51
|
Qiao YL, Zhou JJ, Liang JH, Deng XP, Zhang ZJ, Huang HL, Li S, Dai SF, Liu CQ, Luan ZL, Yu ZL, Sun CP, Ma XC. Uncaria rhynchophylla ameliorates unpredictable chronic mild stress-induced depression in mice via activating 5-HT 1A receptor: Insights from transcriptomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 81:153436. [PMID: 33360346 DOI: 10.1016/j.phymed.2020.153436] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/29/2020] [Accepted: 12/03/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Depression is a pervasive or persistent mental disorder that causes mood, cognitive and memory deficits. Uncaria rhynchophylla has been widely used to treat central nervous system diseases for a long history, although its efficacy and potential mechanism are still uncertain. PURPOSE The present study aimed to investigate anti-depression effect and potential mechanism of U. rhynchophylla extract (URE). STUDY DESIGN AND METHODS A mouse depression model was established using unpredictable chronic mild stress (UCMS). Effects of URE on depression-like behaviours, neurotransmitters, and neuroendocrine hormones were investigated in UCMS-induced mice. The potential target of URE was analyzed by transcriptomics and bioinformatics methods and validated by RT-PCR and Western blot. The agonistic effect on 5-HT1A receptor was assayed by dual-luciferase reporter system. RESULTS URE ameliorated depression-like behaviours, and modulated levels of neurotransmitters and neuroendocrine hormones, including 5-hydroxytryptamine (5-HT), 5-hydroxyindole acetic acid (5-HIAA), dopamine (DA), 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), corticosterone (CORT), corticotropin-releasing hormone (CRH), and adrenocorticotropic hormone (ACTH), in UCMS-induced mice. Transcriptomics and bioinformatics results indicated that URE could regulate glutamatergic, cholinergic, serotonergic, and GABAergic systems, especially neuroactive ligand-receptor and cAMP signaling pathways, revealing that Htr1a encoding 5-HT1A receptor was a potential target of URE. The expression levels of downstream proteins of 5-HT1A signaling pathway 5-HT1A, CREB, BDNF, and PKA were increased in UCMS-induced mice after URE administration, and URE also displayed an agonistic effect against 5-HT1A receptor with an EC50 value of 17.42 μg/ml. CONCLUSION U. rhynchophylla ameliorated depression-like behaviours in UCMS-induced mice through activating 5-HT1A receptor.
Collapse
Affiliation(s)
- Yan-Ling Qiao
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Jun-Jun Zhou
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Jia-Hao Liang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Xiao-Peng Deng
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhan-Jun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Hui-Lian Huang
- Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Song Li
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Shu-Fang Dai
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Chun-Qing Liu
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhi-Lin Luan
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhen-Long Yu
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Cheng-Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China.
| | - Xiao-Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China; State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China.
| |
Collapse
|
52
|
Understanding stress: Insights from rodent models. CURRENT RESEARCH IN NEUROBIOLOGY 2021; 2:100013. [PMID: 36246514 PMCID: PMC9559100 DOI: 10.1016/j.crneur.2021.100013] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 02/01/2023] Open
Abstract
Through incorporating both physical and psychological forms of stressors, a variety of rodent models have provided important insights into the understanding of stress physiology. Rodent models also have provided significant information with regards to the mechanistic basis of the pathophysiology of stress-related disorders such as anxiety disorders, depressive illnesses, cognitive impairment and post-traumatic stress disorder. Additionally, rodent models of stress have served as valuable tools in the area of drug screening and drug development for treatment of stress-induced conditions. Although rodent models do not accurately reproduce the biochemical or physiological parameters of stress response and cannot fully mimic the natural progression of human disorders, yet, animal research has provided answers to many important scientific questions. In this review article, important studies utilizing a variety of stress models are described in terms of their design and apparatus, with specific focus on their capabilities to generate reliable behavioral and biochemical read-out. The review focusses on the utility of rodent models by discussing examples in the literature that offer important mechanistic insights into physiologically relevant questions. The review highlights the utility of rodent models of stress as important tools for advancing the mission of scientific research and inquiry. Stressful life events may lead to the onset of severe psychopathologies in humans. Rodents may model many features of stress exposure in human populations. Induction of stress via pharmacological and psychological manipulations alter rodent behavior. Mechanistic rodent studies reveal key molecular targets critical for new therapeutic targets.
Collapse
|
53
|
Bolton JL, Schulmann A, Garcia-Curran MM, Regev L, Chen Y, Kamei N, Shao M, Singh-Taylor A, Jiang S, Noam Y, Molet J, Mortazavi A, Baram TZ. Unexpected Transcriptional Programs Contribute to Hippocampal Memory Deficits and Neuronal Stunting after Early-Life Adversity. Cell Rep 2020; 33:108511. [PMID: 33326786 PMCID: PMC7817243 DOI: 10.1016/j.celrep.2020.108511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 07/08/2020] [Accepted: 11/19/2020] [Indexed: 01/23/2023] Open
Abstract
Early-life adversity (ELA) is associated with lifelong memory deficits, yet the responsible mechanisms remain unclear. We impose ELA by rearing rat pups in simulated poverty, assess hippocampal memory, and probe changes in gene expression, their transcriptional regulation, and the consequent changes in hippocampal neuronal structure. ELA rats have poor hippocampal memory and stunted hippocampal pyramidal neurons associated with ~140 differentially expressed genes. Upstream regulators of the altered genes include glucocorticoid receptor and, unexpectedly, the transcription factor neuron-restrictive silencer factor (NRSF/REST). NRSF contributes critically to the memory deficits because blocking its function transiently following ELA rescues spatial memory and restores the dendritic arborization of hippocampal pyramidal neurons in ELA rats. Blocking NRSF function in vitro augments dendritic complexity of developing hippocampal neurons, suggesting that NRSF represses genes involved in neuronal maturation. These findings establish important, surprising contributions of NRSF to ELA-induced transcriptional programming that disrupts hippocampal maturation and memory function.
Collapse
Affiliation(s)
- Jessica L Bolton
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Anton Schulmann
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Megan M Garcia-Curran
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Limor Regev
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Yuncai Chen
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Noriko Kamei
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Manlin Shao
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Akanksha Singh-Taylor
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Shan Jiang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Yoav Noam
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Jenny Molet
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Neurology, University of California, Irvine, Irvine, CA 92697-4475, USA.
| |
Collapse
|
54
|
The effects of childhood trauma on the onset, severity and improvement of depression: The role of dysfunctional attitudes and cortisol levels. J Affect Disord 2020; 276:402-410. [PMID: 32871670 DOI: 10.1016/j.jad.2020.07.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/31/2020] [Accepted: 07/05/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Childhood trauma is an important early social risk factor for the development of the major depressive disorder (MDD). Both childhood trauma and depression are associated with dysfunctional attitudes and dysregulation in stress hormones. We aimed to clarify the path from childhood trauma to depression and identify potential predictors of antidepressant treatment outcomes. OBJECTIVES One hundred and thirty-nine MDD patients and 112 healthy controls were included at baseline. Depressive symptoms were assessed with both self-reported and expert-rated scales. Childhood trauma and dysfunctional attitudes were evaluated and blood cortisol levels were assayed. Patients received an open-label antidepressant trial with paroxetine and their depressive symptoms were monitored by the Hamilton Depression Rating Scale (HAMD) during 6 months of treatment. After 6 months, 94 patients received the same assessments as the baseline. RESULTS At baseline, the influence of childhood trauma on depression diagnosis was mediated by dysfunctional attitudes. In patients with MDD, the influence of childhood trauma on depression severity was mediated by both dysfunctional attitudes and cortisol levels. Baseline childhood trauma predicted the antidepressant treatment outcome during early treatment phase and baseline cortisol levels predicted the antidepressant treatment outcome at later treatment phase. After 6-month antidepressant treatment, a significant remission by time effect was found on dysfunctional attitudes and depression severity but not on cortisol levels. CONCLUSION Effect of childhood trauma on depression onset was mediated by dysfunctional attitudes. The relationship between childhood trauma and depressive symptoms was mediated by dysfunctional attitudes and cortisol levels in MDD patients. Baseline childhood trauma and cortisol levels may be moderators for antidepressant treatment response at different treatment phase.
Collapse
|
55
|
Cordier JM, Aguggia JP, Danelon V, Mir FR, Rivarola MA, Mascó D. Postweaning Enriched Environment Enhances Cognitive Function and Brain-Derived Neurotrophic Factor Signaling in the Hippocampus in Maternally Separated Rats. Neuroscience 2020; 453:138-147. [PMID: 33039520 DOI: 10.1016/j.neuroscience.2020.09.058] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022]
Abstract
Adverse environments during early life may lead to different neurophysiological and behavioral consequences, including depression and learning and memory deficits that persist into adulthood. Previously, we demonstrated that exposure to an enriched environment during adolescence mitigates the cognitive impairment observed after maternal separation in a task-specific manner. However, underlying neural mechanisms are still not fully understood. The current study examines the effects of neonatal maternal separation (MS) and postweaning environmental enrichment (EE) on spatial learning and memory performance in a short version of the Barnes Maze, active and passive behaviors in the forced swim test, and on TrkB/BDNF receptor expression in the hippocampus. Our results revealed that MS impaired acquisition learning and that enriched rats performed better than non-enriched rats in acquisition trials, regardless of early conditions. During the probe, enriched-housed rats demonstrated better performance than those reared in standard conditions. No significant differences between groups were found in the forced swim test. Both MS and EE increase full-length TrkB expression, and the combination of MS and EE treatment caused the highest levels of this protein expression. Similarly, truncated TrkB expression was higher in the MS/EE group. Animal facility rearing (AFR) non-enriched groups present the lowest activation of phosphorylated Erk, a canonical downstream kinase of TrkB signaling. Taken together, our results demonstrate the importance of enriched environment as an intervention to ameliorate the effects of maternal separation on spatial learning and memory. TrkB/BDNF signaling could mediate neuroplastic changes related to learning and memory during exposure to enriched environment.
Collapse
Affiliation(s)
- Javier Maximiliano Cordier
- Instituto de Diversidad y Ecología Animal (IDEA), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba -Córdoba, Argentina
| | - Julieta Paola Aguggia
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba, Enrique Barros esq. Enfermera Gordillo. Ciudad Universitaria, CP: 5016, Córdoba, Argentina
| | - Víctor Danelon
- Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 1611, CP: 5016, Córdoba, Argentina
| | - Franco Rafael Mir
- Cátedra de Fisiología Animal, Departamento de Ciencias Exactas Físicas y Naturales, Universidad Nacional de La Rioja, Av. Luis M. de la Fuente S/N, Ciudad Universitaria de la Ciencia y de la Técnica, F5300 La Rioja, Argentina; Cátedra de Fisiología Animal, Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 299 X5000JJC- Córdoba, Argentina
| | - María Angélica Rivarola
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba, Enrique Barros esq. Enfermera Gordillo. Ciudad Universitaria, CP: 5016, Córdoba, Argentina; Cátedra de Fisiología Animal, Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 299 X5000JJC- Córdoba, Argentina.
| | - Daniel Mascó
- Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 1611, CP: 5016, Córdoba, Argentina
| |
Collapse
|
56
|
Notaras M, van den Buuse M. Neurobiology of BDNF in fear memory, sensitivity to stress, and stress-related disorders. Mol Psychiatry 2020; 25:2251-2274. [PMID: 31900428 DOI: 10.1038/s41380-019-0639-2] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/01/2019] [Accepted: 12/12/2019] [Indexed: 01/17/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is widely accepted for its involvement in resilience and antidepressant drug action, is a common genetic locus of risk for mental illnesses, and remains one of the most prominently studied molecules within psychiatry. Stress, which arguably remains the "lowest common denominator" risk factor for several mental illnesses, targets BDNF in disease-implicated brain regions and circuits. Altered stress-related responses have also been observed in animal models of BDNF deficiency in vivo, and BDNF is a common downstream intermediary for environmental factors that potentiate anxiety- and depressive-like behavior. However, BDNF's broad functionality has manifested a heterogeneous literature; likely reflecting that BDNF plays a hitherto under-recognized multifactorial role as both a regulator and target of stress hormone signaling within the brain. The role of BDNF in vulnerability to stress and stress-related disorders, such as posttraumatic stress disorder (PTSD), is a prominent example where inconsistent effects have emerged across numerous models, labs, and disciplines. In the current review we provide a contemporary update on the neurobiology of BDNF including new data from the behavioral neuroscience and neuropsychiatry literature on fear memory consolidation and extinction, stress, and PTSD. First we present an overview of recent advances in knowledge on the role of BDNF within the fear circuitry, as well as address mounting evidence whereby stress hormones interact with endogenous BDNF-TrkB signaling to alter brain homeostasis. Glucocorticoid signaling also acutely recruits BDNF to enhance the expression of fear memory. We then include observations that the functional common BDNF Val66Met polymorphism modulates stress susceptibility as well as stress-related and stress-inducible neuropsychiatric endophenotypes in both man and mouse. We conclude by proposing a BDNF stress-sensitivity hypothesis, which posits that disruption of endogenous BDNF activity by common factors (such as the BDNF Val66Met variant) potentiates sensitivity to stress and, by extension, vulnerability to stress-inducible illnesses. Thus, BDNF may induce plasticity to deleteriously promote the encoding of fear and trauma but, conversely, also enable adaptive plasticity during extinction learning to suppress PTSD-like fear responses. Ergo regulators of BDNF availability, such as the Val66Met polymorphism, may orchestrate sensitivity to stress, trauma, and risk of stress-induced disorders such as PTSD. Given an increasing interest in personalized psychiatry and clinically complex cases, this model provides a framework from which to experimentally disentangle the causal actions of BDNF in stress responses, which likely interact to potentiate, produce, and impair treatment of, stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia. .,College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia. .,Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
57
|
Blues in the Brain and Beyond: Molecular Bases of Major Depressive Disorder and Relative Pharmacological and Non-Pharmacological Treatments. Genes (Basel) 2020; 11:genes11091089. [PMID: 32961910 PMCID: PMC7564223 DOI: 10.3390/genes11091089] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Despite the extensive research conducted in recent decades, the molecular mechanisms underlying major depressive disorder (MDD) and relative evidence-based treatments remain unclear. Various hypotheses have been successively proposed, involving different biological systems. This narrative review aims to critically illustrate the main pathogenic hypotheses of MDD, ranging from the historical ones based on the monoaminergic and neurotrophic theories, through the subsequent neurodevelopmental, glutamatergic, GABAergic, inflammatory/immune and endocrine explanations, until the most recent evidence postulating a role for fatty acids and the gut microbiota. Moreover, the molecular effects of established both pharmacological and non-pharmacological approaches for MDD are also reviewed. Overall, the existing literature indicates that the molecular mechanisms described in the context of these different hypotheses, rather than representing alternative ones to each other, are likely to contribute together, often with reciprocal interactions, to the development of MDD and to the effectiveness of treatments, and points at the need for further research efforts in this field.
Collapse
|
58
|
Yang Y, Yang S, Jia Y, Yin C, Zhao R. Sex-biased transgenerational transmission of betaine-induced epigenetic modifications in glucocorticoid receptor gene and its down-stream BDNF/ERK pathway in rat hippocampus. Nutr Neurosci 2020; 25:746-757. [PMID: 32840180 DOI: 10.1080/1028415x.2020.1807711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Objectives: Glucocorticoid receptor (GR) expressed in hippocampus is critical for the homeostasis of stress responses and susceptible to epigenetic modulation caused by maternal factors. Here we show that maternal methyl nutrition causes sex-biased changes in hippocampal expression of GR exon 1 mRNA variants, associated with promoter DNA methylation, across two offspring generations in rats.Methods: Three-month-old female Sprague-Dawley rats (F0) were fed a diet supplemented with 1% betaine throughout the gestation and lactation. F0 dams and their F1 and F2 offspring of both sexes at weaning were used in the study.Results: A sex-specific transgenerational effect was observed. F2 females, but not males, followed the same pattern of their grand dams showing increased mRNA expression of total GR and its exons 1.4, 1.7, 1.10 and 1.11 variants coincided with promoter DNA hypomethylation in the hippocampus. However, F1 females, but not males, exhibited an opposite pattern, showing decreased expression of GR and its mRNA variants accompanied with promoter hypermethylation. The protein content of phospho-GR and BDNF/ERK in the hippocampus displayed the same sex and generation specificity.Discussion: These results indicate that maternal betaine exerts transgenerational effects on hippocampal GR expression and BDNF/ERK pathway in female rat offspring, with generation-dependent patterns of DNA methylation on alternative GR promoters.
Collapse
Affiliation(s)
- Yang Yang
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, People's Republic of China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Shu Yang
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, People's Republic of China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Yimin Jia
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, People's Republic of China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Chao Yin
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, People's Republic of China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, People's Republic of China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, People's Republic of China
| |
Collapse
|
59
|
Early-life short-term environmental enrichment counteracts the effects of stress on anxiety-like behavior, brain-derived neurotrophic factor and nuclear translocation of glucocorticoid receptors in the basolateral amygdala. Sci Rep 2020; 10:14053. [PMID: 32820184 PMCID: PMC7441150 DOI: 10.1038/s41598-020-70875-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/30/2020] [Indexed: 01/16/2023] Open
Abstract
Early life is a decisive stage for the development of physiological and psychological characteristics of an individual. Any stress or disruption of healthy development at this stage has serious long-lasting consequences for the remaining life. Unfortunately, early life stress is a common occurrence in humans and other animals. In this context, we investigated if the provision of environmental enrichment during the pre-weaning phase of rat pups and dams could alter the consequences of early-life maternal-separation stress. Pre-weaning enrichment rescued the effects of maternal separation on the excess secretion of adrenal stress hormones and anxiety-like behavior during adulthood. Enrichment also reduced the effect of stress on the spine density of basolateral amygdala neurons, a brain region critical for stress-induced facilitation of emotional behaviors. Pre-weaning enrichment, provided during early-life, blunted the effects of maternal separation stress on decreased intra-nuclear translocation of glucocorticoid receptors within the amygdala neurons when tested later in adulthood. Early-life, pre-weaning environmental enrichment also increased the amount of brain-derived neurotrophic factor within adult basolateral amygdala. Our observations showed that environmental manipulation during early formative years could be utilized to build lifelong resilience to stress. Complex naturalistic housing and sensory enrichment is, thus, an useful buffer against an impoverished and stressful childhood.
Collapse
|
60
|
Effects of Maternal Chewing on Prenatal Stress-Induced Cognitive Impairments in the Offspring via Multiple Molecular Pathways. Int J Mol Sci 2020; 21:ijms21165627. [PMID: 32781547 PMCID: PMC7460630 DOI: 10.3390/ijms21165627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022] Open
Abstract
We aimed to investigate the effects of maternal chewing on prenatal stress-induced cognitive impairments in the offspring and to explore the molecular pathways of maternal chewing in a mice model. Maternal chewing ameliorated spatial learning impairments in the offspring in a Morris water maze test. Immunohistochemistry and Western blot findings revealed that maternal chewing alleviated hippocampal neurogenesis impairment and increased the expression of hippocampal brain-derived neurotrophic factor in the offspring. In addition, maternal chewing increased the expression of glucocorticoid receptor (GR) and 11β-hydroxysteroid dehydrogenase isozyme 2 (11β-HSD2) and decreased the expression of 11β-HSD1 in the placenta, thereby attenuating the increase of glucocorticoid in the offspring. Furthermore, maternal chewing increased the expression of 11β-HSD2, FK506-binding protein 51 (FKBP51) and FKBP52 and decreased the expression of 11β-HSD1, thereby increasing hippocampal nuclear GR level. In addition, maternal chewing attenuated the increase in expression of DNMT1 and DNMT3a and the decrease in expression of histone H3 methylation at lysine 4, 9, 27 and histone H3 acetylation at lysine 9 induced by prenatal stress in the offspring. Our findings suggest that maternal chewing could ameliorate prenatal stress-induced cognitive impairments in the offspring at least in part by protecting placenta barrier function, alleviating hippocampal nuclear GR transport impairment and increasing the hippocampal brain-derived neurotrophic factor (BDNF) level.
Collapse
|
61
|
Garcia-Rizo C, Bitanihirwe BKY. Implications of early life stress on fetal metabolic programming of schizophrenia: A focus on epiphenomena underlying morbidity and early mortality. Prog Neuropsychopharmacol Biol Psychiatry 2020; 101:109910. [PMID: 32142745 DOI: 10.1016/j.pnpbp.2020.109910] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/16/2020] [Accepted: 03/03/2020] [Indexed: 12/19/2022]
Abstract
The fetal origin of adult disease hypothesis postulates that a stressful in utero environment can have deleterious consequences on fetal programming, potentially leading to chronic disease in later life. Factors known to impact fetal programming include the timing, intensity, duration and nature of the external stressor during pregnancy. As such, dynamic modulation of fetal programming is heavily involved in shaping health throughout the life course, possibly by influencing metabolic parameters including insulin action, hypothalamic-pituitary-adrenal activity and immune function. The ability of prenatal insults to program adult disease is likely to occur as a result of reduced functional capacity in key organs-a "thrifty" phenotype-where more resources are re-allocated to preserve critical organs such as the brain. Notably, it has been postulated that the manifestation of neuropsychiatric disorders in individuals priorly exposed to prenatal stress may arise from the interaction between hereditary factors and the intrauterine environment, which together precipitate disease onset by disrupting the trajectory of normal brain development. In this review we discuss the evidence linking prenatal programming to neuropsychiatric disorders, mainly schizophrenia, via a "Thrifty psychiatric phenotype" concept. We start by outlining the conception of the thrifty psychiatric phenotype. Next, we discuss the convergence of potential mechanistic pathways through which prenatal insults may trigger epigenetic changes that contribute to the increased morbidity and early mortality observed in neuropsychiatric disorders. Finally, we touch on the public health importance of fetal programming for these disorders. We conclude by providing a brief outlook on the future of this evolving field of research.
Collapse
Affiliation(s)
- Clemente Garcia-Rizo
- Barcelona Clinic Schizophrenia Unit, Neuroscience Institute, Hospital Clinic, Barcelona, Spain; Institute of Biomedical Research Agusti Pi iSunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain.
| | - Byron K Y Bitanihirwe
- Centre for Global Health, Trinity College Dublin, Dublin, Ireland; Department of Psychology, Trinity College Dublin, Dublin, Ireland; School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
62
|
Hoffman KW, Lee JJ, Corcoran CM, Kimhy D, Kranz TM, Malaspina D. Considering the Microbiome in Stress-Related and Neurodevelopmental Trajectories to Schizophrenia. Front Psychiatry 2020; 11:629. [PMID: 32719625 PMCID: PMC7350783 DOI: 10.3389/fpsyt.2020.00629] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Early life adversity and prenatal stress are consistently associated with an increased risk for schizophrenia, although the exact pathogenic mechanisms linking the exposures with the disease remain elusive. Our previous view of the HPA stress axis as an elegant but simple negative feedback loop, orchestrating adaptation to stressors among the hypothalamus, pituitary, and adrenal glands, needs to be updated. Research in the last two decades shows that important bidirectional signaling between the HPA axis and intestinal mucosa modulates brain function and neurochemistry, including effects on glucocorticoid hormones and brain-derived neurotrophic factor (BDNF). The intestinal microbiome in earliest life, which is seeded by the vaginal microbiome during delivery, programs the development of the HPA axis in a critical developmental window, determining stress sensitivity and HPA function as well as immune system development. The crosstalk between the HPA and the Microbiome Gut Brain Axis (MGBA) is particularly high in the hippocampus, the most consistently disrupted neural region in persons with schizophrenia. Animal models suggest that the MGBA remains influential on behavior and physiology across developmental stages, including the perinatal window, early childhood, adolescence, and young adulthood. Understanding the role of the microbiome on critical risk related stressors may enhance or transform of understanding of the origins of schizophrenia and offer new approaches to increase resilience against stress effects for preventing and treating schizophrenia.
Collapse
Affiliation(s)
- Kevin W. Hoffman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jakleen J. Lee
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Cheryl M. Corcoran
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- James J. Peters VA Medical Center, Mental Illness Research, Education and Clinical Centers (MIRECC), New York, NY, United States
| | - David Kimhy
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- James J. Peters VA Medical Center, Mental Illness Research, Education and Clinical Centers (MIRECC), New York, NY, United States
| | - Thorsten M. Kranz
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Dolores Malaspina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
63
|
Watt T, Ceballos N, Kim S, Pan X, Sharma S. The Unique Nature of Depression and Anxiety among College Students with Adverse Childhood Experiences. JOURNAL OF CHILD & ADOLESCENT TRAUMA 2020; 13:163-172. [PMID: 32549928 PMCID: PMC7289944 DOI: 10.1007/s40653-019-00270-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
It is well established that adverse childhood experiences (ACEs) contribute to the development of mental disorders in adulthood. However, less is known about how childhood trauma impacts the mind and the body, whether the resulting mental disorders have different characteristics than those occurring without these antecedent conditions, and if treatment modalities need to reflect the unique nature of mental disorders rooted in trauma. Survey and biomarker data were gathered from a sample of college students (n = 93) to explore the relationship between childhood trauma and mental health. We examine how neuroimmune systems (inflammation and neuroplasticity) relate to depression and anxiety and whether these associations vary for those with and without a history of childhood trauma. Findings reveal that students with 4 or more ACEs are more likely to have depression and anxiety than students without these experiences. In addition, we find that inflammation (CRP) and neuronal health (BDNF) are associated with mental health disorders among students with four or more ACEs, but not for students without this history. These findings suggest that mental disorders associated with four or more ACEs may be uniquely tied to physiological processes, and consequently, warrant tailored treatments. The implications for mental health intervention include, 1) screening for childhood trauma, inflammation, and neuronal health and 2) referral to treatments which are theoretically and empirically tied to the root causes of mental disorders rather than those designed merely to suppress their symptoms.
Collapse
Affiliation(s)
- Toni Watt
- Department of Sociology, Texas State University, 601 University Drive, San Marcos, TX 78666 USA
| | - Natalie Ceballos
- Department of Sociology, Texas State University, 601 University Drive, San Marcos, TX 78666 USA
| | - Seoyoun Kim
- Department of Sociology, Texas State University, 601 University Drive, San Marcos, TX 78666 USA
| | - Xi Pan
- Department of Sociology, Texas State University, 601 University Drive, San Marcos, TX 78666 USA
| | - Shobhit Sharma
- Department of Sociology, Texas State University, 601 University Drive, San Marcos, TX 78666 USA
| |
Collapse
|
64
|
Extremely low birth weight influences the relationship between stress and telomere length in adulthood. J Dev Orig Health Dis 2020; 12:328-334. [PMID: 32468974 DOI: 10.1017/s2040174420000409] [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] [Indexed: 11/07/2022]
Abstract
This study examined the link between two biological markers of stress vulnerability at 22-26 years of age and telomere length at 30-35 among extremely low birth weight (ELBW; <1000 g) survivors and normal birth weight (NBW; >2500 g) control participants. Sixteen ELBW and 22 NBW participants provided baseline afternoon salivary cortisol samples and resting frontal electroencephalogram (EEG) alpha asymmetry data at 22-26 years. Buccal cells were assayed for telomere length at 30-35 years. Analyses controlled for sex, postnatal steroid exposure, childhood socioeconomic status, time of cortisol sample collection, and body mass index at 22-26 years. Salivary cortisol and frontal asymmetry at age 22-26 independently predicted telomere length at age 30-35, such that relatively higher cortisol and greater relative right frontal asymmetry at rest predicted telomere shortening among NBW controls, but not among ELBW survivors. However, similar associations were not noted in ELBW survivors, suggesting that ELBW survivors may have different mechanisms of stress coping as a result of their early-life exposures. These findings offer preliminary evidence in support of the role of stress in the genesis of cellular senescence at least among those born at NBW, but that these links may differ in those born preterm.
Collapse
|
65
|
Prowse N, Dwyer Z, Thompson A, Fortin T, Elson K, Robeson H, Fenner B, Hayley S. Early life selective knockdown of the TrkB receptor and maternal separation modulates adult stress phenotype. Behav Brain Res 2020; 378:112260. [DOI: 10.1016/j.bbr.2019.112260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/23/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022]
|
66
|
Rajan KE, Soundarya S, Karen C, Shanmugapriya V, Radhakrishnan K. Presence of Mother Reduces Early-Life Social Stress: Linking the Alteration in Hypothalamic-Pituitary-Adrenal Axis and Serotonergic System. Dev Neurosci 2019; 41:212-222. [PMID: 31865338 DOI: 10.1159/000504508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 10/30/2019] [Indexed: 11/19/2022] Open
Abstract
In this study, we examined whether the presence of mother suppresses early-life stressful social experience (SSE)-induced anxiety-like behavior and impairment of short-term memory later in life. On postnatal day (PND)-5, mothers with pups were grouped as follows: (i) control; (ii) maternal separation (MS); (iii) pups with mother experience the presence of a stranger (M+P-ST); and (iv) maternal separated pups experience the presence of a stranger (MSP-ST). Individuals were subjected to light-dark box and spontaneous alternation from PND-29 to 32. We observed that the MSP-ST group exhibits anxiety-like behavior and impairment in short-term memory. Further, SSE significantly elevated the adrenocorticotropic hormone, corticosterone and expression of glucocorticoid receptor (GR) in MSP-ST pups. Similarly, serotonin (5-hydroxytryptamine; 5-HT), dopamine, noradrenaline and expression of serotonin transporter levels were significantly elevated in MSP-ST pups. These observations suggest that during early postnatal days, the pups may recognize strangers by the sense of smell, and the presence of mother reduces the SSE-induced stress.
Collapse
Affiliation(s)
- Koilmani Emmanuvel Rajan
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India,
| | - Suba Soundarya
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India.,School of Molecular Sciences, The University of Western Australia, Perth, Washington, Australia
| | - Christopher Karen
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Vasudevan Shanmugapriya
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Karuppasamy Radhakrishnan
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India.,Department of Zoology, Government Arts College, Karur, India
| |
Collapse
|
67
|
Torres-Berrío A, Issler O, Parise EM, Nestler EJ. Unraveling the epigenetic landscape of depression: focus on early life stress
. DIALOGUES IN CLINICAL NEUROSCIENCE 2019; 21:341-357. [PMID: 31949402 PMCID: PMC6952747 DOI: 10.31887/dcns.2019.21.4/enestler] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Depression is a devastating psychiatric disorder caused by a combination of genetic predisposition and life events, mainly exposure to stress. Early life stress (ELS) in particular is known to "scar" the brain, leading to an increased susceptibility to developing depression later in life via epigenetic mechanisms. Epigenetic processes lead to changes in gene expression that are not due to changes in DNA sequence, but achieved via modulation of chromatin modifications, DNA methylation, and noncoding RNAs. Here we review common epigenetic mechanisms including the enzymes that take part in reading, writing, and erasing specific epigenetic marks. We then describe recent developments in understanding how ELS leads to changes in the epigenome that are manifested in increased susceptibility to depression-like abnormalities in animal models. We conclude with highlighting the need for future studies that will potentially enable the utilisation of the understanding of epigenetic changes linked to ELS for the development of much-needed novel therapeutic strategies and biomarker discovery.
.
Collapse
Affiliation(s)
- Angélica Torres-Berrío
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Orna Issler
- Author affiliations: Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Eric M Parise
- Author affiliations: Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Eric J Nestler
- Author affiliations: Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| |
Collapse
|
68
|
Gulyaeva NV. Biochemical Mechanisms and Translational Relevance of Hippocampal Vulnerability to Distant Focal Brain Injury: The Price of Stress Response. BIOCHEMISTRY (MOSCOW) 2019; 84:1306-1328. [PMID: 31760920 DOI: 10.1134/s0006297919110087] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Focal brain injuries (in particular, stroke and traumatic brain injury) induce with high probability the development of delayed (months, years) cognitive and depressive disturbances which are frequently comorbid. The association of these complications with hippocampal alterations (in spite of the lack of a primary injury of this structure), as well as the lack of a clear dependence between the probability of depression and dementia development and primary damage severity and localization served as the basis for a new hypothesis on the distant hippocampal damage as a key link in the pathogenesis of cognitive and psychiatric disturbances. According to this hypothesis, the excess of corticosteroids secreted after a focal brain damage, in particular in patients with abnormal stress-response due to hypothalamic-pituitary-adrenal axis (HPAA) dysfunction, interacts with corticosteroid receptors in the hippocampus inducing signaling pathways which stimulate neuroinflammation and subsequent events including disturbances in neurogenesis and hippocampal neurodegeneration. In this article, the molecular and cellular mechanisms associated with the regulatory role of the HPAA and multiple functions of brain corticosteroid receptors in the hippocampus are analyzed. Functional and structural damage to the hippocampus, a brain region selectively vulnerable to external factors and responding to them by increased cytokine secretion, forms the basis for cognitive function disturbances and psychopathology development. This concept is confirmed by our own experimental data, results of other groups and by prospective clinical studies of post-stroke complications. Clinically relevant biochemical approaches to predict the risks and probability of post-stroke/post-trauma cognitive and depressive disturbances are suggested using the evaluation of biochemical markers of patients' individual stress-response. Pathogenetically justified ways for preventing these consequences of focal brain damage are proposed by targeting key molecular mechanisms underlying hippocampal dysfunction.
Collapse
Affiliation(s)
- N V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia. .,Moscow Research and Clinical Center for Neuropsychiatry, Healthcare Department of Moscow, Moscow, 115419, Russia
| |
Collapse
|
69
|
Music exposure attenuates anxiety- and depression-like behaviors and increases hippocampal spine density in male rats. Behav Brain Res 2019; 372:112023. [DOI: 10.1016/j.bbr.2019.112023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/18/2019] [Accepted: 06/07/2019] [Indexed: 01/14/2023]
|
70
|
Androulakis IP. The quest for digital health: From diseases to patients. Comput Chem Eng 2019. [DOI: 10.1016/j.compchemeng.2019.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
71
|
Comasco E, Schijven D, de Maeyer H, Vrettou M, Nylander I, Sundström-Poromaa I, Olivier JDA. Constitutive Serotonin Transporter Reduction Resembles Maternal Separation with Regard to Stress-Related Gene Expression. ACS Chem Neurosci 2019; 10:3132-3142. [PMID: 30614673 DOI: 10.1021/acschemneuro.8b00595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Interactive effects between allelic variants of the serotonin transporter (5-HTT) promoter-linked polymorphic region (5-HTTLPR) and stressors on depression symptoms have been documented, as well as questioned, by meta-analyses. Translational models of constitutive 5-htt reduction and experimentally controlled stressors often led to inconsistent behavioral and molecular findings and often did not include females. The present study sought to investigate the effect of 5-htt genotype, maternal separation, and sex on the expression of stress-related candidate genes in the rat hippocampus and frontal cortex. The mRNA expression levels of Avp, Pomc, Crh, Crhbp, Crhr1, Bdnf, Ntrk2, Maoa, Maob, and Comt were assessed in the hippocampus and frontal cortex of 5-htt ± and 5-htt +/+ male and female adult rats exposed, or not, to daily maternal separation for 180 min during the first 2 postnatal weeks. Gene- and brain region-dependent, but sex-independent, interactions between 5-htt genotype and maternal separation were found. Gene expression levels were higher in 5-htt +/+ rats not exposed to maternal separation compared with the other experimental groups. Maternal separation and 5-htt +/- genotype did not yield additive effects on gene expression. Correlative relationships, mainly positive, were observed within, but not across, brain regions in all groups except in non-maternally separated 5-htt +/+ rats. Gene expression patterns in the hippocampus and frontal cortex of rats exposed to maternal separation resembled the ones observed in rats with reduced 5-htt expression regardless of sex. These results suggest that floor effects of 5-htt reduction and maternal separation might explain inconsistent findings in humans and rodents.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jocelien D. A. Olivier
- Department Neurobiology, Unit Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9712 CP, The Netherlands
| |
Collapse
|
72
|
Atrooz F, Liu H, Salim S. Stress, psychiatric disorders, molecular targets, and more. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 167:77-105. [PMID: 31601407 DOI: 10.1016/bs.pmbts.2019.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mental health is central to normal health outcomes. A widely accepted theory is that chronic persistent stress during adulthood as well as during early life triggers onset of neuropsychiatric ailments. However, questions related to how that occurs, and why are some individuals resistant to stress while others are not, remain unanswered. An integrated, multisystemic stress response involving neuroinflammatory, neuroendocrine, epigenetic and metabolic cascades have been suggested to have causative links. Several theories have been proposed over the years to conceptualize this link including the cytokine hypothesis, the endocrine hypothesis, the oxidative stress hypothesis and the oxido-neuroinflammation hypothesis. The data discussed in this review describes potential biochemical basis of the link between stress, and stress-induced neuronal, behavioral and emotional deficits, providing insights into potentially novel drug targets.
Collapse
Affiliation(s)
- Fatin Atrooz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Hesong Liu
- Baylor College of Medicine, Houston, TX, United States
| | - Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.
| |
Collapse
|
73
|
Persistence of learning-induced synapses depends on neurotrophic priming of glucocorticoid receptors. Proc Natl Acad Sci U S A 2019; 116:13097-13106. [PMID: 31182610 PMCID: PMC6601006 DOI: 10.1073/pnas.1903203116] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Signal transduction upon activation of receptor tyrosine kinases by neurotrophins and nuclear receptors by glucocorticoids is essential for homeostasis. Phosphorylation (PO4) is one way these receptors communicate with one another to support homeostatic reactions in learning and memory. Using a newly developed glucocorticoid receptor (GR)-PO4–deficient knock-in mouse, we show that consolidation of learning-induced neuroplasticity depends on both GR-PO4 and neurotrophic signaling. Cross-talk between these pathways affects experience-dependent neuroplasticity and behavior, extending previous implications of neurotrophic priming of glucocorticoid response for adaptive plasticity to chronic stress and antidepressant response. Therefore, a disruption of cross-talk between these pathways by, for example, the misalignment of circadian glucocorticoid release and experience-dependent neurotrophic signaling may contribute to the pathophysiology of stress-related disorders. Stress can either promote or impair learning and memory. Such opposing effects depend on whether synapses persist or decay after learning. Maintenance of new synapses formed at the time of learning upon neuronal network activation depends on the stress hormone-activated glucocorticoid receptor (GR) and neurotrophic factor release. Whether and how concurrent GR and neurotrophin signaling integrate to modulate synaptic plasticity and learning is not fully understood. Here, we show that deletion of the neurotrophin brain-derived neurotrophic factor (BDNF)–dependent GR-phosphorylation (PO4) sites impairs long-term memory retention and maintenance of newly formed postsynaptic dendritic spines in the mouse cortex after motor skills training. Chronic stress and the BDNF polymorphism Val66Met disrupt the BDNF-dependent GR-PO4 pathway necessary for preserving training-induced spines and previously acquired memories. Conversely, enrichment living promotes spine formation but fails to salvage training-related spines in mice lacking BDNF-dependent GR-PO4 sites, suggesting it is essential for spine consolidation and memory retention. Mechanistically, spine maturation and persistence in the motor cortex depend on synaptic mobilization of the glutamate receptor subunit A1 (GluA1) mediated by GR-PO4. Together, these findings indicate that regulation of GR-PO4 via activity-dependent BDNF signaling is important for the formation and maintenance of learning-dependent synapses. They also define a signaling mechanism underlying these effects.
Collapse
|
74
|
Couto-Pereira NDS, Lampert C, Vieira ADS, Lazzaretti C, Kincheski GC, Espejo PJ, Molina VA, Quillfeldt JA, Dalmaz C. Resilience and Vulnerability to Trauma: Early Life Interventions Modulate Aversive Memory Reconsolidation in the Dorsal Hippocampus. Front Mol Neurosci 2019; 12:134. [PMID: 31191245 PMCID: PMC6546926 DOI: 10.3389/fnmol.2019.00134] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 05/09/2019] [Indexed: 01/01/2023] Open
Abstract
Early life experiences program lifelong responses to stress. In agreement, resilience and vulnerability to psychopathologies, such as posttraumatic stress disorder (PTSD), have been suggested to depend on the early background. New therapies have targeted memory reconsolidation as a strategy to modify the emotional valence of traumatic memories. Here, we used animal models to study the molecular mechanism through which early experiences may later affect aversive memory reconsolidation. Handling (H)—separation of pups from dams for 10 min—or maternal separation (MS) — 3-h separation—were performed from PDN1–10, using non-handled (NH) litters as controls. Adult males were trained in a contextual fear conditioning (CFC) task; 24 h later, a short reactivation session was conducted in the conditioned or in a novel context, followed by administration of midazolam 3 mg/kg i.p. (mdz), known to disturb reconsolidation, or vehicle; a test session was performed 24 h after. The immunocontent of relevant proteins was studied 15 and 60 min after memory reactivation in the dorsal hippocampus (dHc) and basolateral amygdala complex (BLA). Mdz-treated controls (NH) showed decreased freezing to the conditioned context, consistent with reconsolidation impairment, but H and MS were resistant to labilization. Additionally, MS males showed increased freezing to the novel context, suggesting fear generalization; H rats showed lower freezing than the other groups, in accordance with previous suggestions of reduced emotionality facing adversities. Increased levels of Zif268, GluN2B, β-actin and polyubiquitination found in the BLA of all groups suggest that memory reconsolidation was triggered. In the dHc, only NH showed increased Zif268 levels after memory retrieval; also, a delay in ERK1/2 activation was found in H and MS animals. We showed here that reconsolidation of a contextual fear memory is insensitive to interference by a GABAergic drug in adult male rats exposed to different neonatal experiences; surprisingly, we found no differences in the reconsolidation process in the BLA, but the dHc appears to suffer temporal desynchronization in the engagement of reconsolidation. Our results support a hippocampal-dependent mechanism for reconsolidation resistance in models of early experiences, which aligns with current hypotheses for the etiology of PTSD.
Collapse
Affiliation(s)
- Natividade de Sá Couto-Pereira
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Programa de Pós-graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carine Lampert
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Aline Dos Santos Vieira
- Programa de Pós-graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Camilla Lazzaretti
- Programa de Pós-graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Grasielle Clotildes Kincheski
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Pablo Javier Espejo
- Instituto de Farmacología Experimental de Córdoba, Universidad Nacional de Cordoba (UNC), Cordoba, Argentina
| | - Victor Alejandro Molina
- Instituto de Farmacología Experimental de Córdoba, Universidad Nacional de Cordoba (UNC), Cordoba, Argentina
| | - Jorge Alberto Quillfeldt
- Programa de Pós-graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carla Dalmaz
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Programa de Pós-graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
75
|
Coley EJL, Demaestri C, Ganguly P, Honeycutt JA, Peterzell S, Rose N, Ahmed N, Holschbach M, Trivedi M, Brenhouse HC. Cross-Generational Transmission of Early Life Stress Effects on HPA Regulators and Bdnf Are Mediated by Sex, Lineage, and Upbringing. Front Behav Neurosci 2019; 13:101. [PMID: 31143105 PMCID: PMC6521572 DOI: 10.3389/fnbeh.2019.00101] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/24/2019] [Indexed: 01/10/2023] Open
Abstract
Early life stress (ELS) is a potent developmental disruptor and increases the risk for psychopathology. Various forms of ELS have been studied in both humans and rodents, and have been implicated in altered DNA methylation, gene transcription, stress hormone levels, and behavior. Although recent studies have focused on stress-induced epigenetic changes, the extent to which ELS alters HPA axis function and stress responsivity across generations, whether these effects are sex-specific, and how lineage interacts with upbringing to impact these effects, remain unclear. To address these points, two generations of rodents were utilized, with the first generation subjected to ELS via maternal separation, and the second to a balanced cross-fostering paradigm. We hypothesized that ELS would disrupt normative development in both generations, manifesting as altered methylation and expression of genes associated with stress signaling pathways (Nr3c1, Nr3c2, and Bdnf), blunted corticosterone (CORT), and anxiety-like behaviors. Additionally, we expected deficits in the second generation to be modulated by caretaking environment and for the pattern of results to differ between the sexes. Results suggest that direct exposure to ELS leads to sex-specific effects on gene regulation and HPA functioning in adulthood, with maternal separation leading to increases in Bdnf methylation in both sexes, decreases in Bdnf expression in females, and decreases in Nr3c1 methylation in males, as well as blunted CORT and less anxiety-like behavior in females. These alterations converged with caretaking to impart perturbations upon the subsequent generation. Across sex, ELS lineage led to decreased methylation of Nr3c1, and increased methylation of Bdnf. In fostered animals, upbringing by a previously stressed mother interacted with offspring lineage to impact methylation of Nr3c1 and Bdnf. Upbringing was also implicated in altered anxiety-like behavior in males, and baseline CORT levels in females. Such effects may correspond with observed alterations in maternal behavior across groups. In conclusion, ELS conferred enduring sex-specific alterations, both first-hand and trans-generationally via lineage and upbringing. Importantly, lineage of cross-fostered pups was sufficient to normalize or disturb maternal behavior of foster-dams, an observation requiring further elucidation. These results have implications for multi-generational effects of ELS in humans and may motivate early interventions.
Collapse
Affiliation(s)
- Elena J L Coley
- Developmental Neuropsychobiology Laboratory, Department of Psychology, Northeastern University, Boston, MA, United States
| | - Camila Demaestri
- Developmental Neuropsychobiology Laboratory, Department of Psychology, Northeastern University, Boston, MA, United States
| | - Prabarna Ganguly
- Developmental Neuropsychobiology Laboratory, Department of Psychology, Northeastern University, Boston, MA, United States
| | - Jennifer A Honeycutt
- Developmental Neuropsychobiology Laboratory, Department of Psychology, Northeastern University, Boston, MA, United States
| | - Shayna Peterzell
- Developmental Neuropsychobiology Laboratory, Department of Psychology, Northeastern University, Boston, MA, United States
| | - Natasha Rose
- Neural Metabolism and Epigenetics Laboratory, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Nida Ahmed
- Neural Metabolism and Epigenetics Laboratory, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Mary Holschbach
- Department of Behavioral Neuroscience, College of Psychology, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Malav Trivedi
- Neural Metabolism and Epigenetics Laboratory, Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, United States.,Department of Behavioral Neuroscience, College of Psychology, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Heather C Brenhouse
- Developmental Neuropsychobiology Laboratory, Department of Psychology, Northeastern University, Boston, MA, United States
| |
Collapse
|
76
|
Preventing epigenetic traces of caregiver maltreatment: A role for HDAC inhibition. Int J Dev Neurosci 2019; 78:178-184. [PMID: 31075305 DOI: 10.1016/j.ijdevneu.2019.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/24/2019] [Accepted: 05/06/2019] [Indexed: 01/07/2023] Open
Abstract
Reorganization of the brain's epigenetic landscape occurs alongside early adversity in both human and non-human animals. Whether this reorganization is simply incidental to or is a causal mechanism of the behavioral abnormalities that result from early adversity is important to understand. Using the scarcity-adversity model of low nesting resources in Long Evans rats, our lab has previously reported specific epigenetic and behavioral trajectories occurring in response to early disruption of the caregiving environment. To further probe that relationship, the current work investigates the ability of the epigenome-modifying drug sodium butyrate to prevent maltreatment-induced methylation changes when administered alongside maltreatment. Following exposure to the scarcity-adversity model, during which drug was administered prior to each caregiving session, methylation of Brain-derived Neurotrophic Factor (Bdnf) IX DNA was examined in the Prefrontal Cortex (PFC) of male and female pups at postnatal day (PN) 8. As our previous work reports, increased methylation at this exon of Bdnf in the PFC is a stable epigenetic change across the lifespan that occurs in response to early maltreatment, thus giving us a suitable starting point to investigate pharmacological prevention of maltreatment-induced epigenetic marks. Here we also examined off-target effects of sodium butyrate by assessing methylation in another region of Bdnf (exon IV) not affected in the infant brain as well as global levels of methylation in the brain region of interest. Results indicate that a 400 mg/kg (but not 300 mg/kg) dose of sodium butyrate is effective in preventing the maltreatment-induced rise in methylation at Bdnf exon IX in the PFC of male (but not female) infant pups. Administration of sodium butyrate did not affect the methylation status of Bdnf IV or overall levels of global methylation in the PFC, suggesting potential specificity of this drug. These data provide us an avenue forward for investigating whether the relationship between adversity-induced epigenetic outcomes in our model can be manipulated to improve behavioral outcomes.
Collapse
|
77
|
Dimitriadis M, van den Brink RHS, Comijs HC, Oude Voshaar RC. Prognostic effect of serum BDNF levels in late-life depression: Moderated by childhood trauma and SSRI usage? Psychoneuroendocrinology 2019; 103:276-283. [PMID: 30771710 DOI: 10.1016/j.psyneuen.2019.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) levels decline during depression and normalise after remission, although studies in older patient samples are inconsistent. Whether BDNF serum levels predict depression remission is unclear. We hypothesize that the predictive value of serum BDNF levels in late-life depression is moderated by selective serotonin reuptake inhibitors (SSRI) usage and early traumatization. METHODS Our study sample was a subset of the Netherlands Study of Depression in Older persons (NESDO), a prospective cohort study. It consisted of 267 older persons with a diagnosis of depression, for which follow-up data were available. Depression diagnosis was assessed at baseline and follow up using a structured diagnostic interview (Composite International Diagnostic Interview (CIDI), volume2.1). Logistic regression was performed (adjusted for covariates) with remission of depression after two years as the dependent variable and baseline BDNF serum levels, childhood traumatization and SSRI use as independent variables. Results - The mean age of the subjects was 70.7 years, 65.6% of them were female, their mean BDNF level was 7.7 ng/ml, 80 (30.0%) of them were traumatised in their childhood,71 (26.6%) used SSRIs and 136 (50.9%) no longer had a depressive disorder at the two year follow up. The predictive value of BDNF serum levels was conditional on traumatization and SSRI usage (threeway interaction p = .010). Higher BDNF serum levels predicted remission in traumatized depressed patients without SSRI usage (OR = 1.17, 95% C.I.: 1.00-1.36; p = .048) and in non-traumatized depressed patients who used SSRIs (OR = 1.17, 95% C.I.: 1.00-1.36; p = .052), but not in the other two subgroups. CONCLUSION The association between BDNF serum levels and the course of late-life depression seems to depend on SSRI use and childhood trauma. Based on these results, we hypothesize that childhood trauma may permanently reduce ('blunt') the responsiveness of the neurotrophic system to SSRI usage, and that this responsiveness might be more important for depression course than the actual BDNF serum levels.
Collapse
Affiliation(s)
- M Dimitriadis
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, University of Groningen PO Box 72, 9700 AB, Groningen, the Netherlands.
| | - R H S van den Brink
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, University of Groningen PO Box 72, 9700 AB, Groningen, the Netherlands
| | - H C Comijs
- GGZ in Geest / Department of Psychiatry, VU University Medical Center, The Amsterdam Public Health research institute, VU University Medical Center, De Boelelaan 1105, 1081 HV, Amsterdam, the Netherlands
| | - R C Oude Voshaar
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, University of Groningen PO Box 72, 9700 AB, Groningen, the Netherlands
| |
Collapse
|
78
|
Yang Y, Yang S, Liu J, Feng Y, Qi F, Zhao R. DNA Hypomethylation of GR Promoters is Associated with GR Activation and BDNF/AKT/ERK1/2-Induced Hippocampal Neurogenesis in Mice Derived From Folic-Acid-Supplemented Dams. Mol Nutr Food Res 2019; 63:e1801334. [PMID: 30920123 DOI: 10.1002/mnfr.201801334] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/20/2019] [Indexed: 12/20/2022]
Abstract
SCOPE Glucocorticoid receptor (GR) mediates the nutritional programing of offspring performance. Maternal folic acid has been shown to regulate hippocampal neurogenesis and affect cognitive function in offspring, yet it remains unclear whether and how GR is involved in such effects. METHODS AND RESULTS Adult male mice derived from dams fed basal or folic-acid-supplemented diet (5 mg folic acid/kg) throughout gestation and lactation are used in this study. Maternal folic acid significantly enhances offspring learning and memory with less fear-related behavior. Concurrently, hippocampal neurogenesis is improved with upregulation of brain-derived neurotrophic factor and its downstream AKT/ERK1/2 signaling pathway. More GR immune-positive cells are observed in hippocampus of folic acid group, which are in line with higher GR protein and mRNA abundances. Differential expression of GR exon 1 transcript variants is detected, which is inversely associated with modified DNA methylation on their alternate promoters. CONCLUSION The results indicate that maternal folic acid supplementation promotes hippocampal neurogenesis and improves learning and memory behavior in mouse offspring. The mechanisms involve modification of DNA methylation on GR alternate promoters and GR upregulation in the hippocampus, which is associated with activation of BDNF/AKT/ERK1/2 signaling.
Collapse
Affiliation(s)
- Yang Yang
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, 210095, P. R. China.,Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Shu Yang
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, 210095, P. R. China.,Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Jie Liu
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, 210095, P. R. China.,Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Yue Feng
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, 210095, P. R. China.,Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Fulei Qi
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, 210095, P. R. China.,Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, 210095, P. R. China.,Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| |
Collapse
|
79
|
Umeoka EHL, Robinson EJ, Turimella SL, van Campen JS, Motta-Teixeira LC, Sarabdjitsingh RA, Garcia-Cairasco N, Braun K, de Graan PN, Joëls M. Hyperthermia-induced seizures followed by repetitive stress are associated with age-dependent changes in specific aspects of the mouse stress system. J Neuroendocrinol 2019; 31:e12697. [PMID: 30773738 DOI: 10.1111/jne.12697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/04/2019] [Accepted: 02/13/2019] [Indexed: 11/28/2022]
Abstract
Stress is among the most frequently self-reported factors provoking epileptic seizures in children and adults. It is still unclear, however, why some people display stress-sensitive seizures and others do not. Recently, we showed that young epilepsy patients with stress-sensitive seizures exhibit a dysregulated hypothalamic-pituitary-adrenal (HPA)-axis. Most likely, this dysregulation gradually develops, and is triggered by stressors occurring early in life (early-life stress [ELS]). ELS may be particularly impactful when overlapping with the period of epileptogenesis. To examine this in a controlled and prospective manner, the present study investigated the effect of repetitive variable stressors or control treatment between postnatal day (PND) 12 and 24 in male mice exposed on PND10 to hyperthermia (HT)-induced prolonged seizures (control: normothermia). A number of peripheral and central indices of HPA-axis activity were evaluated at pre-adolescent and young adult age (ie, at PND25 and 90, respectively). At PND25 but not at PND90, body weight gain and absolute as well as relative (to body weight) thymus weight were reduced by ELS (vs control), whereas relative adrenal weight was enhanced, confirming the effectiveness of the stress treatment. Basal and stress-induced corticosterone levels were unaffected, though, by ELS at both ages. HT by itself did not affect any of these peripheral markers of HPA-axis activity, nor did it interact with ELS. However, centrally we did observe age-specific interaction effects of HT and ELS with regard to hippocampal glucocorticoid receptor mRNA expression, neurogenesis with the immature neurone marker doublecortin and the number of hilar (ectopic) granule cells using Prox1 staining. This lends some support to the notion that exposure to repetitive stress after HT-induced seizures may dysregulate central components of the stress system in an age-dependent manner. Such dysregulation could be one of the mechanisms conferring higher vulnerability of individuals with epilepsy to develop seizures in the face of stress.
Collapse
Affiliation(s)
- Eduardo H L Umeoka
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Edward J Robinson
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Sada Lakshmi Turimella
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Jolien S van Campen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lívia C Motta-Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - R Angela Sarabdjitsingh
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Norberto Garcia-Cairasco
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kees Braun
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pierre N de Graan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Marian Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
80
|
Bolton JL, Short AK, Simeone KA, Daglian J, Baram TZ. Programming of Stress-Sensitive Neurons and Circuits by Early-Life Experiences. Front Behav Neurosci 2019; 13:30. [PMID: 30833892 PMCID: PMC6387907 DOI: 10.3389/fnbeh.2019.00030] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/04/2019] [Indexed: 12/22/2022] Open
Abstract
Early-life experiences influence brain structure and function long-term, contributing to resilience or vulnerability to stress and stress-related disorders. Therefore, understanding the mechanisms by which early-life experiences program specific brain cells and circuits to shape life-long cognitive and emotional functions is crucial. We identify the population of corticotropin-releasing hormone (CRH)-expressing neurons in the hypothalamic paraventricular nucleus (PVN) as a key, early target of early-life experiences. Adverse experiences increase excitatory neurotransmission onto PVN CRH cells, whereas optimal experiences, such as augmented and predictable maternal care, reduce the number and function of glutamatergic inputs onto this cell population. Altered synaptic neurotransmission is sufficient to initiate large-scale, enduring epigenetic re-programming within CRH-expressing neurons, associated with stress resilience and additional cognitive and emotional outcomes. Thus, the mechanisms by which early-life experiences influence the brain provide tractable targets for intervention.
Collapse
Affiliation(s)
- Jessica L Bolton
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Annabel Katherine Short
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Kristina A Simeone
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Jennifer Daglian
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Tallie Z Baram
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
81
|
Changes in neuroplasticity following early-life social adversities: the possible role of brain-derived neurotrophic factor. Pediatr Res 2019; 85:225-233. [PMID: 30341412 DOI: 10.1038/s41390-018-0205-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023]
Abstract
Social adversities experienced in childhood can have a profound impact on the developing brain, leading to the emergence of psychopathologies in adulthood. Despite the burden this places on both the individual and society, the neurobiological aspects mediating this transition remain unclear. Recent advances in preclinical and clinical research have begun examining neuroplasticity-the nervous system's ability to form adaptive changes in response to new experience-in the context of early-life vulnerability to social adversities and plasticity-related alterations following such traumatic events. A key mediator of plasticity-related molecular processes is the brain-derived neurotrophic factor (BDNF), which has also been implicated in various psychiatric disorders related to childhood social adversities. Preclinical and clinical data suggest early-life social adversities (ELSA) might be associated with accelerated maturation of social network circuitry, a possible ontogenic adaptation to the adverse environment. Neural plasticity decreases by adulthood, lessening the efficacy of treatment in ELSA-related psychiatric disorders. However, literature data suggest that by increasing BDNF/TrkB signalling through antidepressant treatment a juvenile-like plasticity state can be induced, which allows for reorganization of the social circuitry when guided by psychotherapy and surrounded by a safe and positive environment.
Collapse
|
82
|
Veras AB, Chao MV, Getz M, Goetz R, Cheniaux E, Lopes FL, Nardi AE, Walsh-Messinger J, Malaspina D, Kranz TM. Traumatic experiences and cognitive profiles of schizophrenia cases influenced by the BDNF Val66met polymorphism. Psychiatry Res 2019; 271:111-113. [PMID: 30472504 DOI: 10.1016/j.psychres.2018.11.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 10/27/2022]
Abstract
The association of early trauma exposure with current cognition was examined in a research series of 56 schizophrenia cases with respect to the BDNF Val66Met polymorphism (rs6265, Val66Val, Val66Met, Met66Met), as met allele carriers have reduced neurotrophic activity. The Perceptual Organization Index had a significant negative correlation with trauma exposures only in met carriers, including early physical abuse, general trauma after age 18 years, and physical abuse. Within the Val66Val subgroup, there were no significant correlations between WAIS indices and traumatic experiences.
Collapse
Affiliation(s)
- André B Veras
- Translational Research Group on Mental Health (GPTranSMe), Dom Bosco Catholic University, Campo Grande, Brazil; Institute of Psychiatry, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil; Departments of Psychiatry, Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mt. Sinai Medical Center, New York, NY, USA.
| | - Moses V Chao
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology, Physiology & Neuroscience and Psychiatry, New York University, New York, NY, USA; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Germany
| | - Mara Getz
- Columbia University Mailman School of Public Health, New York, NY, USA
| | - Raymond Goetz
- Department of Psychiatry, Columbia University, New York, NY USA
| | - Elie Cheniaux
- Institute of Psychiatry, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil; State University of Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | | | - Antonio E Nardi
- Translational Research Group on Mental Health (GPTranSMe), Dom Bosco Catholic University, Campo Grande, Brazil
| | | | - Dolores Malaspina
- Departments of Psychiatry, Neuroscience, Genetics and Genomics, Icahn School of Medicine at Mt. Sinai Medical Center, New York, NY, USA
| | - Thorsten M Kranz
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology, Physiology & Neuroscience and Psychiatry, New York University, New York, NY, USA; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Germany
| |
Collapse
|
83
|
Chen H, Amazit L, Lombès M, Le Menuet D. Crosstalk Between Glucocorticoid Receptor and Early-growth Response Protein 1 Accounts for Repression of Brain-derived Neurotrophic Factor Transcript 4 Expression. Neuroscience 2018; 399:12-27. [PMID: 30578973 DOI: 10.1016/j.neuroscience.2018.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/31/2022]
Abstract
The brain-derived neurotrophic factor (BDNF) is a key player in brain functions such as synaptic plasticity, stress, and behavior. Its gene structure in rodents contains 8 untranslated exons (I to VIII) whose expression is finely regulated and which spliced onto a common and unique translated exon IX. Altered Bdnf expression is associated with many pathologies such as depression, Alzheimer's disease and addiction. Through binding to glucocorticoid receptor (GR), glucocorticoids play a pivotal role for stress responses, mood and neuronal plasticity. We recently showed in neuronal primary culture and in the immortalized neuronal-like BZ cells that GR repressed Bdnf expression, notably the bdnf exon IV containing mRNA isoform (Bdnf4) via GR binding to a short 275-bp sequence of Bdnf promoter. Herein, we demonstrate by transient transfection experiments and mutagenesis in BZ cells that GR interacts with an early growth response protein 1 (EGR1) response element (EGR-RE) located in the transcription start site of Bdnf exon IV promoter. Using Chromatin Immunoprecipitation, we find that both GR and EGR1 bind to this promoter sequence in a glucocorticoid-dependent manner and demonstrate by co-immunoprecipitation that GR and EGR1 are interacting physically. Interestingly, EGR1 has been widely characterized as a regulator of brain plasticity. In conclusion, we deciphered a mechanism by which GR downregulates Bdnf expression, identifying a novel functional crosstalk between glucocorticoid pathways, immediate early growth response proteins and Bdnf. As all these factors are well-recognized germane for brain pathophysiology, these findings may have significant implications in neurosciences as well as in therapeutics.
Collapse
Affiliation(s)
- Hui Chen
- Inserm 1185, Fac Med Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Larbi Amazit
- UMS-32, Institut Biomédical de Bicêtre, Le Kremlin-Bicêtre, France
| | - Marc Lombès
- Inserm 1185, Fac Med Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Le Kremlin Bicêtre F-94275, France
| | - Damien Le Menuet
- Inserm 1185, Fac Med Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
84
|
Lemche E. Early Life Stress and Epigenetics in Late-onset Alzheimer's Dementia: A Systematic Review. Curr Genomics 2018; 19:522-602. [PMID: 30386171 PMCID: PMC6194433 DOI: 10.2174/1389202919666171229145156] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 07/27/2017] [Accepted: 12/12/2017] [Indexed: 11/22/2022] Open
Abstract
Involvement of life stress in Late-Onset Alzheimer's Disease (LOAD) has been evinced in longitudinal cohort epidemiological studies, and endocrinologic evidence suggests involvements of catecholamine and corticosteroid systems in LOAD. Early Life Stress (ELS) rodent models have successfully demonstrated sequelae of maternal separation resulting in LOAD-analogous pathology, thereby supporting a role of insulin receptor signalling pertaining to GSK-3beta facilitated tau hyper-phosphorylation and amyloidogenic processing. Discussed are relevant ELS studies, and findings from three mitogen-activated protein kinase pathways (JNK/SAPK pathway, ERK pathway, p38/MAPK pathway) relevant for mediating environmental stresses. Further considered were the roles of autophagy impairment, neuroinflammation, and brain insulin resistance. For the meta-analytic evaluation, 224 candidate gene loci were extracted from reviews of animal studies of LOAD pathophysiological mechanisms, of which 60 had no positive results in human LOAD association studies. These loci were combined with 89 gene loci confirmed as LOAD risk genes in previous GWAS and WES. Of the 313 risk gene loci evaluated, there were 35 human reports on epigenomic modifications in terms of methylation or histone acetylation. 64 microRNA gene regulation mechanisms were published for the compiled loci. Genomic association studies support close relations of both noradrenergic and glucocorticoid systems with LOAD. For HPA involvement, a CRHR1 haplotype with MAPT was described, but further association of only HSD11B1 with LOAD found; however, association of FKBP1 and NC3R1 polymorphisms was documented in support of stress influence to LOAD. In the brain insulin system, IGF2R, INSR, INSRR, and plasticity regulator ARC, were associated with LOAD. Pertaining to compromised myelin stability in LOAD, relevant associations were found for BIN1, RELN, SORL1, SORCS1, CNP, MAG, and MOG. Regarding epigenetic modifications, both methylation variability and de-acetylation were reported for LOAD. The majority of up-to-date epigenomic findings include reported modifications in the well-known LOAD core pathology loci MAPT, BACE1, APP (with FOS, EGR1), PSEN1, PSEN2, and highlight a central role of BDNF. Pertaining to ELS, relevant loci are FKBP5, EGR1, GSK3B; critical roles of inflammation are indicated by CRP, TNFA, NFKB1 modifications; for cholesterol biosynthesis, DHCR24; for myelin stability BIN1, SORL1, CNP; pertaining to (epi)genetic mechanisms, hTERT, MBD2, DNMT1, MTHFR2. Findings on gene regulation were accumulated for BACE1, MAPK signalling, TLR4, BDNF, insulin signalling, with most reports for miR-132 and miR-27. Unclear in epigenomic studies remains the role of noradrenergic signalling, previously demonstrated by neuropathological findings of childhood nucleus caeruleus degeneration for LOAD tauopathy.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| |
Collapse
|
85
|
Thomas M, Knoblich N, Wallisch A, Glowacz K, Becker-Sadzio J, Gundel F, Brückmann C, Nieratschker V. Increased BDNF methylation in saliva, but not blood, of patients with borderline personality disorder. Clin Epigenetics 2018; 10:109. [PMID: 30134995 PMCID: PMC6106893 DOI: 10.1186/s13148-018-0544-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022] Open
Abstract
Background The importance of epigenetic alterations in psychiatric disorders is increasingly acknowledged and the use of DNA methylation patterns as markers of disease is a topic of ongoing investigation. Recent studies suggest that patients suffering from Borderline Personality Disorder (BPD) display differential DNA methylation of various genes relevant for neuropsychiatric conditions. For example, several studies report differential methylation in the promoter region of the brain-derived neurotrophic factor gene (BDNF) in blood. However, little is known about BDNF methylation in other tissues. Results In the present study, we analyzed DNA methylation of the BDNF IV promoter in saliva and blood of 41 BPD patients and 41 matched healthy controls and found significant hypermethylation in the BPD patient’s saliva, but not blood. Further, we report that BDNF methylation in saliva of BPD patients significantly decreased after a 12-week psychotherapeutic intervention. Conclusions Providing a direct comparison of BDNF methylation in blood and saliva of the same individuals, our results demonstrate the importance of choice of tissue for the study of DNA methylation. In addition, they indicate a better suitability of saliva for the study of differential BDNF methylation in BPD patients. Further, our data appear to indicate a reversal of disease-specific alterations in BDNF methylation in response to psychotherapy, though further experiments are necessary to validate these results and determine the specificity of the effect. Electronic supplementary material The online version of this article (10.1186/s13148-018-0544-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mara Thomas
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany.,Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Nora Knoblich
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Annalena Wallisch
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Katarzyna Glowacz
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Julia Becker-Sadzio
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Friederike Gundel
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Christof Brückmann
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Vanessa Nieratschker
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany.
| |
Collapse
|
86
|
Shepard RD, Gouty S, Kassis H, Berenji A, Zhu W, Cox BM, Nugent FS. Targeting histone deacetylation for recovery of maternal deprivation-induced changes in BDNF and AKAP150 expression in the VTA. Exp Neurol 2018; 309:160-168. [PMID: 30102916 DOI: 10.1016/j.expneurol.2018.08.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/17/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022]
Abstract
Severe early life stressors increase the probability of developing psychiatric disorders later in life through modifications in neuronal circuits controlling brain monoaminergic signaling. Our previous work demonstrated that 24 h maternal deprivation (MD) in male Sprague Dawley rats modifies dopamine (DA) signaling from the ventral tegmental area (VTA) through changes at GABAergic synapses that were reversible by in vitro histone deacetylase (HDAC) inhibition which led to restoration of the scaffold A-kinase anchoring protein (AKAP150) signaling and subsequently recovered GABAergic plasticity (Authement et al., 2015). Using a combination of in situ hybridization, Western blots and immunohistochemistry, we confirmed that MD-induced epigenetic modifications at the level of histone acetylation were associated with an upregulation of HDAC2. MD also increased Akap5 mRNA levels in the VTA. Western blot analysis of AKAP150 protein expression showed an increase in synaptic levels of AKAP150 protein in the VTA with an accompanying decrease in synaptic levels of protein kinase A (PKA). Moreover, the abundance of mature brain-derived neurotrophic factor (BDNF) protein of VTA tissues from MD rats was significantly lower than in control groups. In vivo systemic injection with a selective class I HDAC inhibitor (CI-994) was sufficient to reverse MD-induced histone hypoacetylation in the VTA for 24 h after the injection. Furthermore, HDAC inhibition normalized the levels of mBDNF and AKAP150 proteins at 24 h. Our data suggest that HDAC-mediated targeting of BDNF and AKAP-dependent local signaling within VTA could provide novel therapeutics for prevention of later-life psychopathology.
Collapse
Affiliation(s)
- Ryan D Shepard
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Shawn Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Haifa Kassis
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Aylar Berenji
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - William Zhu
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Brian M Cox
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Fereshteh S Nugent
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA.
| |
Collapse
|
87
|
Swart PC, Russell VA, Dimatelis JJ. Maternal separation stress reduced prenatal-ethanol-induced increase in exploratory behaviour and extracellular signal-regulated kinase activity. Behav Brain Res 2018; 356:470-482. [PMID: 29908221 DOI: 10.1016/j.bbr.2018.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/22/2018] [Accepted: 06/06/2018] [Indexed: 01/26/2023]
Abstract
In an attempt to better represent the aetiology of fetal alcohol spectrum disorder (FASD) and the associated psychological deficits, prenatal-ethanol exposure was followed by maternal separation in a rat model in order to account for the effects of early-life adversities in addition to in utero alcohol exposure. Extracellular signal-regulated kinase 1/2 (ERK1/2) and glycogen synthase kinase 3-β (GSK3β) are converging points for many signalling cascades and have been implicated in models of FASD and models of early-life stress. Therefore, these kinases may also contribute to the behavioural changes observed after the combination of both developmental insults. In this study, ethanol-dams voluntarily consumed a 0.066% saccharin-sweetened 10% ethanol (EtOH) solution for 10 days prior to pregnancy and throughout gestation while control-dams had ad libitumaccess to a 0.066% saccharin (sacc) solution. Whole litters were randomly assigned to undergo maternal separation (MS) for 3 h/day from P2 to P14 while the remaining litters were left undisturbed (nMS). This resulted in 4 experimental groups: control (sacc + nMS), MS (sacc + MS), EtOH (EtOH + nMS) and EtOH + MS. Throughout development, EtOH-rats weighed less than control rats. However, subsequent maternal separation stress caused EtOH + MS-rats to weigh more than EtOH-rats. In adulthood both MS- and EtOH-rats were hyperactive but the combination produced activity levels similar to that of control rats. All treated animals (MS-, EtOH- and EtOH + MS-rats) demonstrated a negative affective state shown by increased number and duration of 22 kHz ultrasonic vocalizations compared to control rats. Prenatal-ethanol exposure increased the P-GSK3β/GSK3β ratio in the prefrontal cortex (PFC) and maternal separation decreased the P-GSK3β/GSK3β ratio in the dorsal hippocampus (DH) of adult rats. However, maternal separation stress decreased the effect of prenatal-ethanol exposure on the P-ERK/ERK ratio in the PFC and DH and reduced prenatal-ethanol-induced hyperactivity. Therefore, indicating a significant interaction between prenatal-ethanol exposure and early-life stress on behaviour and the brain and may implicate P-ERK1/2 signalling in exploratory behaviour.
Collapse
Affiliation(s)
- Patricia C Swart
- University of Cape Town, Faculty of Health Sciences, Department of Human Biology, Observatory, 7925 South Africa.
| | - Vivienne A Russell
- University of Cape Town, Faculty of Health Sciences, Department of Human Biology, Observatory, 7925 South Africa.
| | - Jacqueline J Dimatelis
- University of Cape Town, Faculty of Health Sciences, Department of Human Biology, Observatory, 7925 South Africa.
| |
Collapse
|
88
|
Hacimusalar Y, Eşel E. Suggested Biomarkers for Major Depressive Disorder. ACTA ACUST UNITED AC 2018; 55:280-290. [PMID: 30224877 DOI: 10.5152/npa.2017.19482] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/08/2017] [Indexed: 12/21/2022]
Abstract
Currently, the diagnosis of major depressive disorder (MDD) mainly relies on clinical examination and subjective evaluation of depressive symptoms. There is no non-invasive, quantitative test available today for the diagnosis of MDD. In MDD, exploration of biomarkers will be helpful in diagnosing the disorder as well as in choosing a treatment, and predicting the treatment response. In this article, it is aimed to review the findings of suggested biomarkers such as growth factors, cytokines and other inflammatory markers, oxidative stress markers, endocrine markers, energy balance hormones, genetic and epigenetic features, and neuroimaging in MDD and to evaluate how these findings contribute to the pathophysiology of MDD, the prediction of treatment response, severity of the disorder, and identification of subtypes. Among these, the findings related to the brain-derived neurotrophic factor, the hypothalamo-pituitary-adrenal axis, cytokines, and neuroimaging may be strong candidates for being biomarkers MDD, and may provide critical information in understanding biological etiology of depression. Although the findings are not sufficient yet, we think that the results of epigenetic studies will also provide very important contributions to the biomarker research in MDD. The availability of biomarkers in MDD will be an advancement that will facilitate the diagnosis of the disorder, treatment choices in the early stages, and prediction of the course of the disorder.
Collapse
Affiliation(s)
- Yunus Hacimusalar
- Department of Psychiatry, Kayseri Training and Research Hospital, Kayseri, Turkey
| | - Ertuğrul Eşel
- Department of Psychiatry, Erciyes University Faculty of Medicine, Kayseri, Turkey
| |
Collapse
|
89
|
Godoy LD, Umeoka EHL, Ribeiro DE, Santos VR, Antunes-Rodrigues J, Joca SRL, Garcia-Cairasco N. Multimodal early-life stress induces biological changes associated to psychopathologies. Horm Behav 2018; 100:69-80. [PMID: 29548783 DOI: 10.1016/j.yhbeh.2018.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/07/2018] [Accepted: 03/11/2018] [Indexed: 01/21/2023]
Abstract
Evidences suggest the contributive role of early-life stress (ELS) to affective and anxiety disorders. Chronic exposure to the same stressor may generate habituation, while the exposure to different and repeated stressors gradually promotes maladaptive plasticity. Therefore, to further understand the effects of heterotypic stressors during early life period, male Wistar rat pups (P1-P21) were exposed to Multimodal ELS paradigm. Results indicate pups did not habituate to multimodal ELS and neonates respond to both physical and psychogenic stressors. Adult rats that underwent ELS protocol showed significant lower sucrose intake, decreased latency to immobility in the forced swim test and increased latency to light compartment in the light-dark test when compared to control group. Although it has been shown that ELS-induced changes in hippocampus can be used as biomarkers, multimodal ELS did not significantly alter BDNF, Tyrosine Kinase B (TrkB) receptor expression or neurogenesis in the hippocampus. Taken together, these findings indicate that multimodal ELS protocol can be an interesting experimental model for understanding long-term psychiatric disorders associated with stress. Indeed, our data with neurogenesis, BDNF and TrkB, and conflicting data from the literature, suggest that additional studies on synaptic plasticity/intracellular cascades would help to detect the underlying mechanisms.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Eduardo H L Umeoka
- Neurosciences and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Deidiane Elisa Ribeiro
- Pharmacology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | | | - José Antunes-Rodrigues
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Samia Regiane Lourenço Joca
- Physics and Chemistry Department, Ribeirão Preto School of Pharmacy, University of São Paulo, Brazil; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Norberto Garcia-Cairasco
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil; Neurosciences and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil.
| |
Collapse
|
90
|
Wang T, Shi C, Li X, Zhang P, Liu B, Wang H, Wang Y, Yang Y, Wu Y, Li H, Xu ZQD. Injection of oxytocin into paraventricular nucleus reverses depressive-like behaviors in the postpartum depression rat model. Behav Brain Res 2018; 336:236-243. [DOI: 10.1016/j.bbr.2017.09.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/28/2017] [Accepted: 09/04/2017] [Indexed: 01/07/2023]
|
91
|
Xie H, Claycomb Erwin M, Elhai JD, Wall JT, Tamburrino MB, Brickman KR, Kaminski B, McLean SA, Liberzon I, Wang X. Relationship of Hippocampal Volumes and Posttraumatic Stress Disorder Symptoms Over Early Posttrauma Periods. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2017; 3:968-975. [PMID: 30409391 DOI: 10.1016/j.bpsc.2017.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 11/22/2017] [Accepted: 11/22/2017] [Indexed: 11/20/2022]
Abstract
BACKGROUND Smaller hippocampal volume is associated with more severe posttraumatic stress disorder (PTSD) symptoms years after traumatic experiences. Posttraumatic stress symptoms appear early following trauma, but the relationship between hippocampal volume and PTSD symptom severity during early posttrauma periods is not well understood. It is possible that the inverse relationship between hippocampal volume and PTSD symptom severity is already present soon after trauma. To test this possibility, we prospectively examined the association between hippocampal volumes and severity of PTSD symptoms within weeks to months after trauma due to a motor vehicle collision. METHODS Structural magnetic resonance imaging scans of 44 survivors were collected about 2 weeks and again at 3 months after a motor vehicle collision to measure hippocampal volumes. The PTSD Checklist was used to evaluate PTSD symptoms at each scan time. Full (n = 5) or partial (n = 6) PTSD was evaluated using the Clinician-Administered PTSD Scale at 3 months. RESULTS Left hippocampal volumes at both time points negatively correlated with PTSD Checklist scores, and with subscores for re-experiencing symptoms at 3 months. Left hippocampal volumes at 3 months also negatively correlated with hyperarousal symptoms at 3 months. Finally, neither left nor right hippocampal volumes significantly changed between 2 weeks and 3 months posttrauma. CONCLUSIONS The results suggest that small hippocampal volume at early posttrauma weeks is associated with increased risk for PTSD development. Furthermore, the inverse relationship between hippocampal volume and PTSD symptoms at 3 months did not arise from posttrauma shifts in hippocampal volume between 2 weeks and 3 months after trauma.
Collapse
Affiliation(s)
- Hong Xie
- Department of Neurosciences, University of Toledo, Toledo, Ohio
| | | | - Jon D Elhai
- Department of Psychology, University of Toledo, Toledo, Ohio
| | - John T Wall
- Department of Neurosciences, University of Toledo, Toledo, Ohio
| | | | | | - Brian Kaminski
- Department of Emergency Medicine, ProMedica Toledo Hospital, Toledo, Ohio
| | - Samuel A McLean
- Department of Anesthesiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Israel Liberzon
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan
| | - Xin Wang
- Department of Psychiatry, University of Toledo, Toledo, Ohio.
| |
Collapse
|
92
|
Wang SS, Mu RH, Li CF, Dong SQ, Geng D, Liu Q, Yi LT. microRNA-124 targets glucocorticoid receptor and is involved in depression-like behaviors. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:417-425. [PMID: 28764913 DOI: 10.1016/j.pnpbp.2017.07.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/15/2017] [Accepted: 07/28/2017] [Indexed: 11/18/2022]
Abstract
Dysregulation of microRNA (miRNA) has been shown to be involved in early observations of depression. MicroRNA-124-3p (miR-124) is the most abundant microRNA in the brain. Previous studies have shown that miR-124 plays a major role in depression. Here we showed that miR-124 directly targeted glucocorticoid receptor (GR) in HEK 293 cells. In addition, inhibition of miR-124 by its antagomir (2nmol/every two days) could reverse the decrease of sucrose preference and the increase of immobility time in mice exposed to chronic corticosterone (CORT, 40mg/kg) injection. Moreover, these effects on behavioral improvement were coupled to the activation of brain-derived neurotrophic factor (BDNF), TrkB, ERK, and CREB, as well as the induction of synaptogenesis and neuronal proliferation. Altogether, our study suggests that miR-124 can be served as a biomarker for depression and a novel target for drug development, and demonstrates that inhibition of miR-124 may be a strategy for treating depression by activating BDNF-TrkB signaling pathway in the hippocampus.
Collapse
Affiliation(s)
- Shuang-Shuang Wang
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China; Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
| | - Rong-Hao Mu
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China; Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
| | - Cheng-Fu Li
- Xiamen Hospital of Traditional Chinese Medicine, Xiamen 361009, Fujian Province, PR China
| | - Shu-Qi Dong
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China; Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
| | - Di Geng
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China; Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, Fujian Province, PR China; Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
| | - Qing Liu
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China; Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, Fujian Province, PR China; Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
| | - Li-Tao Yi
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China; Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, Fujian Province, PR China; Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
| |
Collapse
|
93
|
do Prado CH, Grassi-Oliveira R, Daruy-Filho L, Wieck A, Bauer ME. Evidence for Immune Activation and Resistance to Glucocorticoids Following Childhood Maltreatment in Adolescents Without Psychopathology. Neuropsychopharmacology 2017; 42:2272-2282. [PMID: 28664925 PMCID: PMC5603807 DOI: 10.1038/npp.2017.137] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 06/22/2017] [Accepted: 06/24/2017] [Indexed: 02/07/2023]
Abstract
Early-life stress (ELS) increases the risk for psychopathology. Immune and endocrine changes have been reported in adults and are associated with maladaptation of stress-responsive systems. Here we investigated the effects of ELS on endocrine and immune pathways in adolescents without psychopathology. Thirty adolescents with a history of childhood maltreatment and 27 adolescents without ELS history were recruited. Blood and hair samples were obtained from all participants. Lymphocytes were isolated and stimulated in vitro. Flow cytometry was used to evaluate lymphocyte subsets, Th1/Th2/Th17 cytokines, mitogen-activated protein kinase (MAPK), and nuclear factor kappa B (NF-κB) signaling pathways, as well as lymphocyte sensitivity to dexamethasone. Brain-derived neurotrophic factor (BDNF) and hair cortisol were assessed with enzyme-linked immunosorbent assays (ELISAs). Adolescents with a history of ELS had increased percentages of T-cell activation markers (CD3+CD4+CD25+ and CD3+CD69+) and senescent T cells (CD8+CD28- and CD4+CD28-), as well as decreased percentages of NK (CD3-CD56+) and NK T cells (CD3+CD56+). Following stimulation, lymphocytes of ELS+ adolescents produced significantly more IL-2, IL-4, IFN-γ, and IL-17 and engaged more MAPK ERK and NF-κB signaling. ELS was associated with increased hair cortisol levels in parallel with increased lymphocyte resistance to dexamethasone and low plasma BDNF levels. These data provide the first indication of the presence of immune activation and pro-inflammatory profiles in healthy adolescents exposed to ELS, which could contribute to increased vulnerability of trauma-related psychopathology later in life. The underlying mechanisms of this impairment may include the enhanced activation of both MAPK and NF-κB signaling in parallel to partial resistance to glucocorticoids.
Collapse
Affiliation(s)
- Carine Hartmann do Prado
- Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rodrigo Grassi-Oliveira
- Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil,Developmental Cognitive Neuroscience Lab (DCNL), Brain Institute of Rio Grande do Sul (InsCer), PUCRS, Porto Alegre, Brazil
| | - Ledo Daruy-Filho
- Developmental Cognitive Neuroscience Lab (DCNL), Brain Institute of Rio Grande do Sul (InsCer), PUCRS, Porto Alegre, Brazil
| | - Andréa Wieck
- Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Moisés Evandro Bauer
- Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil,Faculty of Biosciences, PUCRS, Porto Alegre, Brazil,Instituto de Pesquisas Biomédicas, Hospital São Lucas da PUCRS, Av. Ipiranga 6690, 2° andar. P.O. Box 1429. Porto Alegre, RS 90.610-000, Brazil, Tel: +55 51 33203000, E-mail:
| |
Collapse
|
94
|
Kertes DA, Bhatt SS, Kamin HS, Hughes DA, Rodney NC, Mulligan CJ. BNDF methylation in mothers and newborns is associated with maternal exposure to war trauma. Clin Epigenetics 2017; 9:68. [PMID: 28680507 PMCID: PMC5493129 DOI: 10.1186/s13148-017-0367-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 06/14/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The BDNF gene codes for brain-derived neurotrophic factor, a growth factor involved in neural development, cell differentiation, and synaptic plasticity. Present in both the brain and periphery, BDNF plays critical roles throughout the body and is essential for placental and fetal development. Rodent studies show that early life stress, including prenatal stress, broadly alters BDNF methylation, with presumed changes in gene expression. No studies have assessed prenatal exposure to maternal traumatic stress and BDNF methylation in humans. This study examined associations of prenatal exposure to maternal stress and BDNF methylation at CpG sites across the BDNF gene. RESULTS Among 24 mothers and newborns in the eastern Democratic Republic of Congo, a region with extreme conflict and violence to women, maternal experiences of war trauma and chronic stress were associated with BDNF methylation in umbilical cord blood, placental tissue, and maternal venous blood. Associations of maternal stress and BDNF methylation showed high tissue specificity. The majority of significant associations were observed in putative transcription factor binding regions. CONCLUSIONS This is the first study in humans to examine BDNF methylation in relation to prenatal exposure to maternal stress in three tissues simultaneously and the first in any mammalian species to report associations of prenatal stress and BDNF methylation in placental tissue. The findings add to the growing body of evidence highlighting the importance of considering epigenetic effects when examining the impacts of trauma and stress, not only for adults but also for offspring exposed via effects transmitted before birth.
Collapse
Affiliation(s)
- Darlene A Kertes
- Department of Psychology and University of Florida Genetics Institute, 945 Center Drive, Gainesville, FL 32611-2250 USA
| | - Samarth S Bhatt
- Department of Psychology, University of Florida, Gainesville, FL USA
| | - Hayley S Kamin
- Department of Psychology, University of Florida, Gainesville, FL USA
| | - David A Hughes
- Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Nicole C Rodney
- Department of Anthropology, University of Florida, Gainesville, FL USA
| | - Connie J Mulligan
- Department of Anthropology and University of Florida Genetics Institute, University of Florida, Gainesville, FL USA
| |
Collapse
|
95
|
Influence of catch up growth on spatial learning and memory in a mouse model of intrauterine growth restriction. PLoS One 2017; 12:e0177468. [PMID: 28542302 PMCID: PMC5443512 DOI: 10.1371/journal.pone.0177468] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 04/27/2017] [Indexed: 01/21/2023] Open
Abstract
Background Intrauterine growth restriction (IUGR) and rapid postnatal weight gain or catch up growth (CUG) increase the susceptibility to metabolic syndrome during adult life. Longitudinal studies have also revealed a high incidence of learning difficulties in children with IUGR. The aim of the present study was to investigate the effect of nutrition and CUG on learning memory in an IUGR animal model. We hypothesized that synaptic protein expression and transcription, an essential mechanism for memory consolidation, might be affected by intrauterine undernutrition. Methods IUGR was induced by 50% maternal caloric undernutrition throughout late gestation. During the suckling period, dams were either fed ad libitum or food restricted. The pups were divided into: Normal prenatal diet-Normal postnatal diet (NN), Restricted prenatal diet- Normal postnatal diet + catch up growth (RN+), Normal prenatal diet-Restricted postnatal diet (NR) and Restricted prenatal diet-Restricted postnatal diet (RR). At 4 weeks of age, memory was assessed via a water maze test. To evaluate synaptic function, 2 specific synaptic proteins (postsynaptic density-95 [PSD95], synaptophysin) as well as insulin receptors (IR) were tested by Western Blot and quantitative polymerase chain reaction (qPCR). Brain-derived neurotrophic factor and serum insulin levels were also studied. Results and conclusions The RN+ group presented a learning curve similar to the NN animals. The RR animals without CUG showed learning disabilities. PSD95 was lower in the RR group than in the NN and RN+ mice. In contrast, synaptophysin was similar in all groups. IR showed an inverse expression pattern to that of the PSD95. In conclusion, perinatal nutrition plays an important role in learning. CUG after a period of prenatal malnutrition seems to improve learning skills. The functional alterations observed might be related to lower PSD95 activity and a possible dysfunction in the hormone regulation of synaptic plasticity.
Collapse
|
96
|
Cross D, Fani N, Powers A, Bradley B. Neurobiological Development in the Context of Childhood Trauma. ACTA ACUST UNITED AC 2017; 24:111-124. [PMID: 30906116 DOI: 10.1111/cpsp.12198] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neurobiological systems may be particularly susceptible to deleterious impact of childhood trauma, and the impact of childhood trauma on development and subsequent functional outcomes across the lifespan has been well-documented. The current review addresses the neurobiological impact of exposure to interpersonal trauma in childhood in the context of executive function, emotion regulation, and dissociation/interoceptive awareness. Subsequent risk for PTSD and depression is also discussed. The pathway of risk from childhood trauma to these cognitive, emotional, and psychiatric outcomes is addressed in terms of potential structural and functional alterations within the hippocampus, prefrontal cortex, and amygdala resulting from chronic or repeated activation of the hypothalamic-pituitary-adrenal (HPA) axis and its interaction with and influence on genetic and epigenetic processes during sensitive periods of development. Implications for practice are discussed.
Collapse
Affiliation(s)
- Dorthie Cross
- Department of Psychology, Georgia Southern University
| | - Negar Fani
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine
| | - Abigail Powers
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine
| | - Bekh Bradley
- Department of Psychiatry and Behavioral Sciences, Emory University School of MedicineAtlanta VA Medical Center
| |
Collapse
|
97
|
Chen H, Lombès M, Le Menuet D. Glucocorticoid receptor represses brain-derived neurotrophic factor expression in neuron-like cells. Mol Brain 2017; 10:12. [PMID: 28403881 PMCID: PMC5389111 DOI: 10.1186/s13041-017-0295-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 04/05/2017] [Indexed: 12/20/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is involved in many functions such as neuronal growth, survival, synaptic plasticity and memorization. Altered expression levels are associated with many pathological situations such as depression, epilepsy, Alzheimer’s, Huntington’s and Parkinson’s diseases. Glucocorticoid receptor (GR) is also crucial for neuron functions, via binding of glucocorticoid hormones (GCs). GR actions largely overlap those of BDNF. It has been proposed that GR could be a regulator of BDNF expression, however the molecular mechanisms involved have not been clearly defined yet. Herein, we analyzed the effect of a GC agonist dexamethasone (DEX) on BDNF expression in mouse neuronal primary cultures and in the newly characterized, mouse hippocampal BZ cell line established by targeted oncogenesis. Mouse Bdnf gene exhibits a complex genomic structure with 8 untranslated exons (I to VIII) splicing onto one common and unique coding exon IX. We found that DEX significantly downregulated total BDNF mRNA expression by around 30%. Expression of the highly expressed exon IV and VI containing transcripts was also reduced by DEX. The GR antagonist RU486 abolished this effect, which is consistent with specific GR-mediated action. Transient transfection assays allowed us to define a short 275 bp region within exon IV promoter responsible for GR-mediated Bdnf repression. Chromatin immunoprecipitation experiments demonstrated GR recruitment onto this fragment, through unidentified transcription factor tethering. Altogether, GR downregulates Bdnf expression through direct binding to Bdnf regulatory sequences. These findings bring new insights into the crosstalk between GR and BDNF signaling pathways both playing a major role in physiology and pathology of the central nervous system.
Collapse
Affiliation(s)
- Hui Chen
- Inserm 1185, Fac Med Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Marc Lombès
- Inserm 1185, Fac Med Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Service d'Endocrinologie et des Maladies de la Reproduction, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Damien Le Menuet
- Inserm 1185, Fac Med Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
98
|
Weaver ICG, Korgan AC, Lee K, Wheeler RV, Hundert AS, Goguen D. Stress and the Emerging Roles of Chromatin Remodeling in Signal Integration and Stable Transmission of Reversible Phenotypes. Front Behav Neurosci 2017; 11:41. [PMID: 28360846 PMCID: PMC5350110 DOI: 10.3389/fnbeh.2017.00041] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 02/24/2017] [Indexed: 01/02/2023] Open
Abstract
The influence of early life experience and degree of parental-infant attachment on emotional development in children and adolescents has been comprehensively studied. Structural and mechanistic insight into the biological foundation and maintenance of mammalian defensive systems (metabolic, immune, nervous and behavioral) is slowly advancing through the emerging field of developmental molecular (epi)genetics. Initial evidence revealed that differential nurture early in life generates stable differences in offspring hypothalamic-pituitary-adrenal (HPA) regulation, in part, through chromatin remodeling and changes in DNA methylation of specific genes expressed in the brain, revealing physical, biochemical and molecular paths for the epidemiological concept of gene-environment interactions. Herein, a primary molecular mechanism underpinning the early developmental programming and lifelong maintenance of defensive (emotional) responses in the offspring is the alteration of chromatin domains of specific genomic regions from a condensed state (heterochromatin) to a transcriptionally accessible state (euchromatin). Conversely, DNA methylation promotes the formation of heterochromatin, which is essential for gene silencing, genomic integrity and chromosome segregation. Therefore, inter-individual differences in chromatin modifications and DNA methylation marks hold great potential for assessing the impact of both early life experience and effectiveness of intervention programs—from guided psychosocial strategies focused on changing behavior to pharmacological treatments that target chromatin remodeling and DNA methylation enzymes to dietary approaches that alter cellular pools of metabolic intermediates and methyl donors to affect nutrient bioavailability and metabolism. In this review article, we discuss the potential molecular mechanism(s) of gene regulation associated with chromatin modeling and programming of endocrine (e.g., HPA and metabolic or cardiovascular) and behavioral (e.g., fearfulness, vigilance) responses to stress, including alterations in DNA methylation and the role of DNA repair machinery. From parental history (e.g., drugs, housing, illness, nutrition, socialization) to maternal-offspring exchanges of nutrition, microbiota, antibodies and stimulation, the nature of nurture provides not only mechanistic insight into how experiences propagate from external to internal variables, but also identifies a composite therapeutic target, chromatin modeling, for gestational/prenatal stress, adolescent anxiety/depression and adult-onset neuropsychiatric disease.
Collapse
Affiliation(s)
- Ian C G Weaver
- Department of Psychology and Neuroscience, and Department of Psychiatry, Dalhousie University Halifax, NS, Canada
| | - Austin C Korgan
- Department of Psychology and Neuroscience, and Department of Psychiatry, Dalhousie University Halifax, NS, Canada
| | - Kristen Lee
- Department of Psychology and Neuroscience, and Department of Psychiatry, Dalhousie University Halifax, NS, Canada
| | - Ryan V Wheeler
- Department of Psychology and Neuroscience, and Department of Psychiatry, Dalhousie University Halifax, NS, Canada
| | - Amos S Hundert
- Department of Psychology and Neuroscience, and Department of Psychiatry, Dalhousie University Halifax, NS, Canada
| | - Donna Goguen
- Department of Psychology and Neuroscience, and Department of Psychiatry, Dalhousie University Halifax, NS, Canada
| |
Collapse
|
99
|
Bondar NP, Merkulova TI. Brain-derived neurotrophic factor and early-life stress: Multifaceted interplay. J Biosci 2017; 41:751-758. [PMID: 27966494 DOI: 10.1007/s12038-016-9648-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The brain-derived neurotrophic factor (BDNF) is a key regulator of neural development and plasticity. Longterm changes in the BDNF pathway are associated with childhood adversity and adult depression symptoms. Initially, stress-induced decreases in the BDNF pathway were found in some studies, but subsequent reports indicated the relationship between stress and BDNF to be much more complex, and the concept was significantly revised. In the present mini-review, we focus on the structure and regulation of the Bbnf gene as well as on the stress-BDNF interactions under early-life adverse conditions.
Collapse
Affiliation(s)
- Natalya P Bondar
- Laboratory of Gene Expression Regulation, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia,
| | | |
Collapse
|
100
|
Ohta KI, Suzuki S, Warita K, Kaji T, Kusaka T, Miki T. Prolonged maternal separation attenuates BDNF-ERK signaling correlated with spine formation in the hippocampus during early brain development. J Neurochem 2017; 141:179-194. [PMID: 28178750 DOI: 10.1111/jnc.13977] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/27/2017] [Accepted: 02/03/2017] [Indexed: 12/18/2022]
Abstract
Maternal separation (MS) is known to affect hippocampal function such as learning and memory, yet the molecular mechanism remains unknown. We hypothesized that these impairments are attributed to abnormities of neural circuit formation by MS, and focused on brain-derived neurotrophic factor (BDNF) as key factor because BDNF signaling has an essential role in synapse formation during early brain development. Using rat offspring exposed to MS for 6 h/day during postnatal days (PD) 2-20, we estimated BDNF signaling in the hippocampus during brain development. Our results show that MS attenuated BDNF expression and activation of extracellular signal-regulated kinase (ERK) around PD 7. Moreover, plasticity-related immediate early genes, which are transcriptionally regulated by BDNF-ERK signaling, were also reduced by MS around PD 7. Interestingly, detailed analysis revealed that MS particularly reduced expression of BDNF gene and immediate early genes in the cornu ammonis 1 (CA1) of hippocampus at PD 7. Considering that BDNF-ERK signaling is involved in spine formation, we next evaluated spine formation in the hippocampus during the weaning period. Our results show that MS particularly reduced mature spine density in proximal apical dendrites of CA1 pyramidal neurons at PD 21. These results suggest that MS could attenuate BDNF-ERK signaling during primary synaptogenesis with a region-specific manner, which is likely to lead to decreased spine formation and maturation observed in the hippocampal CA1 region. It is speculated that this incomplete spine formation during early brain development has an influence on learning capabilities throughout adulthood.
Collapse
Affiliation(s)
- Ken-Ichi Ohta
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Shingo Suzuki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Katsuhiko Warita
- Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Tomohiro Kaji
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takanori Miki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|