1
|
Tonna M, Borrelli DF, Marchesi C, Gerra MC, Dallabona C. Childhood obsessive-compulsive disorder, epigenetics, and heterochrony: An evolutionary and developmental approach. Dev Psychopathol 2025:1-15. [PMID: 40099440 DOI: 10.1017/s0954579425000124] [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: 03/19/2025]
Abstract
Childhood obsessive-compulsive disorder (OCD) stems from a bunch of restricted and repetitive behaviors, which are part of normal behavioral repertoire up to the age of 7. The persistence of compulsive-like behaviors after that age is often associated with unique comorbidity patterns, which are age-at-onset dependent and reflect different developmental stages. In particular, OCD synchronically co-occurs with a broad constellation of neurodevelopmental disorders, whereas diachronically it is related to an increased risk of major adult psychoses. Moreover, OCD is associated with trait-like sensory phenomena, suggesting a common disrupted sensorimotor grounding.The present study is aimed at exploring the hypothesis that this specific temporal and comorbidity OCD profile may be due to a developmental heterochronic mechanism of delay in attenuation of ontogenetically early behavioral patterns. The developmental shift of highly evolutionarily conserved behavioral phenotypes might be regulated by epigenetic changes induced by different conditions of sensory unbalance. This evolutionary and developmental model allows capturing childhood OCD in light of the ultimate causes of ritual behavior throughout phylogeny, namely its "homeostatic" function over conditions of unpredictability. Moreover, it may have important clinical implications, as OCD symptoms could represent putative biomarkers of early divergent developmental trajectories, with a pathoplastic effect on course and outcome.
Collapse
Affiliation(s)
- Matteo Tonna
- Department of Medicine and Surgery, Psychiatric Unit, University of Parma, Parma, Italy
- Department of Mental Health, Local Health Service, Parma, Italy
| | - Davide Fausto Borrelli
- Department of Medicine and Surgery, Psychiatric Unit, University of Parma, Parma, Italy
- Department of Mental Health, Local Health Service, Piacenza, Italy
| | - Carlo Marchesi
- Department of Medicine and Surgery, Psychiatric Unit, University of Parma, Parma, Italy
- Department of Mental Health, Local Health Service, Parma, Italy
| | - Maria Carla Gerra
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, PR, Italy
| | - Cristina Dallabona
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, PR, Italy
| |
Collapse
|
2
|
Jang JH, Lee YJ, Ha IH, Park HJ. The analgesic effect of acupuncture in neuropathic pain: regulatory mechanisms of DNA methylation in the brain. Pain Rep 2024; 9:e1200. [PMID: 39450409 PMCID: PMC11500783 DOI: 10.1097/pr9.0000000000001200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 10/26/2024] Open
Abstract
Recent research has demonstrated that chronic pain, resulting from peripheral nerve injury, leads to various symptoms, including not only allodynia and hyperalgesia but also anxiety, depression, and cognitive impairment. These symptoms are believed to arise due to alterations in gene expression and neural function, mediated by epigenetic changes in chromatin structure. Emerging evidence suggests that acupuncture can modulate DNA methylation within the central nervous system, contributing to pain relief and the mitigation of comorbidities. Specifically, acupuncture has been shown to adjust the DNA methylation of genes related to mitochondrial dysfunction, oxidative phosphorylation, and inflammation pathways within cortical regions, such as the prefrontal cortex, anterior cingulate cortex, and primary somatosensory cortex. In addition, it influences the DNA methylation of genes associated with neurogenesis in hippocampal neurons. This evidence indicates that acupuncture, a treatment with fewer side effects compared with conventional medications, could offer an effective strategy for pain management.
Collapse
Affiliation(s)
- Jae-Hwan Jang
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, Republic of Korea
| | - Yoon Jae Lee
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, Republic of Korea
| | - In-Hyuk Ha
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, Republic of Korea
| | - Hi-Joon Park
- Acupuncture and Meridian Science Research Center (AMSRC), College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Leri J, Liu J, Kelly M, Kertes DA. A preliminary investigation of epigenome-wide DNA methylation and temperament during infancy. Dev Psychobiol 2024; 66:e22475. [PMID: 38470455 DOI: 10.1002/dev.22475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 03/13/2024]
Abstract
This study provides preliminary evidence for an epigenetic architecture of infant temperament. At 12 months of age, blood was collected and assayed for DNA methylation and maternally reported infant temperament was assessed using the Infant Behavior Questionnaire in 67 mother-infant dyads. Epigenome-wide analyses showed that the higher order temperament dimensions Surgency and Negative Affect were associated with DNA methylation. The epigenetic signatures of Surgency and Negative Affect were situated at genes involved in synaptic signaling and plasticity. Although replication is required, these results are consistent with a biologically based model of temperament, create new avenues for hypothesis-driven research into epigenetic pathways that underlie individual differences in temperament, and demonstrate that infant temperament has a widespread epigenetic signature in the methylome.
Collapse
Affiliation(s)
- John Leri
- Department of Psychology, University of Florida, Gainesville, Florida, USA
| | - Jingwen Liu
- Department of Psychology, University of Florida, Gainesville, Florida, USA
| | - Maria Kelly
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Darlene A Kertes
- Department of Psychology, University of Florida, Gainesville, Florida, USA
- UF Genetics Institute, University of Florida, Gainesville, USA
| |
Collapse
|
4
|
Chen TJ, Hung HS, Cheng TL, Wang DC. Histone deacetylase inhibitor attenuates the effects of 27-hydroxycholesterol on the rat brain. Neurosci Lett 2024; 818:137533. [PMID: 37865186 DOI: 10.1016/j.neulet.2023.137533] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Hypercholesterolemia is a risk factor for Alzheimer's disease (AD). Plasma cholesterol does not pass the blood-brain barrier whereas its metabolite 27-hydroxycholesterol (27-OHC) can enter the brain. High 27-OHC in the brain has been suggested to mediate hypercholesterolemia-induced impairments of learning and memory through promoting amyloid-β accumulation and facilitating synaptic disruption. In AD brains, the activity of histone deacetylase (HDAC) is elevated. Treating AD animals with HDAC inhibitors decreases amyloid-β levels and synaptic damages, which leads to memory improvement. Whether HDAC activity is involved in the actions of 27-OHC is still uncertain. In this study, 4 weekly injections of 27-OHC/vehicle were given to rats followed by 3 daily injections of HDAC inhibitor trichostatin (TSA)/vehicle. The results of Morris water maze test reveal that all rats have intact spatial learning ability during the 5-d training phase. However, the behavioral performance during the probe trial was impaired by 27-OHC treatment, which was improved by adding TSA treatments. Furthermore, 27-OHC treatments reduced the hippocampal levels of acetylated histone H3, acetylated α tubulin, insulin-degrading enzyme and postsynaptic protein PSD-95, indicating that 27-OHC treatments may induce enhanced HDAC activity, decreased amyloid-β clearance and synaptic disruption. All reduced levels returned to the basal levels by adding TSA treatments. These findings support our hypothesis that HDAC activity is enhanced following long-term exposure to excess 27-OHC.
Collapse
Affiliation(s)
- Tsan-Ju Chen
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Hui-Shan Hung
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tsung-Lin Cheng
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Dean-Chuan Wang
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
5
|
Spies J, Covarrubias-Pinto A, Carcamo C, Arancibia Y, Salazar F, Paredes-Martinez C, Otth C, Castro M, Zambrano A. Modulation of Synaptic Plasticity Genes Associated to DNA Damage in a Model of Huntington's Disease. Neurochem Res 2023; 48:2093-2103. [PMID: 36790580 DOI: 10.1007/s11064-023-03889-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/16/2023]
Abstract
Huntington's disease (HD) is a disease characterized by the progressive degeneration of nerve cells in the brain. DNA damage has been implicated in many neurological disorders; however, the association between this damage and the impaired signaling related to neurodegeneration is still unclear. The transcription factor c-AMP-responsive element binding protein (CREB) has a relevant role in the neuronal plasticity process regulating the expression of several genes, including brain-derived neurotrophic factor (BDNF). Here we analyzed the direct link between DNA damage and the expression of genes involved in neuronal plasticity. The study was performed in model cell lines STHdhQ7 (wild type) and STHdhQ111 (HD model). Treatment with Etoposide (Eto) was used to induce double-strand breaks (DSBs) to evaluate the DNA damage response (DDR) and the expression of synaptic plasticity genes. Eto treatment induced phosphorylation of ATM (p-ATM) and H2AX (γH2AX), markers of DDR, in both cell lines. Interestingly, upon DNA damage, STHdhQ7 cells showed increased expression of activity-regulated cytoskeleton associated protein (Arc) and BDNF when compared to the HD cell line model. Additionally, Eto induced CREB activation with a differential localization of its co-activators in the cell types analyzed. These results suggest that DSBs impact differentially the gene expression patterns of plasticity genes in the normal cell line versus the HD model. This effect is mediated by the impaired localization of CREB-binding protein (CBP) and histone acetylation in the HD model. Our results highlight the role of epigenetics and DNA repair on HD and therefore we suggest that future studies should explore in depth the epigenetic landscape on neuronal pathologies with the goal to further understand molecular mechanisms and pinpoint therapeutic targets.
Collapse
Affiliation(s)
- Johana Spies
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Adriana Covarrubias-Pinto
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Constanza Carcamo
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Yennyfer Arancibia
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Fernanda Salazar
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Carolina Paredes-Martinez
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Carola Otth
- Facultad de Medicina, Instituto de Microbiología Clínica, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Maite Castro
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso (CINV), Valparaíso, Chile
| | - Angara Zambrano
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile.
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
6
|
Maity S, Abbaspour R, Nahabedian D, Connor SA. Norepinephrine, beyond the Synapse: Coordinating Epigenetic Codes for Memory. Int J Mol Sci 2022; 23:ijms23179916. [PMID: 36077313 PMCID: PMC9456295 DOI: 10.3390/ijms23179916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
The noradrenergic system is implicated in neuropathologies contributing to major disorders of the memory, including post-traumatic stress disorder and Alzheimer’s disease. Determining the impact of norepinephrine on cellular function and plasticity is thus essential for making inroads into our understanding of these brain conditions, while expanding our capacity for treating them. Norepinephrine is a neuromodulator within the mammalian central nervous system which plays important roles in cognition and associated synaptic plasticity. Specifically, norepinephrine regulates the formation of memory through the stimulation of β-ARs, increasing the dynamic range of synaptic modifiability. The mechanisms through which NE influences neural circuit function have been extended to the level of the epigenome. This review focuses on recent insights into how the noradrenergic recruitment of epigenetic modifications, including DNA methylation and post-translational modification of histones, contribute to homo- and heterosynaptic plasticity. These advances will be placed in the context of synaptic changes associated with memory formation and linked to brain disorders and neurotherapeutic applications.
Collapse
Affiliation(s)
- Sabyasachi Maity
- Department of Physiology, Neuroscience, and Behavioral Sciences, St. George’s University School of Medicine, True Blue FZ818, Grenada
| | - Raman Abbaspour
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - David Nahabedian
- The Center for Biomedical Visualization, Department of Anatomical Sciences, St. George’s University School of Medicine, True Blue FZ818, Grenada
| | - Steven A. Connor
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
- Correspondence: ; Tel.: +1-(416)-736-2100 (ext. 33803)
| |
Collapse
|
7
|
Wang T, Xu J, Xu Y, Xiao J, Bi N, Gu X, Wang HL. Gut microbiota shapes social dominance through modulating HDAC2 in the medial prefrontal cortex. Cell Rep 2022; 38:110478. [PMID: 35263606 DOI: 10.1016/j.celrep.2022.110478] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/29/2021] [Accepted: 02/08/2022] [Indexed: 12/17/2022] Open
Abstract
Social dominance is a ubiquitous phenomenon among social animals, including humans. To date, individual attributes leading to dominance (after a contest) remain largely elusive. Here, we report that socially dominant rats can be distinguished from subordinates based on their intestinal microbiota. When dysbiosis is induced, rats are predisposed to a subordinate state, while dysbiotic rats reclaim social dominance following microbiota transplantation. Winning hosts are characterized by core microbes, a majority of which are associated with butyrate production, and the sole colonization of Clostridium butyricum is sufficient to restore dominance. Regarding molecular aspects, a histone deacetylase, HDAC2, is responsive to microbial status and mediates competition outcome; however, this occurs only in a restricted population of cells in the medial prefrontal cortex (mPFC). Furthermore, HDAC2 acts by modulating synaptic activity in mPFC. Together, these findings uncover a link between commensals and host dominance, providing insight into the gut-brain mechanisms underlying dominance determination.
Collapse
Affiliation(s)
- Tian Wang
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Jinchun Xu
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Yi Xu
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei 230009, China.
| | - Jie Xiao
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Nanxi Bi
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Xiaozhen Gu
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Hui-Li Wang
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei 230009, China.
| |
Collapse
|
8
|
Christensen J, Beveridge JK, Wang M, Orr SL, Noel M, Mychasiuk R. A Pilot Study Investigating the Role of Gender in the Intergenerational Relationships between Gene Expression, Chronic Pain, and Adverse Childhood Experiences in a Clinical Sample of Youth with Chronic Pain. EPIGENOMES 2021; 5:epigenomes5020009. [PMID: 34968296 PMCID: PMC8594698 DOI: 10.3390/epigenomes5020009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/24/2021] [Accepted: 04/13/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic pain is a highly prevalent and costly issue that often emerges during childhood or adolescence and persists into adulthood. Adverse childhood experiences (ACEs) increase risk for several adverse health conditions, including chronic pain. Recent evidence suggests that parental trauma (ACEs, post-traumatic stress disorder (PTSD) symptoms) confers risk of poor health outcomes in their children. Intergenerational relationships between parental trauma and child chronic pain may be mediated by epigenetic mechanisms. A clinical sample of youth with chronic pain and their parents completed psychometrically sound questionnaires assessing ACEs, PTSD symptoms, and chronic pain, and provided a saliva sample. These were used to investigate the intergenerational relationships between four epigenetic biomarkers (COMT, DRD2, GR, and SERT), trauma, and chronic pain. The results indicated that the significant biomarkers were dependent upon the gender of the child, wherein parental ACEs significantly correlated with changes in DRD2 expression in female children and altered COMT expression in the parents of male children. Additionally, the nature of the ACE (maltreatment vs. household dysfunction) was associated with the specific epigenetic changes. There may be different pathways through which parental ACEs confer risk for poor outcomes for males and females, highlighting the importance of child gender in future investigations.
Collapse
Affiliation(s)
- Jennaya Christensen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia;
| | - Jaimie K. Beveridge
- Department of Psychology, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.K.B.); (M.N.)
| | - Melinda Wang
- Hotchkiss Brain Institute, Calgary, AB T2N 4N1, Canada;
- Alberta Children’s Hospital Research Institute, Calgary, AB T2N 4N1, Canada
| | - Serena L. Orr
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Melanie Noel
- Department of Psychology, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.K.B.); (M.N.)
- Hotchkiss Brain Institute, Calgary, AB T2N 4N1, Canada;
- Alberta Children’s Hospital Research Institute, Calgary, AB T2N 4N1, Canada
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia;
- Department of Psychology, University of Calgary, Calgary, AB T2N 1N4, Canada; (J.K.B.); (M.N.)
- Hotchkiss Brain Institute, Calgary, AB T2N 4N1, Canada;
- Alberta Children’s Hospital Research Institute, Calgary, AB T2N 4N1, Canada
- Correspondence:
| |
Collapse
|
9
|
López AJ, Hecking JK, White AO. The Emerging Role of ATP-Dependent Chromatin Remodeling in Memory and Substance Use Disorders. Int J Mol Sci 2020; 21:E6816. [PMID: 32957495 PMCID: PMC7555352 DOI: 10.3390/ijms21186816] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023] Open
Abstract
Long-term memory formation requires coordinated regulation of gene expression and persistent changes in cell function. For decades, research has implicated histone modifications in regulating chromatin compaction necessary for experience-dependent changes to gene expression and cell function during memory formation. Recent evidence suggests that another epigenetic mechanism, ATP-dependent chromatin remodeling, works in concert with the histone-modifying enzymes to produce large-scale changes to chromatin structure. This review examines how histone-modifying enzymes and chromatin remodelers restructure chromatin to facilitate memory formation. We highlight the emerging evidence implicating ATP-dependent chromatin remodeling as an essential mechanism that mediates activity-dependent gene expression, plasticity, and cell function in developing and adult brains. Finally, we discuss how studies that target chromatin remodelers have expanded our understanding of the role that these complexes play in substance use disorders.
Collapse
Affiliation(s)
- Alberto J. López
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
| | - Julia K. Hecking
- Department of Biological Sciences, Mount Holyoke College, South Hadley, MA 01075, USA;
| | - André O. White
- Department of Biological Sciences, Mount Holyoke College, South Hadley, MA 01075, USA;
| |
Collapse
|
10
|
Guerrero TP, Fickel J, Benhaiem S, Weyrich A. Epigenomics and gene regulation in mammalian social systems. Curr Zool 2020; 66:307-319. [PMID: 32440291 PMCID: PMC7233906 DOI: 10.1093/cz/zoaa005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 02/12/2020] [Indexed: 12/19/2022] Open
Abstract
Social epigenomics is a new field of research that studies how the social environment shapes the epigenome and how in turn the epigenome modulates behavior. We focus on describing known gene-environment interactions (GEIs) and epigenetic mechanisms in different mammalian social systems. To illustrate how epigenetic mechanisms integrate GEIs, we highlight examples where epigenetic mechanisms are associated with social behaviors and with their maintenance through neuroendocrine, locomotor, and metabolic responses. We discuss future research trajectories and open questions for the emerging field of social epigenomics in nonmodel and naturally occurring social systems. Finally, we outline the technological advances that aid the study of epigenetic mechanisms in the establishment of GEIs and vice versa.
Collapse
Affiliation(s)
- Tania P Guerrero
- Department Evolutionary Genetics, Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Str. 17, Berlin, D-10315, Germany
- Faculty of Environment and Natural Resources, Albert Ludwig University of Freiburg, Tennenbacher Str. 4, Freiburg, D-79085, Germany
| | - Jörns Fickel
- Department Evolutionary Genetics, Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Str. 17, Berlin, D-10315, Germany
- Institute for Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, Potsdam, 14476, Germany
| | - Sarah Benhaiem
- Department Ecological Dynamics, Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Str. 17, Berlin, D-10315, Germany
| | - Alexandra Weyrich
- Department Evolutionary Genetics, Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Str. 17, Berlin, D-10315, Germany
| |
Collapse
|
11
|
Torres-Ferrús M, Ursitti F, Alpuente-Ruiz A, Brunello F, Chiappino D, de Vries T, Di Marco S, Ferlisi S, Guerritore L, Gonzalez-Garcia N, Gonzalez-Martinez A, Khutorov D, Kritsilis M, Kyrou A, Makeeva T, Minguez-Olaondo A, Pilati L, Serrien A, Tsurkalenko O, Van den Abbeele D, van Hoogstraten WS, Lampl C. From transformation to chronification of migraine: pathophysiological and clinical aspects. J Headache Pain 2020; 21:42. [PMID: 32349653 PMCID: PMC7189559 DOI: 10.1186/s10194-020-01111-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic migraine is a neurological disorder characterized by 15 or more headache days per month of which at least 8 days show typical migraine features. The process that describes the development from episodic migraine into chronic migraine is commonly referred to as migraine transformation or chronification. Ample studies have attempted to identify factors associated with migraine transformation from different perspectives. Understanding CM as a pathological brain state with trigeminovascular participation where biological changes occur, we have completed a comprehensive review on the clinical, epidemiological, genetic, molecular, structural, functional, physiological and preclinical evidence available.
Collapse
Affiliation(s)
- M. Torres-Ferrús
- Headache and Craniofacial Pain Unit, Neurology Department, Hospital Universitari Vall d’Hebron, Headache and Neurological Pain Research Group, Vall d’Hebron Research Institute (VHIR), Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - F. Ursitti
- Headache Center, Child Neurology Unit, Bambino Gesu’ Children’s Hospital, Rome, Italy
| | - A. Alpuente-Ruiz
- Headache and Craniofacial Pain Unit, Neurology Department, Hospital Universitari Vall d’Hebron, Headache and Neurological Pain Research Group, Vall d’Hebron Research Institute (VHIR), Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - F. Brunello
- Juvenile Headache Centre, Department of Woman’s and Child’s Health, University Hospital of Padua, Padua, Italy
| | - D. Chiappino
- Department of Internal medicine, Sant’Andrea Hospital, University of Rome, Sapienza, Italy
| | - T. de Vries
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S. Di Marco
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Policlinico Paolo Giaccone Hospital, University of Palermo, Palermo, Italy
| | - S. Ferlisi
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Policlinico Paolo Giaccone Hospital, University of Palermo, Palermo, Italy
| | - L. Guerritore
- Department of Internal medicine, Sant’Andrea Hospital, University of Rome, Sapienza, Italy
| | - N. Gonzalez-Garcia
- Headache and Craniofacial Pain Unit, Neurology Department, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - A. Gonzalez-Martinez
- Neurology Department, Hospital Universitario de La Princesa & Instituto de Investigación Sanitaria de La Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - D. Khutorov
- Department of Clinical Neurology and Sleep Medicine, The Nikiforov Russian Center of Emergency and Radiation Medicine of EMERCOM of Russia, Saint-Petersburg, Russia
| | | | - A. Kyrou
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Switzerland University Hospital of Psychiatry, Bern, Switzerland
| | - T. Makeeva
- Headache Unit, Department of Neurology, Medical center “New Medical Technologies”, Voronezh, Russia
| | - A. Minguez-Olaondo
- Department of Neurology, Universitary Hospital of Donostia, San Sebastian, Spain
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Neurology, Hospital Quironsalud Donostia, San Sebastian, Spain
| | - L. Pilati
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Policlinico Paolo Giaccone Hospital, University of Palermo, Palermo, Italy
| | - A. Serrien
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - O. Tsurkalenko
- Department of Neurology and Neurosurgery, State Institution “Dnipropetrovsk medical akademy MOH Ukraine”, Dnipro, Ukraine
| | | | - W. S. van Hoogstraten
- Department of Neuroscience, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - C. Lampl
- Headache Medical Center Linz, Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - On behalf of School of Advanced Studies of European Headache Federation (EHF-SAS)
- Headache and Craniofacial Pain Unit, Neurology Department, Hospital Universitari Vall d’Hebron, Headache and Neurological Pain Research Group, Vall d’Hebron Research Institute (VHIR), Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
- Headache Center, Child Neurology Unit, Bambino Gesu’ Children’s Hospital, Rome, Italy
- Juvenile Headache Centre, Department of Woman’s and Child’s Health, University Hospital of Padua, Padua, Italy
- Department of Internal medicine, Sant’Andrea Hospital, University of Rome, Sapienza, Italy
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Policlinico Paolo Giaccone Hospital, University of Palermo, Palermo, Italy
- Headache and Craniofacial Pain Unit, Neurology Department, Hospital Universitario Clínico San Carlos, Madrid, Spain
- Neurology Department, Hospital Universitario de La Princesa & Instituto de Investigación Sanitaria de La Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Clinical Neurology and Sleep Medicine, The Nikiforov Russian Center of Emergency and Radiation Medicine of EMERCOM of Russia, Saint-Petersburg, Russia
- Grevena General Hospital, Grevena, Greece
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Switzerland University Hospital of Psychiatry, Bern, Switzerland
- Headache Unit, Department of Neurology, Medical center “New Medical Technologies”, Voronezh, Russia
- Department of Neurology, Universitary Hospital of Donostia, San Sebastian, Spain
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Neurology, Hospital Quironsalud Donostia, San Sebastian, Spain
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Neurology and Neurosurgery, State Institution “Dnipropetrovsk medical akademy MOH Ukraine”, Dnipro, Ukraine
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
- Department of Neuroscience, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Headache Medical Center Linz, Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| |
Collapse
|
12
|
Cantacorps L, Alfonso-Loeches S, Guerri C, Valverde O. Long-term epigenetic changes in offspring mice exposed to alcohol during gestation and lactation. J Psychopharmacol 2019; 33:1562-1572. [PMID: 31210079 DOI: 10.1177/0269881119856001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Alcohol exposure impairs brain development and leads to a range of behavioural and cognitive dysfunctions, termed as foetal alcohol spectrum disorders. Although different mechanisms have been proposed to participate in foetal alcohol spectrum disorders, the molecular insights of such effects are still uncertain. Using a mouse model of foetal alcohol spectrum disorder, we have previously shown that maternal binge-like alcohol drinking causes persistent effects on motor, cognitive and emotional-related behaviours associated with neuroimmune dysfunctions. AIMS In this study, we sought to evaluate whether the long-term behavioural alterations found in offspring with early exposure to alcohol are associated with epigenetic changes in the hippocampus and prefrontal cortex. METHODS Pregnant C57BL/6 female mice underwent a model procedure for binge alcohol drinking throughout both the gestation and lactation periods. Subsequently, adult offspring were assessed for their cognitive function in a reversal learning task and brain areas were extracted for epigenetic analyses. RESULTS The results demonstrated that early binge alcohol exposure induces long-term behavioural effects along with alterations in histone acetylation (histone H4 lysine 5 and histone H4 lysine 12) in the hippocampus and prefrontal cortex. The epigenetic effects were linked with an imbalance in histone acetyltransferase activity that was found to be increased in the prefrontal cortex of mice exposed to alcohol. CONCLUSIONS In conclusion, our results reveal that maternal binge-like alcohol consumption induces persistent epigenetic modifications, effects that might be associated with the long-term cognitive and behavioural impairments observed in foetal alcohol spectrum disorder models.
Collapse
Affiliation(s)
- Lídia Cantacorps
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Silvia Alfonso-Loeches
- Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Centre, Valencia, Spain
| | - Consuelo Guerri
- Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Centre, Valencia, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Neuroscience Research Programme, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| |
Collapse
|
13
|
Short AK, Baram TZ. Early-life adversity and neurological disease: age-old questions and novel answers. Nat Rev Neurol 2019; 15:657-669. [PMID: 31530940 PMCID: PMC7261498 DOI: 10.1038/s41582-019-0246-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
Neurological illnesses, including cognitive impairment, memory decline and dementia, affect over 50 million people worldwide, imposing a substantial burden on individuals and society. These disorders arise from a combination of genetic, environmental and experiential factors, with the latter two factors having the greatest impact during sensitive periods in development. In this Review, we focus on the contribution of adverse early-life experiences to aberrant brain maturation, which might underlie vulnerability to cognitive brain disorders. Specifically, we draw on recent robust discoveries from diverse disciplines, encompassing human studies and experimental models. These discoveries suggest that early-life adversity, especially in the perinatal period, influences the maturation of brain circuits involved in cognition. Importantly, new findings suggest that fragmented and unpredictable environmental and parental signals comprise a novel potent type of adversity, which contributes to subsequent vulnerabilities to cognitive illnesses via mechanisms involving disordered maturation of brain 'wiring'.
Collapse
Affiliation(s)
- Annabel K Short
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA.
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA.
- Departments of Neurology, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
14
|
Brandwein NJ, Nguyen PV. A requirement for epigenetic modifications during noradrenergic stabilization of heterosynaptic LTP in the hippocampus. Neurobiol Learn Mem 2019; 161:72-82. [PMID: 30930287 DOI: 10.1016/j.nlm.2019.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 12/31/2022]
Abstract
Beta-adrenergic receptor (b-AR) activation by noradrenaline (NA) enhances memory formation and long-term potentiation (LTP), a form of synaptic plasticity characterized by an activity-dependent increase in synaptic strength. LTP is believed to be a cellular mechanism for contextual learning and memory. In the mammalian hippocampus, LTP can be observed at multiple synaptic pathways after strong stimulation of a single synaptic pathway. This heterosynaptic LTP is believed to involve synaptic tagging of active synapses and capture of plasticity-related proteins that enable heterosynaptic transfer of persistent potentiation. These processes may permit distinct neural pathways to associate information transmitted by separate, but convergent, synaptic inputs. We had previously shown that transcription and epigenetic modifications were necessary for stabilization of homosynaptic LTP. However, it is unclear whether transfer of LTP to a second, heterosynaptic pathway involves b-ARs signalling to the nucleus. Using electrophysiologic recordings in area CA1 of murine hippocampal slices, we show here that pharmacologically inhibiting b-AR activation, transcription, DNA methyltransferase or histone acetyltransferase activation, prevents stabilization of heterosynaptic LTP. Our data suggest that noradrenergic stabilization of heterosynaptic ("tagged") LTP requires not only transcription, but specifically, DNA methylation and histone acetylation. NA promotes stable heterosynaptic plasticity through engagement of nuclear processes that may contribute to prompt consolidation of short-term memories into resilient long-term memories under conditions when the brain's noradrenergic system is recruited.
Collapse
Affiliation(s)
- N J Brandwein
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - P V Nguyen
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada.
| |
Collapse
|
15
|
Murillo-Rodríguez E, Arankowsky-Sandoval G, Barros JA, Rocha NB, Yamamoto T, Machado S, Budde H, Telles-Correia D, Monteiro D, Cid L, Veras AB. Sleep and Neurochemical Modulation by DZNep and GSK-J1: Potential Link With Histone Methylation Status. Front Neurosci 2019; 13:237. [PMID: 30930741 PMCID: PMC6428769 DOI: 10.3389/fnins.2019.00237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/27/2019] [Indexed: 12/24/2022] Open
Abstract
Histone methylation/demethylation plays an important modulatory role in chromatin restructuring, RNA transcription and is essential for controlling a plethora of biological processes. Due to many human diseases have been related to histone methylation/demethylation, several compounds such as 3-deazaneplanocin A (DZNep) or 3-((6-(4,5-Dihydro-1H-benzo[d]azepin-3(2H)-yl)-2-(pyridin-2-yl)pyrimidin-4-yl)amino)propanoic acid; N-[2-(2-pyridinyl)-6-(1,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)-4-pyrimidinyl]-β-Alanine (GSK-J1), have been designed to inhibit histone methylase or suppress histone demethylase, respectively. In the present study, we investigated the effects on the sleep-wake cycle and sleep-related neurochemical levels after systemic injections of DZNep or GSK-J1 given during the light or dark phase in rats. DZNep dose-dependently (0.1, 1.0, or 10 mg/kg, i.p.) prolonged wakefulness (W) duration while decreased slow wave sleep (SWS) and rapid eye movement sleep (REMS) time spent during the lights-on period with no changes observed in dark phase. In opposite direction, GSK-J1 (0.1, 1.0, or 10 mg/kg, i.p.) injected at the beginning of the lights-on period induced no statistical changes in W, SWS, or REMS whereas if administered at darkness, we found a diminution in W and an enhancement in SWS and REMS. Finally, brain microdialysis experiments in freely moving animals were used to evaluate the effects of DZNep or GSK-J1 treatments on contents of sleep-related neurochemicals. The results showed that DZNep boosted extracellular levels of dopamine, norepinephrine, epinephrine, serotonin, adenosine, and acetylcholine if injected at the beginning of the lights-on period whereas GSK-J1 exerted similar outcomes but when administered at darkness. In summary, DZNep and GSK-J1 may control the sleep-wake cycle and sleep-related neurochemicals through histone methylation/demethylation activity.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Mexico.,Intercontinental Neuroscience Research Group, Mérida, Mexico
| | - Gloria Arankowsky-Sandoval
- Centro de Investigaciones Regionales "Dr. Hideyo Noguchi" Universidad Autónoma de Yucatán, Mérida, Mexico
| | - Jorge Aparecido Barros
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Post-graduation Program of Psychology of Health, NACNeuro, Dom Bosco Catholic University, Campo Grande, Mato Grosso del Sur, Brazil
| | - Nuno Barbosa Rocha
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,School of Health, Polytechnic Institute of Porto, Porto, Portugal
| | - Tetsuya Yamamoto
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Sérgio Machado
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Laboratory of Physical Activity Neuroscience, Physical Activity Sciences Postgraduate Program, Salgado de Oliveira University, Niterói, Brazil
| | - Henning Budde
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Faculty of Human Sciences, Medical School Hamburg, Hamburg, Germany.,Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Diogo Telles-Correia
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,University of Lisbon, Faculty of Medicine, Lisbon, Portugal
| | - Diogo Monteiro
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Sport Science School of Rio Maior- Polytechnic Institute of Santarém, Rio Maior, Portugal.,Research Center in Sport, Health and Human Development-CIDESD, Vila Real, Portugal
| | - Luis Cid
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Sport Science School of Rio Maior- Polytechnic Institute of Santarém, Rio Maior, Portugal.,Research Center in Sport, Health and Human Development-CIDESD, Vila Real, Portugal
| | - André Barciela Veras
- Intercontinental Neuroscience Research Group, Mérida, Mexico.,Post-graduation Program of Psychology of Health, NACNeuro, Dom Bosco Catholic University, Campo Grande, Mato Grosso del Sur, Brazil
| |
Collapse
|
16
|
López AJ, Siciliano CA, Calipari ES. Activity-Dependent Epigenetic Remodeling in Cocaine Use Disorder. Handb Exp Pharmacol 2019; 258:231-263. [PMID: 31628597 DOI: 10.1007/164_2019_257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Substance use disorder (SUD) is a behavioral disorder characterized by cycles of abstinence, drug seeking, and relapse. SUD is characterized by aberrant learning processes which develop after repeated exposure to drugs of abuse. At the core of this phenotype is the persistence of symptoms, such as craving and relapse to drug seeking, long after the cessation of drug use. The neural basis of these behavioral changes has been linked to dysfunction in neural circuits across the brain; however, the molecular drivers that allow for these changes to persist beyond the lifespan of any individual protein remain opaque. Epigenetic adaptations - where DNA is modified to increase or decrease the probability of gene expression at key genes - have been identified as a mechanism underlying the long-lasting nature of drug-seeking behavior. Thus, to understand SUD, it is critical to define the interplay between neuronal activation and longer-term changes in transcription and epigenetic remodeling and define their role in addictive behaviors. In this review, we discuss the current understanding of drug-induced changes to circuit function, recent discoveries in epigenetic mechanisms that mediate these changes, and, ultimately, how these adaptations drive the persistent nature of relapse, with emphasis on adaptations in models of cocaine use disorder. Understanding the complex interplay between epigenetic gene regulation and circuit activity will be critical in elucidating the neural mechanisms underlying SUD. This, with the advent of novel genetic-based techniques, will allow for the generation of novel therapeutic avenues to improve treatment outcomes in SUD.
Collapse
Affiliation(s)
- Alberto J López
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Department of Molecular Physiology and Biophysics, Vanderbilt Institute for Infection, Immunology, and Infection, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Department of Psychiatry and Behavioral Sciences, Vanderbilt Institute for Infection, Immunology, and Infection, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
17
|
Postnatal TrkB ablation in corticolimbic interneurons induces social dominance in male mice. Proc Natl Acad Sci U S A 2018; 115:E9909-E9915. [PMID: 30282736 DOI: 10.1073/pnas.1812083115] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The tight balance between synaptic excitation and inhibition (E/I) within neocortical circuits in the mammalian brain is important for complex behavior. Many loss-of-function studies have demonstrated that brain-derived neurotrophic factor (BDNF) and its cognate receptor tropomyosin receptor kinase B (TrkB) are essential for the development of inhibitory GABAergic neurons. However, behavioral consequences of impaired BDNF/TrkB signaling in GABAergic neurons remain unclear, largely due to confounding motor function deficits observed in previous animal models. In this study, we generated conditional knockout mice (TrkB cKO) in which TrkB was ablated from a majority of corticolimbic GABAergic interneurons postnatally. These mice showed intact motor coordination and movement, but exhibited enhanced dominance over other mice in a group-housed setting. In addition, immature fast-spiking GABAergic neurons of TrkB cKO mice resulted in an E/I imbalance in layer 5 microcircuits within the medial prefrontal cortex (mPFC), a key region regulating social dominance. Restoring the E/I imbalance via optogenetic modulation in the mPFC of TrkB cKO mice normalized their social dominance behavior. Taken together, our results provide strong evidence for a role of BDNF/TrkB signaling in inhibitory synaptic modulation and social dominance behavior in mice.
Collapse
|
18
|
Montagud-Romero S, Blanco-Gandía MC, Reguilón MD, Ferrer-Pérez C, Ballestín R, Miñarro J, Rodríguez-Arias M. Social defeat stress: Mechanisms underlying the increase in rewarding effects of drugs of abuse. Eur J Neurosci 2018; 48:2948-2970. [PMID: 30144331 DOI: 10.1111/ejn.14127] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/11/2018] [Accepted: 08/14/2018] [Indexed: 12/27/2022]
Abstract
Social interaction is known to be the main source of stress in human beings, which explains the translational importance of this research in animals. Evidence reported over the last decade has revealed that, when exposed to social defeat experiences (brief episodes of social confrontations during adolescence and adulthood), the rodent brain undergoes remodeling and functional modifications, which in turn lead to an increase in the rewarding and reinstating effects of different drugs of abuse. The mechanisms by which social stress cause changes in the brain and behavior are unknown, and so the objective of this review is to contemplate how social defeat stress induces long-lasting consequences that modify the reward system. First of all, we will describe the most characteristic results of the short- and long-term consequences of social defeat stress on the rewarding effects of drugs of abuse such as psychostimulants and alcohol. Secondly, and throughout the review, we will carefully assess the neurobiological mechanisms underlying these effects, including changes in the dopaminergic system, corticotrophin releasing factor signaling, epigenetic modifications and the neuroinflammatory response. To conclude, we will consider the advantages and disadvantages and the translational value of the social defeat stress model, and will discuss challenges and future directions.
Collapse
Affiliation(s)
- Sandra Montagud-Romero
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | | | - Marina D Reguilón
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Carmen Ferrer-Pérez
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Raul Ballestín
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Jose Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| |
Collapse
|
19
|
Reducing histone acetylation rescues cognitive deficits in a mouse model of Fragile X syndrome. Nat Commun 2018; 9:2494. [PMID: 29950602 PMCID: PMC6021376 DOI: 10.1038/s41467-018-04869-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 05/23/2018] [Indexed: 12/19/2022] Open
Abstract
Fragile X syndrome (FXS) is the most prevalent inherited intellectual disability, resulting from a loss of fragile X mental retardation protein (FMRP). Patients with FXS suffer lifelong cognitive disabilities, but the function of FMRP in the adult brain and the mechanism underlying age-related cognitive decline in FXS is not fully understood. Here, we report that a loss of FMRP results in increased protein synthesis of histone acetyltransferase EP300 and ubiquitination-mediated degradation of histone deacetylase HDAC1 in adult hippocampal neural stem cells (NSCs). Consequently, FMRP-deficient NSCs exhibit elevated histone acetylation and age-related NSC depletion, leading to cognitive impairment in mature adult mice. Reducing histone acetylation rescues both neurogenesis and cognitive deficits in mature adult FMRP-deficient mice. Our work reveals a role for FMRP and histone acetylation in cognition and presents a potential novel therapeutic strategy for treating adult FXS patients. Loss of fragile X mental retardation protein (FMRP) leads to fragile X syndrome, associated with cognitive dysfunction. Here the authors show that mice lacking FMRP show reduced hippocampal neurogenesis and cognitive deficits, which can be rescued by reducing histone acetylation.
Collapse
|
20
|
Restoring Tip60 HAT/HDAC2 Balance in the Neurodegenerative Brain Relieves Epigenetic Transcriptional Repression and Reinstates Cognition. J Neurosci 2018; 38:4569-4583. [PMID: 29654189 DOI: 10.1523/jneurosci.2840-17.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/26/2018] [Accepted: 04/06/2018] [Indexed: 12/16/2022] Open
Abstract
Cognitive decline is a debilitating hallmark during preclinical stages of Alzheimer's disease (AD), yet the causes remain unclear. Because histone acetylation homeostasis is critical for mediating epigenetic gene control throughout neuronal development, we postulated that its misregulation contributes to cognitive impairment preceding AD pathology. Here, we show that disruption of Tip60 histone acetlytransferase (HAT)/histone deacetylase 2 (HDAC2) homeostasis occurs early in the brain of an AD-associated amyloid precursor protein (APP) Drosophila model and triggers epigenetic repression of neuroplasticity genes well before Aβ plaques form in male and female larvae. Repressed genes display enhanced HDAC2 binding and reduced Tip60 and histone acetylation enrichment. Increasing Tip60 in the AD-associated APP brain restores Tip60 HAT/HDAC2 balance by decreasing HDAC2 levels, reverses neuroepigenetic alterations to activate synaptic plasticity genes, and reinstates brain morphology and cognition. Such Drosophila neuroplasticity gene epigenetic signatures are conserved in male and female mouse hippocampus and their expression and Tip60 function is compromised in hippocampus from AD patients. We suggest that Tip60 HAT/HDAC2-mediated epigenetic gene disruption is a critical initial step in AD that is reversed by restoring Tip60 in the brain.SIGNIFICANCE STATEMENT Mild cognitive impairment is a debilitating hallmark during preclinical stages of Alzheimer's disease (AD), yet its causes remain unclear. Although recent findings support elevated histone deacetylase 2 (HDAC2) as a cause for epigenetic repression of synaptic genes that contribute to cognitive deficits, whether alterations in histone acetlytransferase (HAT) levels that counterbalance HDAC2 repressor action occur and the identity of these HATs remain unknown. We demonstrate that disruption of Tip60 HAT/HDAC2 homeostasis occurs early in the AD Drosophila brain and triggers epigenetic repression of neuroplasticity genes before Aβ plaques form. Increasing Tip60 in the AD brain restores Tip60 HAT/HDAC2 balance, reverses neuroepigenetic alterations to activate synaptic genes, and reinstates brain morphology and cognition. Our data suggest that disruption of the Tip60 HAT/HDAC2 balance is a critical initial step in AD.
Collapse
|
21
|
Koberstein JN, Poplawski SG, Wimmer ME, Porcari G, Kao C, Gomes B, Risso D, Hakonarson H, Zhang NR, Schultz RT, Abel T, Peixoto L. Learning-dependent chromatin remodeling highlights noncoding regulatory regions linked to autism. Sci Signal 2018; 11:11/513/eaan6500. [PMID: 29339533 DOI: 10.1126/scisignal.aan6500] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder that is associated with genetic risk factors. Most human disease-associated single-nucleotide polymorphisms (SNPs) are not located in genes but rather are in regulatory regions that control gene expression. The function of regulatory regions is determined through epigenetic mechanisms. Parallels between the cellular basis of development and the formation of long-term memory have long been recognized, particularly the role of epigenetic mechanisms in both processes. We analyzed how learning alters chromatin accessibility in the mouse hippocampus using a new high-throughput sequencing bioinformatics strategy we call DEScan (differential enrichment scan). DEScan, which enabled the analysis of data from epigenomic experiments containing multiple replicates, revealed changes in chromatin accessibility at 2365 regulatory regions-most of which were promoters. Learning-regulated promoters were active during forebrain development in mice and were enriched in epigenetic modifications indicative of bivalent promoters. These promoters were disproportionally intronic, showed a complex relationship with gene expression and alternative splicing during memory consolidation and retrieval, and were enriched in the data set relative to known ASD risk genes. Genotyping in a clinical cohort within one of these promoters (SHANK3 promoter 6) revealed that the SNP rs6010065 was associated with ASD. Our data support the idea that learning recapitulates development at the epigenetic level and demonstrate that behaviorally induced epigenetic changes in mice can highlight regulatory regions relevant to brain disorders in patients.
Collapse
Affiliation(s)
- John N Koberstein
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine. Washington State University, Spokane, WA 99202, USA
| | | | - Mathieu E Wimmer
- Department of Psychology and Program in Neuroscience, College of Liberal Arts, Temple University, Philadelphia, PA 19122, USA
| | - Giulia Porcari
- Vanderbilt University Medical School, Nashville, TN 37232, USA
| | - Charlly Kao
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY 10065, USA
| | - Bruce Gomes
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine. Washington State University, Spokane, WA 99202, USA
| | - Davide Risso
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nancy R Zhang
- Department of Statistics, Wharton School, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert T Schultz
- Center for Autism Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ted Abel
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Lucia Peixoto
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine. Washington State University, Spokane, WA 99202, USA.
| |
Collapse
|
22
|
Berridge MJ. Vitamin D deficiency: infertility and neurodevelopmental diseases (attention deficit hyperactivity disorder, autism, and schizophrenia). Am J Physiol Cell Physiol 2017; 314:C135-C151. [PMID: 29070492 DOI: 10.1152/ajpcell.00188.2017] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The process of development depends on a number of signaling systems that regulates the progressive sequence of developmental events. Infertility and neurodevelopmental diseases, such as attention deficit hyperactivity disorder, autism spectrum disorders, and schizophrenia, are caused by specific alterations in these signaling processes. Calcium signaling plays a prominent role throughout development beginning at fertilization and continuing through early development, implantation, and organ differentiation such as heart and brain development. Vitamin D plays a major role in regulating these signaling processes that control development. There is an increase in infertility and an onset of neurodevelopmental diseases when vitamin D is deficient. The way in which vitamin D deficiency acts to alter development is a major feature of this review. One of the primary functions of vitamin D is to maintain the phenotypic stability of both the Ca2+ and redox signaling pathways that play such a key role throughout development.
Collapse
Affiliation(s)
- Michael J Berridge
- Laboratory of Molecular Signalling, The Babraham Institute , Cambridge , United Kingdom
| |
Collapse
|
23
|
Cross-talk between the epigenome and neural circuits in drug addiction. PROGRESS IN BRAIN RESEARCH 2017; 235:19-63. [PMID: 29054289 DOI: 10.1016/bs.pbr.2017.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drug addiction is a behavioral disorder characterized by dysregulated learning about drugs and associated cues that result in compulsive drug seeking and relapse. Learning about drug rewards and predictive cues is a complex process controlled by a computational network of neural connections interacting with transcriptional and molecular mechanisms within each cell to precisely guide behavior. The interplay between rapid, temporally specific neuronal activation, and longer-term changes in transcription is of critical importance in the expression of appropriate, or in the case of drug addiction, inappropriate behaviors. Thus, these factors and their interactions must be considered together, especially in the context of treatment. Understanding the complex interplay between epigenetic gene regulation and circuit connectivity will allow us to formulate novel therapies to normalize maladaptive reward behaviors, with a goal of modulating addictive behaviors, while leaving natural reward-associated behavior unaffected.
Collapse
|
24
|
Bart Martens M, Frega M, Classen J, Epping L, Bijvank E, Benevento M, van Bokhoven H, Tiesinga P, Schubert D, Nadif Kasri N. Euchromatin histone methyltransferase 1 regulates cortical neuronal network development. Sci Rep 2016; 6:35756. [PMID: 27767173 PMCID: PMC5073331 DOI: 10.1038/srep35756] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/05/2016] [Indexed: 12/12/2022] Open
Abstract
Heterozygous mutations or deletions in the human Euchromatin histone methyltransferase 1 (EHMT1) gene cause Kleefstra syndrome, a neurodevelopmental disorder that is characterized by autistic-like features and severe intellectual disability (ID). Neurodevelopmental disorders including ID and autism may be related to deficits in activity-dependent wiring of brain circuits during development. Although Kleefstra syndrome has been associated with dendritic and synaptic defects in mice and Drosophila, little is known about the role of EHMT1 in the development of cortical neuronal networks. Here we used micro-electrode arrays and whole-cell patch-clamp recordings to investigate the impact of EHMT1 deficiency at the network and single cell level. We show that EHMT1 deficiency impaired neural network activity during the transition from uncorrelated background action potential firing to synchronized network bursting. Spontaneous bursting and excitatory synaptic currents were transiently reduced, whereas miniature excitatory postsynaptic currents were not affected. Finally, we show that loss of function of EHMT1 ultimately resulted in less regular network bursting patterns later in development. These data suggest that the developmental impairments observed in EHMT1-deficient networks may result in a temporal misalignment between activity-dependent developmental processes thereby contributing to the pathophysiology of Kleefstra syndrome.
Collapse
Affiliation(s)
- Marijn Bart Martens
- Department of Neuroinformatics, Radboud University Nijmegen, Faculty of Science, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Monica Frega
- Donders Institute for Brain, Cognition and Behaviour, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
- Department of Cognitive Neuroscience, Radboudumc, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Jessica Classen
- Donders Institute for Brain, Cognition and Behaviour, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
- Department of Cognitive Neuroscience, Radboudumc, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Lisa Epping
- Department of Neuroinformatics, Radboud University Nijmegen, Faculty of Science, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Elske Bijvank
- Donders Institute for Brain, Cognition and Behaviour, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
- Department of Cognitive Neuroscience, Radboudumc, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Marco Benevento
- Donders Institute for Brain, Cognition and Behaviour, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
- Department of Cognitive Neuroscience, Radboudumc, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Hans van Bokhoven
- Donders Institute for Brain, Cognition and Behaviour, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
- Department of Cognitive Neuroscience, Radboudumc, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
- Department of Human Genetics, Radboudumc, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Paul Tiesinga
- Department of Neuroinformatics, Radboud University Nijmegen, Faculty of Science, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Dirk Schubert
- Donders Institute for Brain, Cognition and Behaviour, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
- Department of Cognitive Neuroscience, Radboudumc, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Nael Nadif Kasri
- Donders Institute for Brain, Cognition and Behaviour, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
- Department of Cognitive Neuroscience, Radboudumc, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
- Department of Human Genetics, Radboudumc, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| |
Collapse
|
25
|
Montagud-Romero S, Montesinos J, Pascual M, Aguilar MA, Roger-Sanchez C, Guerri C, Miñarro J, Rodríguez-Arias M. `Up-regulation of histone acetylation induced by social defeat mediates the conditioned rewarding effects of cocaine. Prog Neuropsychopharmacol Biol Psychiatry 2016; 70:39-48. [PMID: 27180319 DOI: 10.1016/j.pnpbp.2016.04.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/24/2016] [Accepted: 04/28/2016] [Indexed: 12/16/2022]
Abstract
Social defeat (SD) induces a long-lasting increase in the rewarding effects of psychostimulants measured using the self-administration and conditioned place procedures (CPP). However, little is known about the epigenetic changes induced by social stress and about their role in the increased response to the rewarding effects of psychostimulants. Considering that histone acetylation regulates transcriptional activity and contributes to drug-induced behavioral changes, we addressed the hypothesis that SD induces transcriptional changes by histone modifications associated with the acquisition of place conditioning. After a fourth defeat, H3(K9) acetylation was decreased in the hippocampus, while there was an increase of HAT and a decrease of HDAC levels in the cortex. Three weeks after the last defeat, mice displayed an increase in histone H4(K12) acetylation and an upregulation of histone acetyl transferase (HAT) activity in the hippocampus. In addition, H3(K4)me3, which is closely associated with transcriptional initiation, was also augmented in the hippocampus three weeks after the last defeat. Inhibition of HAT by curcumin (100mg/kg) before each SD blocked the increase in the conditioned reinforcing effects of 1mg/kg of cocaine, while inhibition of HDAC by valproic acid (500mg/kg) before social stress potentiated cocaine-induced CPP. Preference was reinstated when animals received a priming dose of 0.5mg/kg of cocaine, an effect that was absent in untreated defeated mice. These results suggest that the experience of SD induces chromatin remodeling, alters histone acetylation and methylation, and modifies the effects of cocaine on place conditioning. They also point to epigenetic mechanisms as potential avenues leading to new treatments for the long-term effects of social stress on drug addiction.
Collapse
Affiliation(s)
- S Montagud-Romero
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - J Montesinos
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - M Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - M A Aguilar
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - C Roger-Sanchez
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - C Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - J Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - M Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain.
| |
Collapse
|
26
|
Xu S, Panikker P, Iqbal S, Elefant F. Tip60 HAT Action Mediates Environmental Enrichment Induced Cognitive Restoration. PLoS One 2016; 11:e0159623. [PMID: 27454757 PMCID: PMC4959735 DOI: 10.1371/journal.pone.0159623] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/06/2016] [Indexed: 12/14/2022] Open
Abstract
Environmental enrichment (EE) conditions have beneficial effects for reinstating cognitive ability in neuropathological disorders like Alzheimer's disease (AD). While EE benefits involve epigenetic gene control mechanisms that comprise histone acetylation, the histone acetyltransferases (HATs) involved remain largely unknown. Here, we examine a role for Tip60 HAT action in mediating activity- dependent beneficial neuroadaptations to EE using the Drosophila CNS mushroom body (MB) as a well-characterized cognition model. We show that flies raised under EE conditions display enhanced MB axonal outgrowth, synaptic marker protein production, histone acetylation induction and transcriptional activation of cognition linked genes when compared to their genotypically identical siblings raised under isolated conditions. Further, these beneficial changes are impaired in both Tip60 HAT mutant flies and APP neurodegenerative flies. While EE conditions provide some beneficial neuroadaptive changes in the APP neurodegenerative fly MB, such positive changes are significantly enhanced by increasing MB Tip60 HAT levels. Our results implicate Tip60 as a critical mediator of EE-induced benefits, and provide broad insights into synergistic behavioral and epigenetic based therapeutic approaches for treatment of cognitive disorder.
Collapse
Affiliation(s)
- Songjun Xu
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Priyalakshmi Panikker
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Sahira Iqbal
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Felice Elefant
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| |
Collapse
|
27
|
Xu S, Elefant F. Tip off the HAT- Epigenetic control of learning and memory by Drosophila Tip60. Fly (Austin) 2016; 9:22-8. [PMID: 26327426 DOI: 10.1080/19336934.2015.1080887] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Disruption of epigenetic gene control mechanisms involving histone acetylation in the brain causes cognitive impairment, a debilitating hallmark of most neurodegenerative disorders. Histone acetylation regulates cognitive gene expression via chromatin packaging control in neurons. Unfortunately, the histone acetyltransferases (HATs) that generate such neural epigenetic signatures and their mechanisms of action remain unclear. Our recent findings provide insight into this question by demonstrating that Tip60 HAT action is critical for morphology and function of the mushroom body (MB), the learning and memory center in the Drosophila brain. We show that Tip60 is robustly produced in MB Kenyon cells and extending axonal lobes and that targeted MB Tip60 HAT loss results in axonal outgrowth disruption. Functional consequences of loss and gain of Tip60 HAT levels in the MB are evidenced by defects in memory. Tip60 ChIP-Seq analysis reveals enrichment for genes that function in cognitive processes and accordingly, key genes representing these pathways are misregulated in the Tip60 HAT mutant fly brain. Remarkably, increasing levels of Tip60 in the MB rescues learning and memory deficits resulting from Alzheimer's disease associated amyloid precursor protein (APP) induced neurodegeneration. Our studies highlight the potential of HAT activators as a therapeutic option for cognitive disorders.
Collapse
Affiliation(s)
- Songjun Xu
- a Department of Biology ; Drexel University ; Philadelphia , PA USA
| | | |
Collapse
|
28
|
Dynamic methylation driven by neuronal activity in hippocampal neurons impacts complex behavior. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s11515-015-1369-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
29
|
Peixoto LL, Wimmer ME, Poplawski SG, Tudor JC, Kenworthy CA, Liu S, Mizuno K, Garcia BA, Zhang NR, Giese K, Abel T. Memory acquisition and retrieval impact different epigenetic processes that regulate gene expression. BMC Genomics 2015; 16 Suppl 5:S5. [PMID: 26040834 PMCID: PMC4460846 DOI: 10.1186/1471-2164-16-s5-s5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background A fundamental question in neuroscience is how memories are stored and retrieved in the brain. Long-term memory formation requires transcription, translation and epigenetic processes that control gene expression. Thus, characterizing genome-wide the transcriptional changes that occur after memory acquisition and retrieval is of broad interest and importance. Genome-wide technologies are commonly used to interrogate transcriptional changes in discovery-based approaches. Their ability to increase scientific insight beyond traditional candidate gene approaches, however, is usually hindered by batch effects and other sources of unwanted variation, which are particularly hard to control in the study of brain and behavior. Results We examined genome-wide gene expression after contextual conditioning in the mouse hippocampus, a brain region essential for learning and memory, at all the time-points in which inhibiting transcription has been shown to impair memory formation. We show that most of the variance in gene expression is not due to conditioning and that by removing unwanted variance through additional normalization we are able provide novel biological insights. In particular, we show that genes downregulated by memory acquisition and retrieval impact different functions: chromatin assembly and RNA processing, respectively. Levels of histone 2A variant H2AB are reduced only following acquisition, a finding we confirmed using quantitative proteomics. On the other hand, splicing factor Rbfox1 and NMDA receptor-dependent microRNA miR-219 are only downregulated after retrieval, accompanied by an increase in protein levels of miR-219 target CAMKIIγ. Conclusions We provide a thorough characterization of coding and non-coding gene expression during long-term memory formation. We demonstrate that unwanted variance dominates the signal in transcriptional studies of learning and memory and introduce the removal of unwanted variance through normalization as a necessary step for the analysis of genome-wide transcriptional studies in the context of brain and behavior. We show for the first time that histone variants are downregulated after memory acquisition, and splicing factors and microRNAs after memory retrieval. Our results provide mechanistic insights into the molecular basis of cognition by highlighting the differential involvement of epigenetic mechanisms, such as histone variants and post-transcriptional RNA regulation, after acquisition and retrieval of memory.
Collapse
|
30
|
Roth ED, Roth TL, Money KM, SenGupta S, Eason DE, Sweatt JD. DNA methylation regulates neurophysiological spatial representation in memory formation. ACTA ACUST UNITED AC 2015; 2:1-8. [PMID: 25960947 DOI: 10.1016/j.nepig.2015.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Epigenetic mechanisms including altered DNA methylation are critical for altered gene transcription subserving synaptic plasticity and the retention of learned behavior. Here we tested the idea that one role for activity-dependent altered DNA methylation is stabilization of cognition-associated hippocampal place cell firing in response to novel place learning. We observed that a behavioral protocol (spatial exploration of a novel environment) known to induce hippocampal place cell remapping resulted in alterations of hippocampal Bdnf DNA methylation. Further studies using neurophysiological in vivo single unit recordings revealed that pharmacological manipulations of DNA methylation decreased long-term but not short-term place field stability. Together our data highlight a role for DNA methylation in regulating neurophysiological spatial representation and memory formation.
Collapse
Affiliation(s)
- Eric D Roth
- Department of Psychological and Brian Sciences, University of Delaware, Newark, DE 19716 ; Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Tania L Roth
- Department of Psychological and Brian Sciences, University of Delaware, Newark, DE 19716 ; Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Kelli M Money
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Sonda SenGupta
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Dawn E Eason
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - J David Sweatt
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| |
Collapse
|
31
|
Descalzi G, Ikegami D, Ushijima T, Nestler EJ, Zachariou V, Narita M. Epigenetic mechanisms of chronic pain. Trends Neurosci 2015; 38:237-46. [PMID: 25765319 DOI: 10.1016/j.tins.2015.02.001] [Citation(s) in RCA: 270] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/05/2015] [Accepted: 02/09/2015] [Indexed: 12/12/2022]
Abstract
Neuropathic and inflammatory pain promote a large number of persisting adaptations at the cellular and molecular level, allowing even transient tissue or nerve damage to elicit changes in cells that contribute to the development of chronic pain and associated symptoms. There is evidence that injury-induced changes in chromatin structure drive stable changes in gene expression and neural function, which may cause several symptoms, including allodynia, hyperalgesia, anxiety, and depression. Recent findings on epigenetic changes in the spinal cord and brain during chronic pain may guide fundamental advances in new treatments. Here, we provide a brief overview of epigenetic regulation in the nervous system and then discuss the still-limited literature that directly implicates epigenetic modifications in chronic pain syndromes.
Collapse
Affiliation(s)
- Giannina Descalzi
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daigo Ikegami
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Toshikazu Ushijima
- Division of Epigenomics, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan; Life Science Tokyo Advanced Research Center (L-StaR), 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Eric J Nestler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Venetia Zachariou
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Minoru Narita
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan; Life Science Tokyo Advanced Research Center (L-StaR), 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| |
Collapse
|
32
|
Schuch V, Utsumi DA, Costa TVMM, Kulikowski LD, Muszkat M. Attention Deficit Hyperactivity Disorder in the Light of the Epigenetic Paradigm. Front Psychiatry 2015; 6:126. [PMID: 26441687 PMCID: PMC4585002 DOI: 10.3389/fpsyt.2015.00126] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/31/2015] [Indexed: 12/22/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a highly prevalent neurodevelopmental disorder characterized by a definite behavioral pattern that might lead to performance problems in the social, educational, or work environments. In the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, the symptoms of ADHD were restricted to those associated with cognitive (attention deficit) and behavioral (hyperactivity/impulsivity) deficits, while deficient emotional self-regulation, a relevant source of morbidity, was left out. The etiology of it is complex, as its exact causes have not yet been fully elucidated. ADHD seems to arise from a combination of various genetic and environmental factors that alter the developing brain, resulting in structural and functional abnormalities. The aim of this paper was to review epigenetics and ADHD focused on how multidimensional mechanisms influence the behavioral phenotype.
Collapse
Affiliation(s)
- Viviane Schuch
- Núcleo de Atendimento Neuropsicológico Infantil Interdisciplinar (NANI), Centro Paulista de Neuropsicologia, Departamento de Psicobiologia da Universidade Federal de São Paulo - UNIFESP , São Paulo , Brazil
| | - Daniel Augusto Utsumi
- Núcleo de Atendimento Neuropsicológico Infantil Interdisciplinar (NANI), Centro Paulista de Neuropsicologia, Departamento de Psicobiologia da Universidade Federal de São Paulo - UNIFESP , São Paulo , Brazil
| | | | - Leslie Domenici Kulikowski
- Laboratório de Citogenômica, LIM 03, Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo , São Paulo , Brazil
| | - Mauro Muszkat
- Núcleo de Atendimento Neuropsicológico Infantil Interdisciplinar (NANI), Centro Paulista de Neuropsicologia, Departamento de Psicobiologia da Universidade Federal de São Paulo - UNIFESP , São Paulo , Brazil
| |
Collapse
|
33
|
Satterlee JS, Beckel-Mitchener A, Little R, Procaccini D, Rutter JL, Lossie AC. Neuroepigenomics: Resources, Obstacles, and Opportunities. NEUROEPIGENETICS 2015; 1:2-13. [PMID: 25722961 PMCID: PMC4337407 DOI: 10.1016/j.nepig.2014.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Long-lived post-mitotic cells, such as the majority of human neurons, must respond effectively to ongoing changes in neuronal stimulation or microenvironmental cues through transcriptional and epigenomic regulation of gene expression. The role of epigenomic regulation in neuronal function is of fundamental interest to the neuroscience community, as these types of studies have transformed our understanding of gene regulation in post-mitotic cells. This perspective article highlights many of the resources available to researchers interested in neuroepigenomic investigations and discusses some of the current obstacles and opportunities in neuroepigenomics.
Collapse
Affiliation(s)
- John S. Satterlee
- National Institute on Drug Abuse (NIDA), Division of Basic Neurobiology and Behavioral Research, 6001 Executive Boulevard, Bethesda, MD 20850, USA
| | - Andrea Beckel-Mitchener
- National Institute on Mental Health (NIMH), Division of Neuroscience and Basic Behavioral Science, 6001 Executive Boulevard Bethesda, MD 20892-9641, USA
| | - Roger Little
- National Institute on Drug Abuse (NIDA), Division of Basic Neurobiology and Behavioral Research, 6001 Executive Boulevard, Bethesda, MD 20850, USA
| | - Dena Procaccini
- National Institute on Drug Abuse (NIDA), Division of Basic Neurobiology and Behavioral Research, 6001 Executive Boulevard, Bethesda, MD 20850, USA
| | - Joni L. Rutter
- National Institute on Drug Abuse (NIDA), Division of Basic Neurobiology and Behavioral Research, 6001 Executive Boulevard, Bethesda, MD 20850, USA
| | - Amy C. Lossie
- Office of Behavioral and Social Sciences Research (OBSSR), Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director/National Institutes of Health (NIH), 31 Center Drive, Bethesda, MD 20892, USA
| |
Collapse
|
34
|
Abstract
Disruption of epigenetic gene control mechanisms in the brain causes significant cognitive impairment that is a debilitating hallmark of most neurodegenerative disorders, including Alzheimer's disease (AD). Histone acetylation is one of the best characterized of these epigenetic mechanisms that is critical for regulating learning- and memory- associated gene expression profiles, yet the specific histone acetyltransferases (HATs) that mediate these effects have yet to be fully characterized. Here, we investigate an epigenetic role for the HAT Tip60 in learning and memory formation using the Drosophila CNS mushroom body (MB) as a well-characterized cognition model. We show that Tip60 is endogenously expressed in the Kenyon cells, the intrinsic neurons of the MB, and in the MB axonal lobes. Targeted loss of Tip60 HAT activity in the MB causes thinner and shorter axonal lobes while increasing Tip60 HAT levels cause no morphological defects. Functional consequences of both loss and gain of Tip60 HAT levels in the MB are evidenced by defects in immediate-recall memory. Our ChIP-Seq analysis reveals that Tip60 target genes are enriched for functions in cognitive processes, and, accordingly, key genes representing these pathways are misregulated in the Tip60 HAT mutant fly brain. Remarkably, we find that both learning and immediate-recall memory deficits that occur under AD-associated, amyloid precursor protein (APP)-induced neurodegenerative conditions can be effectively rescued by increasing Tip60 HAT levels specifically in the MB. Together, our findings uncover an epigenetic transcriptional regulatory role for Tip60 in cognitive function and highlight the potential of HAT activators as a therapeutic option for neurodegenerative disorders.
Collapse
|
35
|
Jonsson B, Jonsson N. Early environment influences later performance in fishes. JOURNAL OF FISH BIOLOGY 2014; 85:151-88. [PMID: 24961386 DOI: 10.1111/jfb.12432] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 04/28/2014] [Indexed: 05/19/2023]
Abstract
Conditions fish encounter during embryogenesis and early life history can leave lasting effects not only on morphology, but also on growth rate, life-history and behavioural traits. The ecology of offspring can be affected by conditions experienced by their parents and mother in particular. This review summarizes such early impacts and their ecological influences for a variety of teleost species, but with special reference to salmonids. Growth and adult body size, sex ratio, egg size, lifespan and tendency to migrate can all be affected by early influences. Mechanisms behind such phenotypically plastic impacts are not well known, but epigenetic change appears to be one central mechanism. The thermal regime during development and incubation is particularly important, but also early food consumption and intraspecific density can all be responsible for later life-history variation. For behavioural traits, early experiences with effects on brain, sensory development and cognition appear essential. This may also influence boldness and other social behaviours such as mate choice. At the end of the review, several issues and questions for future studies are given.
Collapse
Affiliation(s)
- B Jonsson
- Norwegian Institute for Nature Research, Gaustadalléen 21, N-0349 Oslo, Norway
| | | |
Collapse
|
36
|
The histone acetyltransferase MOF activates hypothalamic polysialylation to prevent diet-induced obesity in mice. Mol Metab 2014; 3:619-29. [PMID: 25161885 PMCID: PMC4142401 DOI: 10.1016/j.molmet.2014.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/27/2014] [Accepted: 05/28/2014] [Indexed: 12/11/2022] Open
Abstract
Overfeeding causes rapid synaptic remodeling in hypothalamus feeding circuits. Polysialylation of cell surface molecules is a key step in this neuronal rewiring and allows normalization of food intake. Here we examined the role of hypothalamic polysialylation in the long-term maintenance of body weight, and deciphered the molecular sequence underlying its nutritional regulation. We found that upon high fat diet (HFD), reduced hypothalamic polysialylation exacerbated the diet-induced obese phenotype in mice. Upon HFD, the histone acetyltransferase MOF was rapidly recruited on the St8sia4 polysialyltransferase-encoding gene. Mof silencing in the mediobasal hypothalamus of adult mice prevented activation of the St8sia4 gene transcription, reduced polysialylation, altered the acute homeostatic feeding response to HFD and increased the body weight gain. These findings indicate that impaired hypothalamic polysialylation contribute to the development of obesity, and establish a role for MOF in the brain control of energy balance.
Collapse
|
37
|
Levina AS, Shiryaeva NV, Vaido AI, Dyuzhikova NA. Effect of NMDA receptor activity on histone H3 methylation and its asymmetry in the hippocampal pyramidal neurons of rats with different excitability thresholds under normal and stress conditions. J EVOL BIOCHEM PHYS+ 2014. [DOI: 10.1134/s0022093013060091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Guo JU, Su Y, Shin JH, Shin J, Li H, Xie B, Zhong C, Hu S, Le T, Fan G, Zhu H, Chang Q, Gao Y, Ming GL, Song H. Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain. Nat Neurosci 2014; 17:215-22. [PMID: 24362762 PMCID: PMC3970219 DOI: 10.1038/nn.3607] [Citation(s) in RCA: 528] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/19/2013] [Indexed: 12/12/2022]
Abstract
DNA methylation has critical roles in the nervous system and has been traditionally considered to be restricted to CpG dinucleotides in metazoan genomes. Here we show that the single base-resolution DNA methylome from adult mouse dentate neurons consists of both CpG (~75%) and CpH (~25%) methylation (H = A/C/T). Neuronal CpH methylation is conserved in human brains, enriched in regions of low CpG density, depleted at protein-DNA interaction sites and anticorrelated with gene expression. Functionally, both methylated CpGs (mCpGs) and mCpHs can repress transcription in vitro and are recognized by methyl-CpG binding protein 2 (MeCP2) in neurons in vivo. Unlike most CpG methylation, CpH methylation is established de novo during neuronal maturation and requires DNA methyltransferase 3A (DNMT3A) for active maintenance in postmitotic neurons. These characteristics of CpH methylation suggest that a substantially expanded proportion of the neuronal genome is under cytosine methylation regulation and provide a new foundation for understanding the role of this key epigenetic modification in the nervous system.
Collapse
Affiliation(s)
- Junjie U. Guo
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yijing Su
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jaehoon Shin
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hongda Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Bin Xie
- Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chun Zhong
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shaohui Hu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thuc Le
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yuan Gao
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guo-li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
39
|
|
40
|
Bókkon I, Vas JP, Császár N, Lukács T. Challenges to free will: transgenerational epigenetic information, unconscious processes, and vanishing twin syndrome. Rev Neurosci 2014; 25:163-75. [DOI: 10.1515/revneuro-2013-0036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 10/17/2013] [Indexed: 01/27/2023]
|
41
|
Simmons RK, Stringfellow SA, Glover ME, Wagle AA, Clinton SM. DNA methylation markers in the postnatal developing rat brain. Brain Res 2013; 1533:26-36. [PMID: 23954679 PMCID: PMC3838910 DOI: 10.1016/j.brainres.2013.08.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 07/30/2013] [Accepted: 08/03/2013] [Indexed: 01/03/2023]
Abstract
In spite of intense interest in how altered epigenetic processes including DNA methylation may contribute to psychiatric and neurodevelopmental disorders, there is a limited understanding of how methylation processes change during early postnatal brain development. The present study used in situ hybridization to assess mRNA expression for the three major DNA methyltranserases (DNMTs)--DNMT1, DNMT3a and DNMT3b--in the developing rat brain at seven developmental timepoints: postnatal days (P) 1, 4, 7, 10, 14, 21, and 75. We also assessed 5-methylcytosine levels (an indicator of global DNA methylation) in selected brain regions during the first three postnatal weeks. DNMT1, DNMT3a and DNMT3b mRNAs are widely expressed throughout the adult and postnatal developing rat brain. Overall, DNMT mRNA levels reached their highest point in the first week of life and gradually decreased over the first three postnatal weeks within the hippocampus, amygdala, striatum, cingulate and lateral septum. Global DNA methylation levels did not follow this developmental pattern; methylation levels gradually increased over the first three postnatal weeks in the hippocampus, and remained stable in the developing amygdala and prefrontal cortex. Our results contribute to a growing understanding of how DNA methylation markers unfold in the developing brain, and highlight how these developmental processes may differ within distinct brain regions.
Collapse
Affiliation(s)
- Rebecca K. Simmons
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sara A. Stringfellow
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Matthew E. Glover
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anjali A. Wagle
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sarah M. Clinton
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
42
|
Bókkon I, Mallick BN, Tuszynski JA. Near death experiences: a multidisciplinary hypothesis. Front Hum Neurosci 2013; 7:533. [PMID: 24062655 PMCID: PMC3769617 DOI: 10.3389/fnhum.2013.00533] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 08/16/2013] [Indexed: 12/16/2022] Open
Abstract
Recently, we proposed a novel biophysical concept regarding on the appearance of brilliant lights during near death experiences (NDEs) (Bókkon and Salari, 2012). Specifically, perceiving brilliant light in NDEs has been proposed to arise due to the reperfusion that produces unregulated overproduction of free radicals and energetically excited molecules that can generate a transient enhancement of bioluminescent biophotons in different areas of the brain, including retinotopic visual areas. If this excess of bioluminescent photon emission exceeds a threshold in retinotopic visual areas, this can appear as (phosphene) lights because the brain interprets these intrinsic retinotopic bioluminescent photons as if they originated from the external physical world. Here, we review relevant literature that reported experimental studies (Imaizumi et al., 1984; Suzuki et al., 1985) that essentially support our previously published conception, i.e., that seeing lights in NDEs may be due to the transient enhancement of bioluminescent biophotons. Next, we briefly describe our biophysical visual representation model that may explain brilliant lights experienced during NDEs (by phosphenes as biophotons) and REM sleep associated dream-like intrinsic visual imageries through biophotons in NDEs. Finally, we link our biophysical visual representation notion to self-consciousness that may involve extremely low-energy quantum entanglements. This article is intended to introduce novel concepts for discussion and does not pretend to give the ultimate explanation for the currently unanswerable questions about matter, life and soul; their creation and their interrelationship.
Collapse
Affiliation(s)
- István Bókkon
- Neuroscience Department, Vision Research Institute Lowell, MA, USA
| | | | | |
Collapse
|
43
|
Meethal SV, Hogan KJ, Mayanil CS, Iskandar BJ. Folate and epigenetic mechanisms in neural tube development and defects. Childs Nerv Syst 2013; 29:1427-33. [PMID: 24013316 DOI: 10.1007/s00381-013-2162-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 05/13/2013] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Multiple genetic and epigenetic factors involved in central nervous system (CNS) development influence the incidence of neural tube defects (NTDs). DISCUSSION The beneficial effect of periconceptional folic acid on NTD prevention denotes a vital role for the single-carbon biochemical pathway in NTD genesis. Indeed, NTDs are associated with polymorphisms in a diversity of genes that encode folate pathway enzymes. Recent evidence suggests that CNS development and function, and consequently NTDs, are also associated with epigenetic mechanisms, many of which participate in the folate cycle and its input and output pathways. We provide an overview with select examples drawn from the authors' research.
Collapse
Affiliation(s)
- Sivan Vadakkadath Meethal
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, WI 53792, USA
| | | | | | | |
Collapse
|
44
|
Activity-dependent NPAS4 expression and the regulation of gene programs underlying plasticity in the central nervous system. Neural Plast 2013; 2013:683909. [PMID: 24024041 PMCID: PMC3759247 DOI: 10.1155/2013/683909] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 07/09/2013] [Indexed: 11/17/2022] Open
Abstract
The capability of the brain to change functionally in response to sensory experience is most active during early stages of development but it decreases later in life when major alterations of neuronal network structures no longer take place in response to experience. This view has been recently challenged by experimental strategies based on the enhancement of environmental stimulation levels, genetic manipulations, and pharmacological treatments, which all have demonstrated that the adult brain retains a degree of plasticity that allows for a rewiring of neuronal circuitries over the entire life course. A hot spot in the field of neuronal plasticity centres on gene programs that underlie plastic phenomena in adulthood. Here, I discuss the role of the recently discovered neuronal-specific and activity-dependent transcription factor NPAS4 as a critical mediator of plasticity in the nervous system. A better understanding of how modifications in the connectivity of neuronal networks occur may shed light on the treatment of pathological conditions such as brain damage or disease in adult life, some of which were once considered untreatable.
Collapse
|
45
|
Interdisciplinary implications on autism, savantism, Asperger syndrome and the biophysical picture representation: Thinking in pictures. COGN SYST RES 2013. [DOI: 10.1016/j.cogsys.2012.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
46
|
Krasnova IN, Chiflikyan M, Justinova Z, McCoy MT, Ladenheim B, Jayanthi S, Quintero C, Brannock C, Barnes C, Adair JE, Lehrmann E, Kobeissy FH, Gold MS, Becker KG, Goldberg SR, Cadet JL. CREB phosphorylation regulates striatal transcriptional responses in the self-administration model of methamphetamine addiction in the rat. Neurobiol Dis 2013; 58:132-43. [PMID: 23726845 DOI: 10.1016/j.nbd.2013.05.009] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/08/2013] [Accepted: 05/20/2013] [Indexed: 10/26/2022] Open
Abstract
Neuroplastic changes in the dorsal striatum participate in the transition from casual to habitual drug use and might play a critical role in the development of methamphetamine (METH) addiction. We examined the influence of METH self-administration on gene and protein expression that may form substrates for METH-induced neuronal plasticity in the dorsal striatum. Male Sprague-Dawley rats self-administered METH (0.1mg/kg/injection, i.v.) or received yoked saline infusions during eight 15-h sessions and were euthanized 2h, 24h, or 1month after cessation of METH exposure. Changes in gene and protein expression were assessed using microarray analysis, RT-PCR and Western blots. Chromatin immunoprecipitation (ChIP) followed by PCR was used to examine epigenetic regulation of METH-induced transcription. METH self-administration caused increases in mRNA expression of the transcription factors, c-fos and fosb, the neurotrophic factor, Bdnf, and the synaptic protein, synaptophysin (Syp) in the dorsal striatum. METH also caused changes in ΔFosB, BDNF and TrkB protein levels, with increases after 2 and 24h, but decreases after 1month of drug abstinence. Importantly, ChIP-PCR showed that METH self-administration caused enrichment of phosphorylated CREB (pCREB), but not of histone H3 trimethylated at lysine 4 (H3K4me3), on promoters of c-fos, fosb, Bdnf and Syp at 2h after cessation of drug intake. These findings show that METH-induced changes in gene expression are mediated, in part, by pCREB-dependent epigenetic phenomena. Thus, METH self-administration might trigger epigenetic changes that mediate alterations in expression of genes and proteins serving as substrates for addiction-related synaptic plasticity.
Collapse
Affiliation(s)
- Irina N Krasnova
- Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
The neuron-specific chromatin regulatory subunit BAF53b is necessary for synaptic plasticity and memory. Nat Neurosci 2013; 16:552-61. [PMID: 23525042 PMCID: PMC3777648 DOI: 10.1038/nn.3359] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 02/11/2013] [Indexed: 12/13/2022]
Abstract
Recent exome sequencing studies have implicated polymorphic BAF complexes (mammalian SWI/SNF chromatin remodeling complexes) in several human intellectual disabilities and cognitive disorders. However, it is currently unknown how mutations in BAF complexes result in impaired cognitive function. Post mitotic neurons express a neuron specific assembly, nBAF, characterized by the neuron-specific subunit BAF53b. Mice harboring selective genetic manipulations of BAF53b have severe defects in longterm memory and long-lasting forms of hippocampal synaptic plasticity. We rescued memory impairments in BAF53b mutant mice by reintroducing BAF53b in the adult hippocampus, indicating a role for BAF53b beyond neuronal development. The defects in BAF53b mutant mice appear to derive from alterations in gene expression that produce abnormal postsynaptic components, such as spine structure and function, and ultimately lead to deficits in synaptic plasticity. Our studies provide new insight into the role of dominant mutations in subunits of BAF complexes in human intellectual and cognitive disorders.
Collapse
|
48
|
Eising E, A Datson N, van den Maagdenberg AMJM, Ferrari MD. Epigenetic mechanisms in migraine: a promising avenue? BMC Med 2013; 11:26. [PMID: 23379668 PMCID: PMC3584973 DOI: 10.1186/1741-7015-11-26] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 02/04/2013] [Indexed: 01/07/2023] Open
Abstract
Migraine is a disabling common brain disorder typically characterized by attacks of severe headache and associated with autonomic and neurological symptoms. Its etiology is far from resolved. This review will focus on evidence that epigenetic mechanisms play an important role in disease etiology. Epigenetics comprise both DNA methylation and post-translational modifications of the tails of histone proteins, affecting chromatin structure and gene expression. Besides playing a role in establishing cellular and developmental stage-specific regulation of gene expression, epigenetic processes are also important for programming lasting cellular responses to environmental signals. Epigenetic mechanisms may explain how non-genetic endogenous and exogenous factors such as female sex hormones, stress hormones and inflammation trigger may modulate attack frequency. Developing drugs that specifically target epigenetic mechanisms may open up exciting new avenues for the prophylactic treatment of migraine.
Collapse
Affiliation(s)
- Else Eising
- Department of Human Genetics, Leiden University Medical Centre, Einthovenweg 20, Leiden 2333 ZC, The Netherlands
| | | | | | | |
Collapse
|
49
|
Social defeat induces changes in histone acetylation and expression of histone modifying enzymes in the ventral hippocampus, prefrontal cortex, and dorsal raphe nucleus. Neuroscience 2013; 264:88-98. [PMID: 23370319 DOI: 10.1016/j.neuroscience.2013.01.024] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 01/09/2013] [Accepted: 01/14/2013] [Indexed: 12/12/2022]
Abstract
Chronic exposure to stress is associated with a number of psychiatric disorders, but little is known about the epigenetic mechanisms that underlie the stress response or resilience to chronic stress. We investigated histone acetylation in seven different brain regions of rats exposed to chronic social defeat stress: the dorsal hippocampus (dHPC), ventral hippocampus (vHPC), medial prefrontal cortex (mPFC), basolateral amygdala (BLA), locus coeruleus (LC), paraventricular thalamus (PVT), and dorsal raphe (DR) nucleus. This stress paradigm was unique in that it allowed rats to display resilience in the form of an active coping mechanism. We found that there was an increase in acetylation of H3K9/14 (H3K9/14ac) and bulk acetylation of H4K5,8,12,16 (H4K5,8,12,16ac) in the DR nucleus of rats that were less resilient. Less resilient rats also displayed increased levels of H3K18 acetylation (H3K18ac) in the mPFC when compared to non-stressed controls. In the vHPC, there was an increase in H3K18ac and H4K12 (H4K12ac) in rats that were less resilient when compared to non-stressed control rats. In addition, there was a decrease in levels of H4K8 acetylation (H4K8ac) in both resilient and non-resilient rats as compared to controls. We assessed expression of histone modifying enzymes in the vHPC and the mPFC using quantitative real-time polymerase chain reaction (PCR) and found changes in expression of a number of targets. These included changes in Sirt1 and Sirt2 in the vHPC and changes in Kat5 in the mPFC. Overall, these results suggest that changes in histone acetylation and expression of histone modifying enzymes in these regions correlate with the behavioral response to stress in socially defeated rats.
Collapse
|
50
|
Abstract
Social plasticity is a ubiquitous feature of animal behaviour. Animals must adjust the expression of their social behaviour to the nuances of daily social life and to the transitions between life-history stages, and the ability to do so affects their Darwinian fitness. Here, an integrative framework is proposed for understanding the proximate mechanisms and ultimate consequences of social plasticity. According to this framework, social plasticity is achieved by rewiring or by biochemically switching nodes of the neural network underlying social behaviour in response to perceived social information. Therefore, at the molecular level, it depends on the social regulation of gene expression, so that different brain genomic and epigenetic states correspond to different behavioural responses and the switches between states are orchestrated by signalling pathways that interface the social environment and the genotype. At the evolutionary scale, social plasticity can be seen as an adaptive trait that can be under positive selection when changes in the environment outpace the rate of genetic evolutionary change. In cases when social plasticity is too costly or incomplete, behavioural consistency can emerge by directional selection that recruits gene modules corresponding to favoured behavioural states in that environment. As a result of this integrative approach, how knowledge of the proximate mechanisms underlying social plasticity is crucial to understanding its costs, limits and evolutionary consequences is shown, thereby highlighting the fact that proximate mechanisms contribute to the dynamics of selection. The role of teleosts as a premier model to study social plasticity is also highlighted, given the diversity and plasticity that this group exhibits in terms of social behaviour. Finally, the proposed integrative framework to social plasticity also illustrates how reciprocal causation analysis of biological phenomena (i.e. considering the interaction between proximate factors and evolutionary explanations) can be a more useful approach than the traditional proximate-ultimate dichotomy, according to which evolutionary processes can be understood without knowledge on proximate causes, thereby black-boxing developmental and physiological mechanisms.
Collapse
Affiliation(s)
- R F Oliveira
- Unidade de Investigação em Eco-Etologia, ISPA - Instituto Universitário, Rua Jardim do Tabaco 34, 1149-041 Lisboa, Portugal.
| |
Collapse
|