at any given time, hormonal contraceptives trigger depression or anxiety symptoms. How hormonal contraceptives contribute to these responses and who is at risk for adverse outcomes remain unknown. In this paper, we discussstudies of hormonal contraceptive use in humans and describe the ways in which laboratory animal models of contraceptive hormone exposure will be an essential tool for expanding findings to understand the precise mechanisms by which hormonal contraceptives influence the brain, stress responses, and depression risk.
Collapse
Affiliation(s)
- Natalie C Tronson
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA.
| | - Kristen M Schuh
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
13
|
Lee W, Milewski TM, Dwortz MF, Young RL, Gaudet AD, Fonken LK, Champagne FA, Curley JP. Distinct immune and transcriptomic profiles in dominant versus subordinate males in mouse social hierarchies. Brain Behav Immun 2022; 103:130-144. [PMID: 35447300 DOI: 10.1016/j.bbi.2022.04.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 03/31/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022] Open
Abstract
Social status is a critical factor determining health outcomes in human and nonhuman social species. In social hierarchies with reproductive skew, individuals compete to monopolize resources and increase mating opportunities. This can come at a significant energetic cost leading to trade-offs between different physiological systems. In particular, changes in energetic investment in the immune system can have significant short and long-term effects on fitness and health. We have previously found that dominant alpha male mice living in social hierarchies have increased metabolic demands related to territorial defense. In this study, we tested the hypothesis that high-ranking male mice favor adaptive immunity, while subordinate mice show higher investment in innate immunity. We housed 12 groups of 10 outbred CD-1 male mice in a social housing system. All formed linear social hierarchies and subordinate mice had higher concentrations of plasma corticosterone (CORT) than alpha males. This difference was heightened in highly despotic hierarchies. Using flow cytometry, we found that dominant status was associated with a significant shift in immunophenotypes towards favoring adaptive versus innate immunity. Using Tag-Seq to profile hepatic and splenic transcriptomes of alpha and subordinate males, we identified genes that regulate metabolic and immune defense pathways that are associated with status and/or CORT concentration. In the liver, dominant animals showed a relatively higher expression of specific genes involved in major urinary production and catabolic processes, whereas subordinate animals showed relatively higher expression of genes promoting biosynthetic processes, wound healing, and proinflammatory responses. In spleen, subordinate mice showed relatively higher expression of genes facilitating oxidative phosphorylation and DNA repair and CORT was negatively associated with genes involved in lymphocyte proliferation and activation. Together, our findings suggest that dominant and subordinate animals adaptively shift immune profiles and peripheral gene expression to match their contextual needs.
Collapse
Affiliation(s)
- Won Lee
- Department of Psychology, University of Texas at Austin, Austin, TX, USA; Department of In Vivo Pharmacology Services, The Jackson Laboratory, Sacramento, CA, USA
| | - Tyler M Milewski
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Madeleine F Dwortz
- Department of Psychology, University of Texas at Austin, Austin, TX, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Rebecca L Young
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - Andrew D Gaudet
- Department of Psychology, University of Texas at Austin, Austin, TX, USA; Department of Neurology, University of Texas at Austin, Austin, TX, USA
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, USA
| | | | - James P Curley
- Department of Psychology, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
14
|
Armignacco R, Jouinot A, Bouys L, Septier A, Lartigue T, Neou M, Gaspar C, Perlemoine K, Braun L, Riester A, Bonnet-Serrano F, Blanchard A, Amar L, Scaroni C, Ceccato F, Rossi GP, Williams TA, Larsen CK, Allassonnière S, Zennaro MC, Beuschlein F, Reincke M, Bertherat J, Assié G. Identification of glucocorticoid-related molecular signature by whole blood methylome analysis. Eur J Endocrinol 2022; 186:297-308. [PMID: 34914631 PMCID: PMC8789024 DOI: 10.1530/eje-21-0907] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/16/2021] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Cushing's syndrome represents a state of excessive glucocorticoids related to glucocorticoid treatments or to endogenous hypercortisolism. Cushing's syndrome is associated with high morbidity, with significant inter-individual variability. Likewise, adrenal insufficiency is a life-threatening condition of cortisol deprivation. Currently, hormone assays contribute to identify Cushing's syndrome or adrenal insufficiency. However, no biomarker directly quantifies the biological glucocorticoid action. The aim of this study was to identify such markers. DESIGN We evaluated whole blood DNA methylome in 94 samples obtained from patients with different glucocorticoid states (Cushing's syndrome, eucortisolism, adrenal insufficiency). We used an independent cohort of 91 samples for validation. METHODS Leukocyte DNA was obtained from whole blood samples. Methylome was determined using the Illumina methylation chip array (~850 000 CpG sites). Both unsupervised (principal component analysis) and supervised (Limma) methods were used to explore methylome profiles. A Lasso-penalized regression was used to select optimal discriminating features. RESULTS Whole blood methylation profile was able to discriminate samples by their glucocorticoid status: glucocorticoid excess was associated with DNA hypomethylation, recovering within months after Cushing's syndrome correction. In Cushing's syndrome, an enrichment in hypomethylated CpG sites was observed in the region of FKBP5 gene locus. A methylation predictor of glucocorticoid excess was built on a training cohort and validated on two independent cohorts. Potential CpG sites associated with the risk for specific complications, such as glucocorticoid-related hypertension or osteoporosis, were identified, needing now to be confirmed on independent cohorts. CONCLUSIONS Whole blood DNA methylome is dynamically impacted by glucocorticoids. This biomarker could contribute to better assessment of glucocorticoid action beyond hormone assays.
Collapse
Affiliation(s)
- Roberta Armignacco
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
- Correspondence should be addressed to R Armignacco or G Assié; or
| | - Anne Jouinot
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
| | - Lucas Bouys
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
| | - Amandine Septier
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
| | - Thomas Lartigue
- ARAMIS Project-Team, Inria Paris, France
- CMAP, UMR 7641, CNRS, École polytechnique, I.P. Paris, France
| | - Mario Neou
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
| | - Cassandra Gaspar
- Sorbonne Université, Inserm, UMS Pass, Plateforme Post-génomique de la Pitié-Salpêtrière, P3S, Paris, France
| | - Karine Perlemoine
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
| | - Leah Braun
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anna Riester
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fidéline Bonnet-Serrano
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Service d’Hormonologie, Paris, France
| | - Anne Blanchard
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Centre d’Investigations Cliniques 9201, Paris, France
| | - Laurence Amar
- Université de Paris, PARCC, INSERM, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Unité Hypertension Artérielle, Paris, France
| | - Carla Scaroni
- UOC Endocrinologia, Dipartimento di Medicina DIMED, Azienda Ospedaliera-Università di Padova, Padua, Italy
| | - Filippo Ceccato
- UOC Endocrinologia, Dipartimento di Medicina DIMED, Azienda Ospedaliera-Università di Padova, Padua, Italy
| | - Gian Paolo Rossi
- Clinica dell’Ipertensione Arteriosa, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Tracy Ann Williams
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | | | | | - Maria-Christina Zennaro
- Université de Paris, PARCC, INSERM, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, UniversitätsSpital Zürich, Zürich, Switzerland
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jérôme Bertherat
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Service d’Endocrinologie, Center for Rare Adrenal Diseases, Paris, France
| | - Guillaume Assié
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Service d’Endocrinologie, Center for Rare Adrenal Diseases, Paris, France
- Correspondence should be addressed to R Armignacco or G Assié; or
| |
Collapse
|
15
|
Espina JEC, Bagamasbad PD. Synergistic gene regulation by thyroid hormone and glucocorticoid in the hippocampus. VITAMINS AND HORMONES 2021; 118:35-81. [PMID: 35180933 DOI: 10.1016/bs.vh.2021.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The hippocampus is considered the center for learning and memory in the brain, and its development and function is greatly affected by the thyroid and stress axes. Thyroid hormone (TH) and glucocorticoids (GC) are known to have a synergistic effect on developmental programs across several vertebrate species, and their effects on hippocampal structure and function are well-documented. However, there are few studies that focus on the processes and genes that are cooperatively regulated by the two hormone axes. Cross-regulation of the thyroid and stress axes in the hippocampus occurs on multiple levels such that TH can regulate the expression of the GC receptor (GR) while GC can modulate tissue sensitivity to TH by controlling the expression of TH receptor (TR) and enzymes involved in TH biosynthesis. Thyroid hormone and GC are also known to synergistically regulate the transcription of genes associated with neuronal function and development. Synergistic gene regulation by TH and GC may occur through the direct, cooperative action of TR and GR on common target genes, or by indirect mechanisms involving gene regulatory cascades activated by TR and GR. In this chapter, we describe the known physiological effects and underlying molecular mechanisms of TH and GC synergistic gene regulation in the hippocampus.
Collapse
Affiliation(s)
- Jose Ezekiel C Espina
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines
| | - Pia D Bagamasbad
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines.
| |
Collapse
|
16
|
Lee RS, Zandi PP, Santos A, Aulinas A, Carey JL, Webb SM, McCaul ME, Resmini E, Wand GS. Cross-species Association Between Telomere Length and Glucocorticoid Exposure. J Clin Endocrinol Metab 2021; 106:e5124-e5135. [PMID: 34265046 PMCID: PMC8787853 DOI: 10.1210/clinem/dgab519] [Citation(s) in RCA: 4] [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: 03/22/2021] [Indexed: 12/12/2022]
Abstract
CONTEXT Chronic exposure to glucocorticoids (GCs) or stress increases the risk of medical disorders, including cardiovascular and neuropsychiatric disorders. GCs contribute to accelerated aging; however, while the link between chronic GC exposure and disease onset is well established, the underpinning mechanisms are not clear. OBJECTIVE We explored the potential nexus between GCs or stress exposure and telomere length. METHODS In addition to rats exposed to 3 weeks of chronic stress, an iatrogenic mouse model of Cushing syndrome (CS), and a mouse neuronal cell line, we studied 32 patients with CS and age-matched controls and another cohort of 75 healthy humans. RESULTS (1) Exposure to stress in rats was associated with a 54.5% (P = 0.036) reduction in telomere length in T cells. Genomic DNA (gDNA) extracted from the dentate gyrus of stressed and unstressed rats showed 43.2% reduction in telomere length (P = 0.006). (2) Mice exposed to corticosterone had a 61.4% reduction in telomere length in blood gDNA (P = 5.75 × 10-5) and 58.8% reduction in telomere length in the dentate gyrus (P = 0.002). (3) We observed a 40.8% reduction in the telomere length in patients with active CS compared to healthy controls (P = 0.006). There was a 17.8% reduction in telomere length in cured CS patients, which was not different from that of healthy controls (P = 0.08). For both cured and active CS, telomere length correlated significantly with duration of hypercortisolism (R2 = 0.22, P = 0.007). (4) There was a 27.6% reduction in telomere length between low and high tertiles in bedtime cortisol levels of healthy participants (P = 0.019). CONCLUSION Our findings demonstrate that exposure to stress and/or GCs is associated with shortened telomeres, which may be partially reversible.
Collapse
Affiliation(s)
- Richard S Lee
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Peter P Zandi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Mental Health, Johns Hopkins School of Public Health, Baltimore, MD 21205, USA
| | - Alicia Santos
- Endocrinology/Medicine Department, Hospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unit747), IIB-Sant Pau, ISCIII and Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Anna Aulinas
- Endocrinology/Medicine Department, Hospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unit747), IIB-Sant Pau, ISCIII and Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Jenny L Carey
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Susan M Webb
- Endocrinology/Medicine Department, Hospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unit747), IIB-Sant Pau, ISCIII and Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Mary E McCaul
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Eugenia Resmini
- Correspondence: Eugenia Resmini, MD, PhD, Endocrinology/Medicine Department, Hospital Sant Pau, CIBER-ER, Unit747, IIB-Sant Pau, ISCIII, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.
| | - Gary S Wand
- Gary S. Wand, MD, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
17
|
Lin YL, Wei CW, Lerdall TA, Nhieu J, Wei LN. Crabp1 Modulates HPA Axis Homeostasis and Anxiety-like Behaviors by Altering FKBP5 Expression. Int J Mol Sci 2021; 22:12240. [PMID: 34830120 PMCID: PMC8619219 DOI: 10.3390/ijms222212240] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 12/22/2022] Open
Abstract
Retinoic acid (RA), the principal active metabolite of vitamin A, is known to be involved in stress-related disorders. However, its mechanism of action in this regard remains unclear. This study reports that, in mice, endogenous cellular RA binding protein 1 (Crabp1) is highly expressed in the hypothalamus and pituitary glands. Crabp1 knockout (CKO) mice exhibit reduced anxiety-like behaviors accompanied by a lowered stress induced-corticosterone level. Furthermore, CRH/DEX tests show an increased sensitivity (hypersensitivity) of their feedback inhibition in the hypothalamic-pituitary-adrenal (HPA) axis. Gene expression studies show reduced FKBP5 expression in CKO mice; this would decrease the suppression of glucocorticoid receptor (GR) signaling thereby enhancing their feedback inhibition, consistent with their dampened corticosterone level and anxiety-like behaviors upon stress induction. In AtT20, a pituitary gland adenoma cell line elevating or reducing Crabp1 level correspondingly increases or decreases FKBP5 expression, and its endogenous Crabp1 level is elevated by GR agonist dexamethasone or RA treatment. This study shows, for the first time, that Crabp1 regulates feedback inhibition of the the HPA axis by modulating FKBP5 expression. Furthermore, RA and stress can increase Crabp1 level, which would up-regulate FKBP5 thereby de-sensitizing feedback inhibition of HPA axis (by decreasing GR signaling) and increasing the risk of stress-related disorders.
Collapse
Affiliation(s)
| | | | | | | | - Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (Y.-L.L.); (C.-W.W.); (T.A.L.); (J.N.)
| |
Collapse
|
18
|
Zimmer C, Hanson HE, Martin LB. FKBP5 expression is related to HPA flexibility and the capacity to cope with stressors in female and male house sparrows. Horm Behav 2021; 135:105038. [PMID: 34280702 DOI: 10.1016/j.yhbeh.2021.105038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis and its end products, the glucocorticoids, are critical to responding appropriately to stressors. Subsequently, many studies have sought relationships between glucocorticoids and measures of health or fitness, but such relationships are at best highly context dependent. Recently, some endocrinologists have started to suggest that a focus on HPA flexibility, the ability of an individual to mount appropriate responses to different stressors, could be useful. Here, we tested the hypothesis that expression of FKBP5, a cochaperone in the glucocorticoid receptor complex, is a simple and reliable proxy of HPA flexibility in a wild songbird, the house sparrow (Passer domesticus). We quantified HPA flexibility in a novel way, using guidance from research on heart rhythm regulation. As predicted, we found that adult sparrows with low stress-induced FKBP5 expression in the hypothalamus exhibited high HPA flexibility. Moreover, low FKBP5 expression was associated with greater exploratory disposition and were better at maintaining body mass under stressful conditions. Altogether, these results suggest that FKBP5 may be important in the regulation of HPA flexibility, potentially affecting how individuals cope with natural and anthropogenic adversity.
Collapse
Affiliation(s)
- Cedric Zimmer
- Global Health and Infectious Disease Research, University of South Florida, Tampa, FL, USA.
| | - Haley E Hanson
- Global Health and Infectious Disease Research, University of South Florida, Tampa, FL, USA
| | - Lynn B Martin
- Global Health and Infectious Disease Research, University of South Florida, Tampa, FL, USA
| |
Collapse
|
19
|
Naturalistic Stress Hormone Levels Drive Cumulative Epigenomic Changes along the Cellular Lifespan. Int J Mol Sci 2021; 22:ijms22168778. [PMID: 34445485 PMCID: PMC8395735 DOI: 10.3390/ijms22168778] [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: 07/29/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 11/17/2022] Open
Abstract
Environmental stress is ubiquitous in modern societies and can exert a profound and cumulative impact on cell function and health phenotypes. This impact is thought to be in large part mediated by the action of glucocorticoid stress hormones, primarily cortisol in humans. While the underlying molecular mechanisms are unclear, epigenetics-the chemical changes that regulate genomic function without altering the genetic code-has emerged as a key link between environmental exposures and phenotypic outcomes. The present study assessed genome-wide DNA (CpG) methylation, one of the key epigenetic mechanisms, at three timepoints during prolonged (51-day) exposure of cultured human fibroblasts to naturalistic cortisol levels, which can be reached in human tissues during in vivo stress. The findings support a spatiotemporal model of profound and widespread stress hormone-driven methylomic changes that emerge at selected CpG sites, are more likely to spread to nearby located CpGs, and quantitatively accrue at open sea, glucocorticoid receptor binding, and chromatin-accessible sites. Taken together, these findings provide novel insights into how prolonged stress may impact the epigenome, with potentially important implications for stress-related phenotypes.
Collapse
|
20
|
Rivera-Bonet CN, Birn RM, Ladd CO, Meyerand ME, Abercrombie HC. Cortisol effects on brain functional connectivity during emotion processing in women with depression. J Affect Disord 2021; 287:247-254. [PMID: 33799044 PMCID: PMC8128282 DOI: 10.1016/j.jad.2021.03.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND Depression is associated with altered functional connectivity and altered cortisol sensitivity, but the effects of cortisol on functional connectivity in depression are unknown. Previous research shows that brief cortisol augmentation (CORT) has beneficial neurocognitive effects in depression. METHODS We investigated the effects of CORT (20mg oral cortisol) on functional connectivity during emotion processing in women with depression. Participants included 75 women with no depression or a depressive disorder. In a double-blind, crossover study, we used functional magnetic resonance imaging to measure effects of CORT vs. placebo on task-based functional connectivity during presentation of emotionally-laden images. We performed psychophysiological interaction (PPI) to test interactions among depression severity, cortisol administration, and task-dependent functional connectivity using the hippocampus and amygdala as seeds. RESULTS During the presentation of negative images, CORT (vs. placebo) increased functional connectivity between the hippocampus and putamen in association with depression severity. During the presentation of positive pictures CORT increased functional connectivity between the hippocampus and middle frontal gyrus as well as superior temporal gyrus in association with depression. LIMITATIONS Because cortisol was pharmacologically manipulated, results cannot be extrapolated to endogenous increases in cortisol levels. The sample did not permit investigation of differences due to race, ethnicity, or sex. Co-morbidities such as anxiety or PTSD were not accounted for. CONCLUSIONS The results suggest that CORT has normalizing effects on task-dependent functional connectivity in women with depression during emotion processing. Increasing cortisol availability or signaling may have therapeutic benefits within affective disorders.
Collapse
Affiliation(s)
| | - Rasmus M Birn
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States; Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
| | - Charlotte O Ladd
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| | - Mary E Meyerand
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Heather C Abercrombie
- Center for Healthy Minds, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
21
|
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
|
22
|
Zimmer C, Hanson HE, Wildman DE, Uddin M, Martin LB. FKBP5: A Key Mediator of How Vertebrates Flexibly Cope with Adversity. Bioscience 2020. [DOI: 10.1093/biosci/biaa114] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Flexibility in the regulation of the hypothalamic–pituitary–adrenal (HPA) axis is an important mediator of stress resilience as it helps organisms adjust to, avoid, or compensate for acute and chronic challenges across changing environmental contexts. Glucocorticoids remain the favorite metric from medicine to conservation biology to attempt to quantify stress resilience despite the skepticism around their consistency in relation to individual health, welfare, and fitness. We suggest that a cochaperone molecule related to heat shock proteins and involved in glucocorticoid receptor activity, FKBP5, may mediate HPA flexibility and therefore stress resilience because it affects how individuals can regulate glucocorticoids and therefore capacitates their abilities to adjust phenotypes appropriately to prevailing, adverse conditions. Although the molecule is well studied in the biomedical literature, FKBP5 research in wild vertebrates is limited. In the present article, we highlight the potential major role of FKBP5 as mediator of HPA axis flexibility in response to adversity in humans and lab rodents.
Collapse
Affiliation(s)
- Cedric Zimmer
- Global and Planetary Health Department of the College of Public Health, University of South Florida, Tampa, Florida
| | - Haley E Hanson
- Global and Planetary Health Department of the College of Public Health, University of South Florida, Tampa, Florida
| | - Derek E Wildman
- Global and Planetary Health Department of the College of Public Health, University of South Florida, Tampa, Florida
| | - Monica Uddin
- Global and Planetary Health Department of the College of Public Health, University of South Florida, Tampa, Florida
| | - Lynn B Martin
- Global and Planetary Health Department of the College of Public Health, University of South Florida, Tampa, Florida
| |
Collapse
|
23
|
Wood NM, Trebilco T, Cohen-Woods S. Scars of childhood socioeconomic stress: A systematic review. Neurosci Biobehav Rev 2020; 118:397-410. [PMID: 32795493 DOI: 10.1016/j.neubiorev.2020.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 12/17/2022]
Abstract
Childhood socioeconomic position (SEP) is associated with the development of adult psychological outcomes, with DNA methylation (DNAm) as a mechanism to potentially explain these changes. We present the first systematic review synthesising the literature investigating childhood SEP and DNAm. Thirty-two publications were included. Seventeen studies focused on candidate genes, typically focusing on genes implicated with the stress response and/or development of psychiatric conditions. These studies typically investigated different regions of the genes, which revealed inconsistent results. Six studies calculated epigenetic age, with a small number revealing an elevated significant association with childhood SEP. Epigenome-wide studies revealed altered patterns of DNAm which varied between the nine studies. This research area is emerging and demonstrated great variance in findings with no clear patterns identified across studies. Multiple methodological shortcomings are identified, including at the phenotypic level where construct validity of childhood SEP is highly inconsistent, with studies using a wide range of measures. Larger cohorts will be required with international collaborations to strengthen this research area.
Collapse
Affiliation(s)
- Natasha M Wood
- Discipline of Psychology, College of Education, Psychology, and Social Work, Flinders University, Adelaide, SA, Australia
| | - Thomas Trebilco
- Discipline of Psychology, College of Education, Psychology, and Social Work, Flinders University, Adelaide, SA, Australia
| | - Sarah Cohen-Woods
- Discipline of Psychology, College of Education, Psychology, and Social Work, Flinders University, Adelaide, SA, Australia; Órama Institute, Flinders University, Adelaide, SA, Australia; Flinders Centre for Innovation in Cancer, Adelaide, SA, Australia.
| |
Collapse
|
24
|
Siller SJ, Rubenstein DR. A Tissue Comparison of DNA Methylation of the Glucocorticoid Receptor Gene (Nr3c1) in European Starlings. Integr Comp Biol 2019; 59:264-272. [PMID: 31076777 DOI: 10.1093/icb/icz034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Negative feedback of the vertebrate stress response via the hypothalamic-pituitary-adrenal (HPA) axis is regulated by glucocorticoid receptors in the brain. Epigenetic modification of the glucocorticoid receptor gene (Nr3c1), including DNA methylation of the promoter region, can influence expression of these receptors, impacting behavior, physiology, and fitness. However, we still know little about the long-term effects of these modifications on fitness. To better understand these fitness effects, we must first develop a non-lethal method to assess DNA methylation in the brain that allows for multiple measurements throughout an organism's lifetime. In this study, we aimed to determine if blood is a viable biomarker for Nr3c1 DNA methylation in two brain regions (hippocampus and hypothalamus) in adult European starlings (Sturnus vulgaris). We found that DNA methylation of CpG sites in the complete Nr3c1 putative promoter varied among tissue types and was lowest in blood. Although we identified a similar cluster of correlated Nr3c1 putative promoter CpG sites within each tissue, this cluster did not show any correlation in DNA methylation among tissues. Additional studies should consider the role of the developmental environment in producing epigenetic modifications in different tissues.
Collapse
Affiliation(s)
- Stefanie J Siller
- Department of Ecology, Evolution and Environmental Biology, Columbia University, 10th Floor Schermerhorn Extension, 1200 Amsterdam Avenue, New York, NY, USA
| | - Dustin R Rubenstein
- Department of Ecology, Evolution and Environmental Biology, Columbia University, 10th Floor Schermerhorn Extension, 1200 Amsterdam Avenue, New York, NY, USA
| |
Collapse
|
25
|
Géranton SM. Does epigenetic 'memory' of early-life stress predispose to chronic pain in later life? A potential role for the stress regulator FKBP5. Philos Trans R Soc Lond B Biol Sci 2019; 374:20190283. [PMID: 31544613 DOI: 10.1098/rstb.2019.0283] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Animal behaviours are affected not only by inherited genes but also by environmental experiences. For example, in both rats and humans, stressful early-life events such as being reared by an inattentive mother can leave a lasting trace and affect later stress response in adult life. This is owing to a chemical trace left on the chromatin attributed to so-called epigenetic mechanisms. Such an epigenetic trace often has consequences, sometimes long-lasting, on the functioning of our genes, thereby allowing individuals to rapidly adapt to a new environment. One gene under such epigenetic control is FKBP5, the gene that encodes the protein FKPB51, a crucial regulator of the stress axis and a significant driver of chronic pain states. In this article, we will discuss the possibility that exposure to stress could drive the susceptibly to chronic pain via epigenetic modifications of genes within the stress axis such as FKBP5. The possibility that such modifications, and therefore, the susceptibility to chronic pain, could be transmitted across generations in mammals and whether such mechanisms may be evolutionarily conserved across phyla will also be debated. This article is part of the Theo Murphy meeting issue 'Evolution of mechanisms and behaviour important for pain'.
Collapse
Affiliation(s)
- S M Géranton
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
26
|
Epigenetic upregulation of FKBP5 by aging and stress contributes to NF-κB-driven inflammation and cardiovascular risk. Proc Natl Acad Sci U S A 2019; 116:11370-11379. [PMID: 31113877 PMCID: PMC6561294 DOI: 10.1073/pnas.1816847116] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Diseases of the aging are the leading cause of morbidity and mortality. Elucidating the molecular mechanisms through which modifiable factors, such as psychosocial stress, confer risk for aging-related disease can have profound implications. By combining studies in humans with experiments in cells, we show that aging and stress synergize to epigenetically upregulate FKBP5, a protein implicated in stress physiology. Higher FKBP5 promotes inflammation by activating the master immune regulator NF-κB, whereas opposing FKBP5, either genetically or pharmacologically, prevents the effects on NF-κB. Further, the aging/stress-related epigenetic signature of FKBP5 is associated with history of myocardial infarction, a disease linked to inflammation. These findings provide molecular insights into stress-related disease, pointing to biomarker and treatment possibilities. Aging and psychosocial stress are associated with increased inflammation and disease risk, but the underlying molecular mechanisms are unclear. Because both aging and stress are also associated with lasting epigenetic changes, a plausible hypothesis is that stress along the lifespan could confer disease risk through epigenetic effects on molecules involved in inflammatory processes. Here, by combining large-scale analyses in human cohorts with experiments in cells, we report that FKBP5, a protein implicated in stress physiology, contributes to these relations. Across independent human cohorts (total n > 3,000), aging synergized with stress-related phenotypes, measured with childhood trauma and major depression questionnaires, to epigenetically up-regulate FKBP5 expression. These age/stress-related epigenetic effects were recapitulated in a cellular model of replicative senescence, whereby we exposed replicating human fibroblasts to stress (glucocorticoid) hormones. Unbiased genome-wide analyses in human blood linked higher FKBP5 mRNA with a proinflammatory profile and altered NF-κB–related gene networks. Accordingly, experiments in immune cells showed that higher FKBP5 promotes inflammation by strengthening the interactions of NF-κB regulatory kinases, whereas opposing FKBP5 either by genetic deletion (CRISPR/Cas9-mediated) or selective pharmacological inhibition prevented the effects on NF-κB. Further, the age/stress-related epigenetic signature enhanced FKBP5 response to NF-κB through a positive feedback loop and was present in individuals with a history of acute myocardial infarction, a disease state linked to peripheral inflammation. These findings suggest that aging/stress-driven FKBP5–NF-κB signaling mediates inflammation, potentially contributing to cardiovascular risk, and may thus point to novel biomarker and treatment possibilities.
Collapse
|
27
|
Occupational noise exposure is associated with hypertension in China: Results from project ELEFANT. PLoS One 2018; 13:e0209041. [PMID: 30596670 PMCID: PMC6312221 DOI: 10.1371/journal.pone.0209041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/27/2018] [Indexed: 11/20/2022] Open
Abstract
Objectives We investigated the association between occupational noise exposure and the risk of elevated blood pressure and hypertension by stage in young adults. Methods We utilized 124,286 young adults (18–40 years) from the Project ELEFANT study. We categorized occupational noise exposure as high (75 dBA noise exposure for more than 4 hours per day) or low, and measured blood pressure (mmHg) and categorized participants by hypertension stage (normal, elevated, Stage 1, Stage 2). We applied adjusted logistic regression models to identify associations with hypertension risk, and we further examined the noise-BMI, noise-gender, and noise-residence interactions on hypertension risk in separate models. Results High occupational noise exposure was associated with increases in blood pressure among participants with elevated blood pressure (Estimate = 0.23, 95% CI: 1.09, 1.46, p = 0.0009), in Stage 1 hypertension (Estimate = 0.15, 95% CI: 1.06, 1.25, p = 0.0008), and in Stage 2 hypertension (Estimate = 0.41 95% CI: 1.31, 1.73, p<0.0001). Likewise, noise exposure-BMI interaction was consistently positively associated with increases in blood pressure in participants with elevated blood pressure (Estimate = 0.71, 95% CI: 1.55, 2.69, p<0.0001), in Stage 1 hypertension (Estimate = 0.78, 95% CI: 1.82, 2.61, p<0.0001), and in Stage 2 hypertension (Estimate = 2.06, 95% CI: 5.64, 10.81, p<0.0001). The noise exposure-male interaction showed higher risk for hypertension compared to the noise exposure-female interaction in participants with elevated blood pressure (Estimate = 1.24, 95% CI: 2.56, 4.71, p<0.0001), Stage 1 (Estimate = 1.67, 95% CI: 4.34, 6.42, p<0.0001) and Stage 2 hypertension (Estimate = 1.70, 95% CI: 3.86, 7.77, p<0.0001). Finally, we found that noise exposure-urban interaction was consistently associated with an increase in blood pressure in elevated blood pressure (Estimate = 0.32, 95% CI: 1.19, 1.62, p<0.0001) and in Stage 2 hypertension (Estimate = 0.44, 95% CI: 1.31, 1.80, p<0.0001).
Collapse
|
28
|
Duis J, Cox OH, Ji Y, Seifuddin F, Lee RS, Wang X. Effect of Genotype and Maternal Affective Disorder on Intronic Methylation of FK506 Binding Protein 5 in Cord Blood DNA. Front Genet 2018; 9:648. [PMID: 30619472 PMCID: PMC6305129 DOI: 10.3389/fgene.2018.00648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
A single nucleotide polymorphism (SNP: rs1360780) in FKBP5 (FK506 Binding Protein 5) has been shown to interact with exposure to childhood adversity to promote loss of methylation and increase in gene expression in adults. We asked whether rs1360780 can influence FKBP5 intronic methylation in the context of exposure to maternal affective disorders in utero. Sixty cord blood DNA samples from the Boston Birth Cohort were genotyped at rs1360780 and studied for methylation changes as they relate to genotype and exposure to affective disorders during pregnancy. Linear regression was employed to contrast the risk (TT) genotype to the heterozygous (CT) and homozygous (CC) genotypes with adjustment for potential confounders. The recessive genotype (TT) was associated with increased methylation at multiple CpGs in the FKBP5 intron 5 region (p < 0.01). These findings were enhanced among cases exposed to maternal affective disorders (p = 0.02). A human cell line treated with cortisol showed that changes in intron 5 CpG methylation and FKBP5 expression were inversely associated. These findings suggest that rs1360780 can influence FKBP5 intronic methylation by acting in cis as a methylation quantitative locus and highlight the impact of genotypic risk on methylation in utero. Additionally, prenatal stress exposure compounded with the risk genotype may lead to a compensatory increase in methylation.
Collapse
Affiliation(s)
- Jessica Duis
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Olivia H Cox
- Department of Psychiatry and Behavioral Sciences, Mood Disorders Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Yuelong Ji
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Fayaz Seifuddin
- Department of Psychiatry and Behavioral Sciences, Mood Disorders Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Richard S Lee
- Department of Psychiatry and Behavioral Sciences, Mood Disorders Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Xiaobin Wang
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
29
|
Lux V. Epigenetic Programming Effects of Early Life Stress: A Dual-Activation Hypothesis. Curr Genomics 2018; 19:638-652. [PMID: 30532644 PMCID: PMC6225448 DOI: 10.2174/1389202919666180307151358] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/04/2017] [Accepted: 08/04/2017] [Indexed: 12/30/2022] Open
Abstract
Epigenetic processes during early brain development can function as 'developmental switches' that contribute to the stability of long-term effects of early environmental influences by programming central feedback mechanisms of the HPA axis and other neural networks. In this thematic review, we summarize accumulated evidence for a dual-activation of stress-related and sensory networks underlying the epigenetic programming effects of early life stress. We discuss findings indicating epigenetic programming of stress-related genes with impact on HPA axis function, the interaction of epigenetic mechanisms with neural activity in stress-related neural networks, epigenetic effects of glucocorticoid exposure, and the impact of stress on sensory development. Based on these findings, we propose that the combined activation of stress-related neural networks and stressor-specific sensory networks leads to both neural and hormonal priming of the epigenetic machinery, which sensitizes these networks for developmental programming effects. This allows stressor-specific adaptations later in life, but may also lead to functional mal-adaptations, depending on timing and intensity of the stressor. Finally, we discuss methodological and clinical implications of the dual-activation hypothesis. We emphasize that, in addition to modifications in stress-related networks, we need to account for functional modifications in sensory networks and their epigenetic underpinnings to elucidate the long-term effects of early life stress.
Collapse
Affiliation(s)
- Vanessa Lux
- Department of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
30
|
Lee RS, Mahon PB, Zandi PP, McCaul ME, Yang X, Bali U, Wand GS. DNA methylation and sex-specific expression of FKBP5 as correlates of one-month bedtime cortisol levels in healthy individuals. Psychoneuroendocrinology 2018; 97:164-173. [PMID: 30036794 PMCID: PMC6366448 DOI: 10.1016/j.psyneuen.2018.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/17/2018] [Accepted: 07/03/2018] [Indexed: 11/30/2022]
Abstract
Chronic exposure to cortisol is associated with cardiovascular, metabolic, and psychiatric disorders. Although cortisol can be readily measured from peripheral sources such as blood, urine, or saliva, multiple samplings spanning several days to weeks are necessary to reliably assess chronic cortisol exposure levels (referred to as cortisol load). Although cortisol levels in hair have been proposed as a measure of cortisol load, measurement is cumbersome and many people are not candidates due to short hair length and use of hair dyes. To date, there are no blood biomarkers that capture cortisol load. To identify a blood biomarker capable of integrating one-month cortisol exposure levels, 75 healthy participants provided 30+ days of awakening and bedtime saliva cortisol and completed psychosocial measures of anxiety, depression, and stress. Mean daily awakening and bedtime cortisol levels were then compared to CpG methylation levels, gene expression, and genotypes of the stress response gene FKBP5 obtained from blood drawn on the last day of the study. We found a correlation between FKBP5 methylation levels and mean 30+day awakening and bedtime cortisol levels (|r|≥0.32, p ≤ 0.006). We also observed a sex-specific correlation between bedtime cortisol levels and FKBP5 mRNA expression in female participants (r = 0.42, p = 0.005). Dividing the 30-day sampling period into four weekly bins showed that the correlations for both methylation and expression were not being driven by cortisol levels in the week preceding the blood draw. We also identified a female-specific association between FKBP5 mRNA expression and scores on the Beck Anxiety Inventory (r = 0.37, p = 0.013) and Beck Depression Inventory-II (r = 0.32, p = 0.033). Finally, DNA was genotyped at four SNPs, and variation in rs4713902 was shown to have an effect on FKBP5 expression under a codominant model (f = 3.41, p = 0.048) for females only. Our results suggest that blood FKBP5 DNA methylation and mRNA expression levels may be a useful marker for determining general or sex-specific 30-day cortisol load and justifies genome-wide approaches that can potentially identify additional cortisol markers with broader clinical utility.
Collapse
Affiliation(s)
- Richard S Lee
- Department of Psychiatry and Behavioral Sciences and Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, United States
| | - Pamela B Mahon
- Department of Psychiatry and Behavioral Sciences and Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, United States; Department of Psychiatry, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Peter P Zandi
- Department of Psychiatry and Behavioral Sciences and Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, United States; Department of Mental Health, Johns Hopkins School of Public Health, Baltimore, MD, 21205, United States
| | - Mary E McCaul
- Department of Psychiatry and Behavioral Sciences and Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, United States
| | - Xiaoju Yang
- Department of Psychiatry and Behavioral Sciences and Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, United States
| | - Utsav Bali
- Sygnature Discovery, Nottingham, NG1 1GF, UK
| | - Gary S Wand
- Department of Psychiatry and Behavioral Sciences and Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, United States.
| |
Collapse
|
31
|
Carey JL, Cox OH, Seifuddin F, Marque L, Tamashiro KLK, Zandi PP, Wand GS, Lee RS. A Rat Methyl-Seq Platform to Identify Epigenetic Changes Associated with Stress Exposure. J Vis Exp 2018. [PMID: 30417882 PMCID: PMC6235597 DOI: 10.3791/58617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
As genomes of a wider variety of animals become available, there is an increasing need for tools that can capture dynamic epigenetic changes in these animal models. The rat is one particular model animal where an epigenetic tool can complement many pharmacological and behavioral studies to provide insightful mechanistic information. To this end, we adapted the SureSelect Target Capture System (referred to as Methyl-Seq) for the rat, which can assess DNA methylation levels across the rat genome. The rat design targeted promoters, CpG islands, island shores, and GC-rich regions from all RefSeq genes. To implement the platform on a rat experiment, male Sprague Dawley rats were exposed to chronic variable stress for 3 weeks, after which blood samples were collected for genomic DNA extraction. Methyl-Seq libraries were constructed from the rat DNA samples by shearing, adapter ligation, target enrichment, bisulfite conversion, and multiplexing. Libraries were sequenced on a next-generation sequencing platform and the sequenced reads were analyzed to identify DMRs between DNA of stressed and unstressed rats. Top candidate DMRs were independently validated by bisulfite pyrosequencing to confirm the robustness of the platform. Results demonstrate that the rat Methyl-Seq platform is a useful epigenetic tool that can capture methylation changes induced by exposure to stress.
Collapse
Affiliation(s)
- Jenny L Carey
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine
| | - Olivia H Cox
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine
| | - Fayaz Seifuddin
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine
| | - Leonard Marque
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine
| | - Kellie L K Tamashiro
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine
| | - Peter P Zandi
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine; Department of Mental Health, Johns Hopkins School of Public Health
| | - Gary S Wand
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine; Department of Medicine, Johns Hopkins School of Medicine
| | - Richard S Lee
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine;
| |
Collapse
|
32
|
Criado-Marrero M, Rein T, Binder EB, Porter JT, Koren J, Blair LJ. Hsp90 and FKBP51: complex regulators of psychiatric diseases. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0532. [PMID: 29203717 DOI: 10.1098/rstb.2016.0532] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 09/11/2017] [Indexed: 01/30/2023] Open
Abstract
Mood disorders affect nearly a quarter of the world's population. Therefore, understanding the molecular mechanisms underlying these conditions is of great importance. FK-506 binding protein 5 (FKBP5) encodes the FKBP51 protein, a heat shock protein 90 kDa (Hsp90) co-chaperone, and is a risk factor for several affective disorders. FKBP51, in coordination with Hsp90, regulates glucocorticoid receptor (GR) activity via a short negative feedback loop. This signalling pathway rapidly restores homeostasis in the hypothalamic-pituitary-adrenal (HPA) axis following stress. Expression of FKBP5 increases with age through reduced DNA methylation. High levels of FKBP51 are linked to GR resistance and reduced stress coping behaviour. Moreover, common allelic variants in the FKBP5 gene are associated with increased risk of developing affective disorders like anxiety, depression and post-traumatic stress disorder (PTSD). This review highlights the current understanding of the Hsp90 co-chaperone, FKBP5, in disease from both human and animal studies. In addition, FKBP5 genetic implications in the clinic involving life stress exposure, gender differences and treatment outcomes are discussed.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- Marangelie Criado-Marrero
- Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, FL 33613, USA
| | - Theo Rein
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - James T Porter
- Department of Basic Sciences, Ponce Health Sciences University-School of Medicine/Ponce Research Institute, Ponce, Puerto Rico 00732, USA
| | - John Koren
- Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, FL 33613, USA
| | - Laura J Blair
- Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, FL 33613, USA
| |
Collapse
|
33
|
Type 2 diabetes and cardiometabolic risk may be associated with increase in DNA methylation of FKBP5. Clin Epigenetics 2018; 10:82. [PMID: 29951131 PMCID: PMC6010037 DOI: 10.1186/s13148-018-0513-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 06/05/2018] [Indexed: 12/11/2022] Open
Abstract
Background Subclinical hypercortisolism and hypothalamic-pituitary-adrenal (HPA) axis dysfunction are associated with type 2 diabetes (T2DM), cardiovascular disease, and metabolic dysfunction. Intronic methylation of FKBP5 has been implicated as a potential indicator of chronic cortisol exposure. Our overall objective in this study was to determine the association of chronic cortisol exposure, measured via percent methylation of FKBP5 at intron 2, with percent glycosylated hemoglobin (HbA1c), low-density lipoprotein cholesterol (LDL-cholesterol), waist circumference (WC), and body mass index (BMI), in a clinic-based sample of 43 individuals with T2DM. Results Greater percent methylation of the FKBP5 intron 2 at one CpG-dinucleotide region was significantly associated with higher HbA1c (β = 0.535, p = 0.003) and LDL cholesterol (β = 0.344, p = 0.037) and a second CpG-dinucleotide region was significantly associated with higher BMI and WC (β = 0.516, p = 0.001; β = 0.403, p = 0.006, respectively). Conclusions FKBP5 methylation may be a marker of higher metabolic risk in T2DM, possibly secondary to higher exposure to cortisol. Further work should aim to assess the longitudinal association of FKBP5 with cardiovascular disease and glycemic outcomes in T2DM as a first step in understanding potential preventive and treatment-related interventions targeting the HPA axis.
Collapse
|
34
|
Bishop JR, Lee AM, Mills LJ, Thuras PD, Eum S, Clancy D, Erbes CR, Polusny MA, Lamberty GJ, Lim KO. Methylation of FKBP5 and SLC6A4 in Relation to Treatment Response to Mindfulness Based Stress Reduction for Posttraumatic Stress Disorder. Front Psychiatry 2018; 9:418. [PMID: 30279666 PMCID: PMC6153325 DOI: 10.3389/fpsyt.2018.00418] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 08/15/2018] [Indexed: 01/18/2023] Open
Abstract
Mindfulness Based Stress Reduction (MBSR) is an effective non-pharmacologic treatment for veterans with PTSD. Extensive work has identified epigenetic factors related to PTSD disease risk and pathophysiology, but how these factors influence treatment response is unclear. Serotonin signaling and hypothalamic-pituitary-adrenal (HPA) axis functioning may be perturbed in PTSD and are molecular pathways targeted by PTSD treatments. To identify potential biomarkers for treatment response, we utilized genomic DNA isolated from peripheral blood samples from veterans with PTSD who were responders (n = 11) or non-responders (n = 11) to MBSR as part of a clinical trial. We assessed methylation levels at CpG sites in regions of the serotonin transporter (SLC6A4) previously associated with expression and depression outcomes, as well as the Intron 7 region of the FK506 binding protein 5 (FKBP5) containing known glucocorticoid response elements suggested to regulate this gene. Selected subjects were matched across MBSR responder status by baseline symptoms, age, sex, current smoking status, and current antidepressant use. Percent methylation was compared between responders and non-responders at baseline (pre-MBSR treatment). Additionally, percent change in methylation from baseline to post-treatment was compared between responders and non-responders. There was a significant time x responder group interaction for methylation in FKBP5 intron 7 bin 2 [F (1, 19) = 7.492, p = 0.013] whereby responders had a decrease in methylation and non-responders had an increase in methylation from before to after treatment in this region. Analyses of the three CpG sites within bin 2 revealed a significant time x responder group interaction for CpG_35558513 [F (1, 19) = 5.551, p = 0.029] which resides in a known glucocorticoid response element (GRE). Decreases in FKBP5 methylation after treatment in responders as compared to increases in non-responders suggest that effective meditation intervention may be associated with stress-related pathways at the molecular level. These preliminary findings suggest that DNA methylation signatures within FKBP5 are potential indicators of response to meditation treatment in PTSD and require validation in larger cohorts.
Collapse
Affiliation(s)
- Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, United States.,Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Adam M Lee
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, United States
| | - Lauren J Mills
- University of Minnesota Supercomputing Institute, Minneapolis, MN, United States
| | - Paul D Thuras
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, United States.,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, United States
| | - Seenae Eum
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, United States
| | - Doris Clancy
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Christopher R Erbes
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, United States.,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, United States
| | - Melissa A Polusny
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, United States.,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, United States
| | - Gregory J Lamberty
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, United States.,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, United States
| | - Kelvin O Lim
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, United States.,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, United States.,Defense Veterans Brain Injury Center, Minneapolis, MN, United States
| |
Collapse
|
35
|
Mehta D, Bruenig D, Carrillo-Roa T, Lawford B, Harvey W, Morris CP, Smith AK, Binder EB, Young RM, Voisey J. Genomewide DNA methylation analysis in combat veterans reveals a novel locus for PTSD. Acta Psychiatr Scand 2017; 136:493-505. [PMID: 28795405 DOI: 10.1111/acps.12778] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Epigenetic modifications such as DNA methylation may play a key role in the aetiology and serve as biomarkers for post-traumatic stress disorder (PTSD). We performed a genomewide analysis to identify genes whose DNA methylation levels are associated with PTSD. METHOD A total of 211 individuals comprising Australian male Vietnam War veterans (n = 96) and males from a general population belonging to the Grady Trauma Project (n = 115) were included. Genomewide DNA methylation was performed from peripheral blood using the Illumina arrays. Data analysis was performed using generalized linear regression models. RESULTS Differential DNA methylation of 17 previously reported PTSD candidate genes was associated with PTSD symptom severity. Genomewide analyses revealed CpG sites spanning BRSK1, LCN8, NFG and DOCK2 genes were associated with PTSD symptom severity. We replicated the findings of DOCK2 in an independent cohort. Pathway analysis revealed that among the associated genes, genes within actin cytoskeleton and focal adhesion molecular pathways were enriched. CONCLUSION These data highlight the role of DNA methylation as biomarkers of PTSD. The results support the role of previous candidates and uncover novel genes associated with PTSD, such as DOCK2. This study contributes to our understanding of the biological underpinnings of PTSD.
Collapse
Affiliation(s)
- D Mehta
- School of Psychology and Counselling, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - D Bruenig
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Greenslopes, QLD, Australia
| | - T Carrillo-Roa
- Department of Translational Research, Max Planck Institute of Psychiatry, Munich, Germany
| | - B Lawford
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - W Harvey
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Greenslopes, QLD, Australia
| | - C P Morris
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - A K Smith
- Genetics and Molecular Biology Program, Emory University, Atlanta, GA, USA.,Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - E B Binder
- Department of Translational Research, Max Planck Institute of Psychiatry, Munich, Germany.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - R McD Young
- Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - J Voisey
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| |
Collapse
|
36
|
Gray JD, Kogan JF, Marrocco J, McEwen BS. Genomic and epigenomic mechanisms of glucocorticoids in the brain. Nat Rev Endocrinol 2017; 13:661-673. [PMID: 28862266 DOI: 10.1038/nrendo.2017.97] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Following the discovery of glucocorticoid receptors in the hippocampus and other brain regions, research has focused on understanding the effects of glucocorticoids in the brain and their role in regulating emotion and cognition. Glucocorticoids are essential for adaptation to stressors (allostasis) and in maladaptation resulting from allostatic load and overload. Allostatic overload, which can occur during chronic stress, can reshape the hypothalamic-pituitary-adrenal axis through epigenetic modification of genes in the hippocampus, hypothalamus and other stress-responsive brain regions. Glucocorticoids exert their effects on the brain through genomic mechanisms that involve both glucocorticoid receptors and mineralocorticoid receptors directly binding to DNA, as well as by non-genomic mechanisms. Furthermore, glucocorticoids synergize both genomically and non-genomically with neurotransmitters, neurotrophic factors, sex hormones and other stress mediators to shape an organism's present and future responses to a stressful environment. Here, we discuss the mechanisms of glucocorticoid action in the brain and review how glucocorticoids interact with stress mediators in the context of allostasis, allostatic load and stress-induced neuroplasticity.
Collapse
Affiliation(s)
- Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065. USA
| | - Joshua F Kogan
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065. USA
| | - Jordan Marrocco
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065. USA
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065. USA
| |
Collapse
|
37
|
Winkler BK, Lehnert H, Oster H, Kirchner H, Harbeck B. FKBP5 methylation as a possible marker for cortisol state and transient cortisol exposure in healthy human subjects. Epigenomics 2017; 9:1279-1286. [PMID: 28875708 DOI: 10.2217/epi-2017-0057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Current glucocorticoid replacement regimens, in adrenal insufficiency, fail to mimic the physiological cortisol secretion, thereby fostering serious side effects. AIM To experimentally evaluate the impact of CpG methylation within the FKBP5 gene as a possible short- and long-term marker for cortisol exposure in humans. MATERIALS & METHODS An ACTH-stimulation test was carried out and methylation status of the FKBP5 gene in leukocytes was determined. RESULTS A negative correlation between basal levels of methylation and serum cortisol was observed. Individual changes in FKBP5 methylation after 24 h correlated with cortisol responses. CONCLUSION Considering previous studies conducted with murine leucocytes, FKBP5 methylation may be suitable as a long-term biomarker, rather than acute glucocorticoid exposure, also in humans.
Collapse
Affiliation(s)
- Britta K Winkler
- Department of Internal Medicine I, University of Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
| | - Hendrik Lehnert
- Department of Internal Medicine I, University of Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
| | - Henrik Oster
- Department of Internal Medicine I, University of Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
| | - Henriette Kirchner
- Department of Internal Medicine I, University of Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
| | - Birgit Harbeck
- Department of Internal Medicine I, University of Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
| |
Collapse
|
38
|
Smith JA, Zhao W, Wang X, Ratliff SM, Mukherjee B, Kardia SLR, Liu Y, Roux AVD, Needham BL. Neighborhood characteristics influence DNA methylation of genes involved in stress response and inflammation: The Multi-Ethnic Study of Atherosclerosis. Epigenetics 2017; 12:662-673. [PMID: 28678593 PMCID: PMC5687339 DOI: 10.1080/15592294.2017.1341026] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Living in a disadvantaged neighborhood is associated with poor health outcomes even after accounting for individual-level socioeconomic factors. The chronic stress of unfavorable neighborhood conditions may lead to dysregulation of the stress reactivity and inflammatory pathways, potentially mediated through epigenetic mechanisms such as DNA methylation. We used multi-level models to examine the relationship between 2 neighborhood conditions and methylation levels of 18 genes related to stress reactivity and inflammation in purified monocytes from 1,226 participants of the Multi-Ethnic Study of Atherosclerosis (MESA), a population-based sample of US adults. Neighborhood socioeconomic disadvantage, a summary of 16 census-based metrics, was associated with DNA methylation [False discovery rate (FDR) q-value ≤ 0.1] in 2 out of 7 stress-related genes evaluated (CRF, SLC6A4) and 2 out of 11 inflammation-related genes (F8, TLR1). Neighborhood social environment, a summary measure of aesthetic quality, safety, and social cohesion, was associated with methylation in 4 of the 7 stress-related genes (AVP, BDNF, FKBP5, SLC6A4) and 7 of the 11 inflammation-related genes (CCL1, CD1D, F8, KLRG1, NLRP12, SLAMF7, TLR1). High socioeconomic disadvantage and worse social environment were primarily associated with increased methylation. In 5 genes with significant associations between neighborhood and methylation (FKBP5, CD1D, F8, KLRG1, NLRP12), methylation was associated with gene expression of at least one transcript. These results demonstrate that multiple dimensions of neighborhood context may influence methylation levels and subsequent gene expression of stress- and inflammation-related genes, even after accounting for individual socioeconomic factors. Further elucidating the molecular mechanisms underlying these relationships will be important for understanding the etiology of health disparities.
Collapse
Affiliation(s)
- Jennifer A Smith
- a Department of Epidemiology , School of Public Health, University of Michigan , Ann Arbor , MI , USA.,b Survey Research Center, Institute for Social Research, University of Michigan , Ann Arbor , MI , USA
| | - Wei Zhao
- a Department of Epidemiology , School of Public Health, University of Michigan , Ann Arbor , MI , USA
| | - Xu Wang
- c Department of Epidemiology and Biostatistics , Dornsife School of Public Health, Drexel University , Philadelphia , PA , USA
| | - Scott M Ratliff
- a Department of Epidemiology , School of Public Health, University of Michigan , Ann Arbor , MI , USA
| | - Bhramar Mukherjee
- d Department of Biostatistics , School of Public Health, University of Michigan , Ann Arbor , MI , USA
| | - Sharon L R Kardia
- a Department of Epidemiology , School of Public Health, University of Michigan , Ann Arbor , MI , USA
| | - Yongmei Liu
- e Department of Epidemiology and Prevention , School of Medicine, Wake Forest University , Winston-Salem , NC , USA
| | - Ava V Diez Roux
- c Department of Epidemiology and Biostatistics , Dornsife School of Public Health, Drexel University , Philadelphia , PA , USA
| | - Belinda L Needham
- a Department of Epidemiology , School of Public Health, University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
39
|
Seifuddin F, Wand G, Cox O, Pirooznia M, Moody L, Yang X, Tai J, Boersma G, Tamashiro K, Zandi P, Lee R. Genome-wide Methyl-Seq analysis of blood-brain targets of glucocorticoid exposure. Epigenetics 2017; 12:637-652. [PMID: 28557603 DOI: 10.1080/15592294.2017.1334025] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Chronic exposure to glucocorticoids (GCs) can lead to psychiatric complications through epigenetic mechanisms such as DNA methylation (DNAm). We sought to determine whether epigenetic changes in a peripheral tissue can serve as a surrogate for those in a relatively inaccessible tissue such as the brain. DNA extracted from the hippocampus and blood of mice treated with GCs or vehicle solution was assayed using a genome-wide DNAm platform (Methyl-Seq) to identify differentially methylated regions (DMRs) induced by GC treatment. We observed that ∼70% of the DMRs in both tissues lost methylation following GC treatment. Of the 3,095 DMRs that mapped to the same genes in both tissues, 1,853 DMRs underwent DNAm changes in the same direction. Interestingly, only 209 DMRs (<7%) overlapped in genomic coordinates between the 2 tissues, suggesting tissue-specific differences in GC-targeted loci. Pathway analysis showed that the DMR-associated genes were members of pathways involved in metabolism, immune function, and neurodevelopment. Also, changes in cell type composition of blood and brain were examined by fluorescence-activated cell sorting. Separation of the cortex into neuronal and non-neuronal fractions and the leukocytes into T-cells, B-cells, and neutrophils showed that GC-induced methylation changes primarily occurred in neurons and T-cells, with the blood tissue also undergoing a shift in the proportion of constituent cell types while the proportion of neurons and glia in the brain remained stable. From the current pilot study, we found that despite tissue-specific epigenetic changes and cellular heterogeneity, blood can serve as a surrogate for GC-induced changes in the brain.
Collapse
Affiliation(s)
- Fayaz Seifuddin
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Gary Wand
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA.,b Department of Medicine, Division of Endocrinology , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Olivia Cox
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Mehdi Pirooznia
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Laura Moody
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Xiaoju Yang
- b Department of Medicine, Division of Endocrinology , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Jonathan Tai
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Gretha Boersma
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Kellie Tamashiro
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Peter Zandi
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| | - Richard Lee
- a Mood Disorders Center, Department of Psychiatry and Behavioral Sciences , Johns Hopkins University School of Medicine , Baltimore , MD, USA
| |
Collapse
|
40
|
Epigenetic programming by stress and glucocorticoids along the human lifespan. Mol Psychiatry 2017; 22:640-646. [PMID: 28289275 DOI: 10.1038/mp.2017.35] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 01/08/2017] [Accepted: 01/17/2017] [Indexed: 12/19/2022]
Abstract
Psychosocial stress triggers a set of behavioral, neural, hormonal, and molecular responses that can be a driving force for survival when adaptive and time-limited, but may also contribute to a host of disease states if dysregulated or chronic. The beneficial or detrimental effects of stress are largely mediated by the hypothalamic-pituitary axis, a highly conserved neurohormonal cascade that culminates in systemic secretion of glucocorticoids. Glucocorticoids activate the glucocorticoid receptor, a ubiquitous nuclear receptor that not only causes widespread changes in transcriptional programs, but also induces lasting epigenetic modifications in many target tissues. While the epigenome remains sensitive to stressors throughout life, we propose two key principles that may govern the epigenetics of stress and glucocorticoids along the lifespan: first, the presence of distinct life periods, during which the epigenome shows heightened plasticity to stress exposure, such as in early development and at advanced age; and, second, the potential of stress-induced epigenetic changes to accumulate throughout life both in select chromatin regions and at the genome-wide level. These principles have important clinical and translational implications, and they show striking parallels with the existence of sensitive developmental periods and the cumulative impact of stressful experiences on the development of stress-related phenotypes. We hope that this conceptual mechanistic framework will stimulate fruitful research that aims at unraveling the molecular pathways through which our life stories sculpt genomic function to contribute to complex behavioral and somatic phenotypes.
Collapse
|
41
|
Illam SP, Narayanankutty A, Raghavamenon AC. Polyphenols of virgin coconut oil prevent pro-oxidant mediated cell death. Toxicol Mech Methods 2017; 27:442-450. [DOI: 10.1080/15376516.2017.1320458] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
42
|
Glad CAM, Andersson-Assarsson JC, Berglund P, Bergthorsdottir R, Ragnarsson O, Johannsson G. Reduced DNA methylation and psychopathology following endogenous hypercortisolism - a genome-wide study. Sci Rep 2017; 7:44445. [PMID: 28300138 PMCID: PMC5353706 DOI: 10.1038/srep44445] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/08/2017] [Indexed: 01/21/2023] Open
Abstract
Patients with Cushing’s Syndrome (CS) in remission were used as a model to test the hypothesis that long-standing excessive cortisol exposure induces changes in DNA methylation that are associated with persisting neuropsychological consequences. Genome-wide DNA methylation was assessed in 48 women with CS in long-term remission (cases) and 16 controls matched for age, gender and education. The Fatigue impact scale and the comprehensive psychopathological rating scale were used to evaluate fatigue, depression and anxiety. Cases had lower average global DNA methylation than controls (81.2% vs 82.7%; p = 0.002). Four hundred and sixty-one differentially methylated regions, containing 3,246 probes mapping to 337 genes were identified. After adjustment for age and smoking, 731 probes in 236 genes were associated with psychopathology (fatigue, depression and/or anxiety). Twenty-four gene ontology terms were associated with psychopathology; terms related to retinoic acid receptor signalling were the most common (adjusted p = 0.0007). One gene in particular, COL11A2, was associated with fatigue following a false discovery rate correction. Our findings indicate that hypomethylation of FKBP5 and retinoic acid receptor related genes serve a potential mechanistic explanation for long-lasting GC-induced psychopathology.
Collapse
Affiliation(s)
- Camilla A M Glad
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg and Department of Endocrinology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johanna C Andersson-Assarsson
- Department of Molecular and Clinical Medicine, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Berglund
- Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ragnhildur Bergthorsdottir
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg and Department of Endocrinology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Oskar Ragnarsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg and Department of Endocrinology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gudmundur Johannsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg and Department of Endocrinology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
43
|
Needham BL, Smith JA, Zhao W, Wang X, Mukherjee B, Kardia SLR, Shively CA, Seeman TE, Liu Y, Diez Roux AV. Life course socioeconomic status and DNA methylation in genes related to stress reactivity and inflammation: The multi-ethnic study of atherosclerosis. Epigenetics 2016; 10:958-69. [PMID: 26295359 DOI: 10.1080/15592294.2015.1085139] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Epigenetic changes, such as DNA methylation, have been hypothesized to provide a link between the social environment and disease development. The purpose of this study was to examine associations between life course measures of socioeconomic status (SES) and DNA methylation (DNAm) in 18 genes related to stress reactivity and inflammation using a multi-level modeling approach that treats DNAm measurements as repeat measures within an individual. DNAm and gene expression were assessed in purified monocytes for a random subsample of 1,264 non-Hispanic white, African-American, and Hispanic participants aged 55-94 from the Multi-Ethnic Study of Atherosclerosis (MESA). After correction for multiple testing, we found that low childhood SES was associated with DNAm in 3 stress-related genes (AVP, FKBP5, OXTR) and 2 inflammation-related genes (CCL1, CD1D), low adult SES was associated with DNAm in one stress-related gene (AVP) and 5 inflammation-related genes (CD1D, F8, KLRG1, NLRP12, TLR3), and social mobility was associated with DNAm in 3 stress-related genes (AVP, FKBP5, OXTR) and 7 inflammation-related genes (CCL1, CD1D, F8, KLRG1, NLRP12, PYDC1, TLR3). In general, low SES was associated with increased DNAm. Expression data was available for 7 genes that showed a significant relationship between SES and DNAm. In 5 of these 7 genes (CD1D, F8, FKBP5, KLRG1, NLRP12), DNAm was associated with gene expression for at least one transcript, providing evidence of the potential functional consequences of alterations in DNAm related to SES. The results of this study reflect the biological complexity of epigenetic data and underscore the need for multi-disciplinary approaches to study how DNAm may contribute to the social patterning of disease.
Collapse
Affiliation(s)
- Belinda L Needham
- a Department of Epidemiology; University of Michigan; Ann Arbor, MI USA
| | - Jennifer A Smith
- a Department of Epidemiology; University of Michigan; Ann Arbor, MI USA
| | - Wei Zhao
- a Department of Epidemiology; University of Michigan; Ann Arbor, MI USA
| | - Xu Wang
- b Department of Epidemiology; Drexel University; Philadelphia, PA USA
| | - Bhramar Mukherjee
- c Department of Biostatistics; University of Michigan; Ann Arbor, MI USA
| | - Sharon L R Kardia
- a Department of Epidemiology; University of Michigan; Ann Arbor, MI USA
| | - Carol A Shively
- d Section on Comparative Medicine; Wake Forest University; Winston-Salem, NC USA
| | - Teresa E Seeman
- e Department of Medicine; University of California ; Los Angeles, Los Angeles, CA USA
| | - Yongmei Liu
- f Department of Epidemiology and Prevention; Wake Forest University; Winston-Salem, NC USA
| | - Ava V Diez Roux
- b Department of Epidemiology; Drexel University; Philadelphia, PA USA
| |
Collapse
|
44
|
Gassen NC, Chrousos GP, Binder EB, Zannas AS. Life stress, glucocorticoid signaling, and the aging epigenome: Implications for aging-related diseases. Neurosci Biobehav Rev 2016; 74:356-365. [PMID: 27343999 DOI: 10.1016/j.neubiorev.2016.06.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 05/24/2016] [Accepted: 06/06/2016] [Indexed: 02/06/2023]
Abstract
Life stress has been associated with accelerated cellular aging and increased risk for developing aging-related diseases; however, the underlying molecular mechanisms remain elusive. A highly relevant process that may underlie this association is epigenetic regulation. In this review, we build upon existing evidence to propose a model whereby exposure to life stress, in part via its effects on the hypothalamic-pituitary axis and the glucocorticoid signaling system, may alter the epigenetic landscape across the lifespan and, consequently, influence genomic regulation and function in ways that are conducive to the development of aging-related diseases. This model is supported by recent studies showing that life stressors and stress-related phenotypes can accelerate epigenetic aging, a measure that is based on DNA methylation prediction of chronological age and has been associated with several aging-related disease phenotypes. We discuss the implications of this model for the prevention and treatment of aging-related diseases, as well as the challenges and limitations of this line of research.
Collapse
Affiliation(s)
- Nils C Gassen
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - George P Chrousos
- First Department of Pediatrics, University of Athens Medical School, Athens, Greece
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany; Department of Psychiatry and Behavioral Sciences, Emory University Medical School, Atlanta, GA, USA
| | - Anthony S Zannas
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
45
|
Abstract
A growing body of evidence suggests that alterations of the stress response system may be a mechanism by which childhood maltreatment alters risk for psychopathology. FK506 binding protein 51 (FKBP5) binds to the glucocorticoid receptor and alters its ability to respond to stress signaling. The aim of the present study was to examine methylation of the FKBP5 gene (FKBP5), and the role of an FKBP5 genetic variant, in relation to childhood maltreatment in a sample of impoverished preschool-aged children. One hundred seventy-four families participated in this study, including 69 with child welfare documentation of moderate to severe maltreatment in the past 6 months. The children, who ranged in age from 3 to 5 years, were racially and ethnically diverse. Structured record review and interviews in the home were used to assess a history of maltreatment, other traumas, and contextual life stressors; and a composite variable assessed the number exposures to these adversities. Methylation of two sites in intron 7 of FKBP5 was measured via sodium bisulfite pyrosequencing. Maltreated children had significantly lower levels of methylation at both CpG sites (p < .05). Lifetime contextual stress exposure showed a trend for lower levels of methylation at one of the sites, and a trend for an interaction with the FKBP5 polymorphism. A composite adversity variable was associated with lower levels of methylation at one of the sites as well (p < .05). FKBP5 alters glucocorticoid receptor responsiveness, and FKBP5 gene methylation may be a mechanism of the biobehavioral effects of adverse exposures in young children.
Collapse
|
46
|
Sawamura T, Klengel T, Armario A, Jovanovic T, Norrholm SD, Ressler KJ, Andero R. Dexamethasone Treatment Leads to Enhanced Fear Extinction and Dynamic Fkbp5 Regulation in Amygdala. Neuropsychopharmacology 2016; 41:832-46. [PMID: 26174596 PMCID: PMC4707829 DOI: 10.1038/npp.2015.210] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 06/09/2015] [Accepted: 06/28/2015] [Indexed: 12/19/2022]
Abstract
Posttraumatic stress disorder (PTSD) is both a prevalent and debilitating trauma-related disorder associated with dysregulated fear learning at the core of many of its signs and symptoms. Improvements in the currently available psychological and pharmacological treatments are needed in order to improve PTSD treatment outcomes and to prevent symptom relapse. In the present study, we used a putative animal model of PTSD that included presentation of immobilization stress (IMO) followed by fear conditioning (FC) a week later. We then investigated the acute effects of GR receptor activation on the extinction (EXT) of conditioned freezing, using dexamethasone administered systemically which is known to result in suppression of the HPA axis. In our previous work, IMO followed by tone-shock-mediated FC was associated with impaired fear EXT. In this study, we administered dexamethasone 4 h before EXT training and then examined EXT retention (RET) 24 h later to determine whether dexamethasone suppression rescued EXT deficits. Dexamethasone treatment produced dose-dependent enhancement of both EXT and RET. Dexamethasone was also associated with reduced amygdala Fkbp5 mRNA expression following EXT and after RET. Moreover, DNA methylation of the Fkbp5 gene occurred in a dose-dependent and time course-dependent manner within the amygdala. Additionally, we found dynamic changes in epigenetic regulation, including Dnmt and Tet gene pathways, as a function of both fear EXT and dexamethasone suppression of the HPA axis. Together, these data suggest that dexamethasone may serve to enhance EXT by altering Fkbp5-mediated glucocorticoid sensitivity via epigenetic regulation of Fkbp5 expression.
Collapse
Affiliation(s)
- Takehito Sawamura
- Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, GA, USA,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA,Self Defense Forces Yokosuka Hospital, Yokosuka City, Japan
| | - Torsten Klengel
- Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, GA, USA,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Antonio Armario
- Institut de Neurociènces, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Tanja Jovanovic
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Seth D Norrholm
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA,Atlanta VA Medical Center, Decatur, GA, USA
| | - Kerry J Ressler
- Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, GA, USA,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA,Howard Hughes Medical Institute, Emory University, Atlanta, GA, USA
| | - Raül Andero
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA,Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA, Tel: +617 855 4216, Fax: 617 855 3479, E-mail:
| |
Collapse
|
47
|
Gene-Stress-Epigenetic Regulation of FKBP5: Clinical and Translational Implications. Neuropsychopharmacology 2016; 41:261-74. [PMID: 26250598 PMCID: PMC4677131 DOI: 10.1038/npp.2015.235] [Citation(s) in RCA: 391] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 12/13/2022]
Abstract
Stress responses and related outcomes vary markedly across individuals. Elucidating the molecular underpinnings of this variability is of great relevance for developing individualized prevention strategies and treatments for stress-related disorders. An important modulator of stress responses is the FK506-binding protein 51 (FKBP5/FKBP51). FKBP5 acts as a co-chaperone that modulates not only glucocorticoid receptor activity in response to stressors but also a multitude of other cellular processes in both the brain and periphery. Notably, the FKBP5 gene is regulated via complex interactions among environmental stressors, FKBP5 genetic variants, and epigenetic modifications of glucocorticoid-responsive genomic sites. These interactions can result in FKBP5 disinhibition that has been shown to contribute to a number of aberrant phenotypes in both rodents and humans. Consequently, FKBP5 blockade may hold promise as treatment intervention for stress-related disorders, and recently developed selective FKBP5 blockers show encouraging results in vitro and in rodent models. Although risk for stress-related disorders is conferred by multiple environmental and genetic factors, the findings related to FKBP5 illustrate how a deeper understanding of the molecular and systemic mechanisms underlying specific gene-environment interactions may provide insights into the pathogenesis of stress-related disorders.
Collapse
|
48
|
Nutritional Omega-3 Deficiency Alters Glucocorticoid Receptor-Signaling Pathway and Neuronal Morphology in Regionally Distinct Brain Structures Associated with Emotional Deficits. Neural Plast 2015; 2016:8574830. [PMID: 27057368 PMCID: PMC4710953 DOI: 10.1155/2016/8574830] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/07/2015] [Accepted: 10/19/2015] [Indexed: 12/31/2022] Open
Abstract
Extensive evidence suggests that long term dietary n-3 polyunsaturated fatty acids (PUFAs) deficiency results in altered emotional behaviour. We have recently demonstrated that n-3 PUFAs deficiency induces emotional alterations through abnormal corticosterone secretion which leads to altered dendritic arborisation in the prefrontal cortex (PFC). Here we show that hypothalamic-pituitary-adrenal (HPA) axis feedback inhibition was not compromised in n-3 deficient mice. Rather, glucocorticoid receptor (GR) signaling pathway was inactivated in the PFC but not in the hippocampus of n-3 deficient mice. Consequently, only dendritic arborisation in PFC was affected by dietary n-3 PUFAs deficiency. In addition, occlusion experiment with GR blockade altered GR signaling in the PFC of control mice, with no further alterations in n-3 deficient mice. In conclusion, n-3 PUFAs deficiency compromised PFC, leading to dendritic atrophy, but did not change hippocampal GR function and dendritic arborisation. We argue that this GR sensitivity contributes to n-3 PUFAs deficiency-related emotional behaviour deficits.
Collapse
|
49
|
Zannas AS, Provençal N, Binder EB. Epigenetics of Posttraumatic Stress Disorder: Current Evidence, Challenges, and Future Directions. Biol Psychiatry 2015; 78:327-35. [PMID: 25979620 DOI: 10.1016/j.biopsych.2015.04.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/20/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a stress-related psychiatric disorder that is thought to emerge from complex interactions among traumatic events and multiple genetic factors. Epigenetic regulation lies at the heart of these interactions and mediates the lasting effects of the environment on gene regulation. An increasing body of evidence in human subjects with PTSD supports a role for epigenetic regulation of distinct genes and pathways in the pathogenesis of PTSD. The role of epigenetic regulation is further supported by studies examining fear conditioning in rodent models. Although this line of research offers an exciting outlook for future epigenetic research in PTSD, important limitations include the tissue specificity of epigenetic modifications, the phenomenologic definition of the disorder, and the challenge of translating molecular evidence across species. These limitations call for studies that combine data from postmortem human brain tissue and animal models, assess longitudinal epigenetic changes in living subjects, and examine dimensional phenotypes in addition to diagnoses. Moreover, examining the environmental, genetic, and epigenetic factors that promote resilience to trauma may lead to important advances in the field.
Collapse
Affiliation(s)
- Anthony S Zannas
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - Nadine Provençal
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany; Department of Psychiatry and Behavioral Sciences, Emory University Medical School, Atlanta, Georgia.
| |
Collapse
|
50
|
Klengel T, Binder EB. Epigenetics of Stress-Related Psychiatric Disorders and Gene × Environment Interactions. Neuron 2015; 86:1343-57. [PMID: 26087162 DOI: 10.1016/j.neuron.2015.05.036] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A deeper understanding of the pathomechanisms leading to stress-related psychiatric disorders is important for the development of more efficient preventive and therapeutic strategies. Epidemiological studies indicate a combined contribution of genetic and environmental factors in the risk for disease. The environment, particularly early life severe stress or trauma, can lead to lifelong molecular changes in the form of epigenetic modifications that can set the organism off on trajectories to health or disease. Epigenetic modifications are capable of shaping and storing the molecular response of a cell to its environment as a function of genetic predisposition. This provides a potential mechanism for gene-environment interactions. Here, we review epigenetic mechanisms associated with the response to stress and trauma exposure and the development of stress-related psychiatric disorders. We also look at how they may contribute to our understanding of the combined effects of genetic and environmental factors in shaping disease risk.
Collapse
Affiliation(s)
- Torsten Klengel
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|