401
|
Ito F, Yamada Y, Shigemitsu A, Akinishi M, Kaniwa H, Miyake R, Yamanaka S, Kobayashi H. Role of Oxidative Stress in Epigenetic Modification in Endometriosis. Reprod Sci 2017; 24:1493-1502. [PMID: 28443478 DOI: 10.1177/1933719117704909] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Aberrant DNA methylation and histone modification are associated with an increased risk of reproductive disorders such as endometriosis. However, a cause-effect relationship between epigenetic mechanisms and endometriosis development has not been fully determined. This review provides current information based on oxidative stress in epigenetic modification in endometriosis. This article reviews the English-language literature on epigenetics, DNA methylation, histone modification, and oxidative stress associated with endometriosis in an effort to identify epigenetic modification that causes a predisposition to endometriosis. Oxidative stress, secondary to the influx of hemoglobin, heme, and iron during retrograde menstruation, is involved in the expression of CpG demethylases, ten-eleven translocation, and jumonji (JMJ). Ten-eleven translocation and JMJ recognize a wide range of endogenous DNA methyltransferases (DNMTs). The increased expression levels of DNMTs may be involved in the subsequent downregulation of the decidualization-related genes. This review supports the hypothesis that there are at least 2 distinct phases of epigenetic modification in endometriosis: the initial wave of iron-induced oxidative stress would be followed by the second big wave of epigenetic modulation of endometriosis susceptibility genes. We summarize the recent advances in our understanding of the underlying epigenetic mechanisms focusing on oxidative stress in endometriosis.
Collapse
Affiliation(s)
- Fuminori Ito
- 1 Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Yuki Yamada
- 1 Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Aiko Shigemitsu
- 1 Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Mika Akinishi
- 1 Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Hiroko Kaniwa
- 1 Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Ryuta Miyake
- 1 Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Shoichiro Yamanaka
- 1 Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Hiroshi Kobayashi
- 1 Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| |
Collapse
|
402
|
Rosonina E, Akhter A, Dou Y, Babu J, Sri Theivakadadcham VS. Regulation of transcription factors by sumoylation. Transcription 2017; 8:220-231. [PMID: 28379052 PMCID: PMC5574528 DOI: 10.1080/21541264.2017.1311829] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Transcription factors (TFs) are among the most frequently detected targets of sumoylation, and effects of the modification have been studied for about 200 individual TFs to date. TF sumoylation is most often associated with reduced target gene expression, which can be mediated by enhanced interactions with corepressors or by interference with protein modifications that promote transcription. However, recent studies show that sumoylation also regulates gene expression by controlling the levels of TFs that are associated with chromatin. SUMO can mediate this by modulating TF DNA-binding activity, promoting clearance of TFs from chromatin, or indirectly, by influencing TF abundance or localization.
Collapse
Affiliation(s)
- Emanuel Rosonina
- a Department of Biology , York University , Toronto , ON , Canada
| | - Akhi Akhter
- a Department of Biology , York University , Toronto , ON , Canada
| | - Yimo Dou
- a Department of Biology , York University , Toronto , ON , Canada
| | - John Babu
- a Department of Biology , York University , Toronto , ON , Canada
| | | |
Collapse
|
403
|
Zhao Y, Cao Q, He Y, Xue Q, Xie L, Yan Y. Impairment of endoplasmic reticulum is involved in β-cell dysfunction induced by microcystin-LR. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 223:587-594. [PMID: 28162804 DOI: 10.1016/j.envpol.2017.01.061] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 01/02/2017] [Accepted: 01/21/2017] [Indexed: 06/06/2023]
Abstract
Microcystins (MCs) widely distributed in freshwaters have posed a significant risk to human health. Previous studies have demonstrated that exposure to MC-LR impairs pancreatic islet function, however, the underlying mechanisms still remain unclear. In the present study, we explored the role of endoplasmic reticulum (ER) impairment in β-cell dysfunction caused by MC-LR. The result showed that MC-LR modified ER morphology evidenced by increased ER amount and size at low doses (15, 30 or 60 μM) and vacuolar and dilated ER ultrastructure at high doses (100 or 200 μM). Also, insulin content showed increased at 15 or 30 μM but declined at 60, 100, or 200 μM, which was highly accordant with ER morphological alteration. Transcriptomic analysis identified a number of factors and several pathways associated with ER protein processing, ER stress, apoptosis, and diabetes mellitus in the cells treated with MC-LR compared with non-treated cells. Furthermore, MC-LR-induced ER stress significantly promoted the expression of PERK/eIF2α and their downstream targets (ATF4, CHOP, and Gadd34), which indicates that PERK-eIF2α-ATF4 pathway is involved in MC-LR-induced insulin deficiency. These results suggest that ER impairment is an important contributor to MC-LR-caused β-cell failure and provide a new insight into the association between MCs contamination and the occurrence of human diseases.
Collapse
Affiliation(s)
- Yanyan Zhao
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, 73 East Beijing Road, Nanjing 210008, PR China
| | - Qing Cao
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, 73 East Beijing Road, Nanjing 210008, PR China
| | - Yaojia He
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, PR China
| | - Qingju Xue
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, 73 East Beijing Road, Nanjing 210008, PR China
| | - Liqiang Xie
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, 73 East Beijing Road, Nanjing 210008, PR China.
| | - Yunjun Yan
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, PR China.
| |
Collapse
|
404
|
Pons-Espinal M, de Luca E, Marzi MJ, Beckervordersandforth R, Armirotti A, Nicassio F, Fabel K, Kempermann G, De Pietri Tonelli D. Synergic Functions of miRNAs Determine Neuronal Fate of Adult Neural Stem Cells. Stem Cell Reports 2017; 8:1046-1061. [PMID: 28330621 PMCID: PMC5390108 DOI: 10.1016/j.stemcr.2017.02.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 02/10/2017] [Accepted: 02/10/2017] [Indexed: 02/04/2023] Open
Abstract
Adult neurogenesis requires the precise control of neuronal versus astrocyte lineage determination in neural stem cells. While microRNAs (miRNAs) are critically involved in this step during development, their actions in adult hippocampal neural stem cells (aNSCs) has been unclear. As entry point to address that question we chose DICER, an endoribonuclease essential for miRNA biogenesis and other RNAi-related processes. By specific ablation of Dicer in aNSCs in vivo and in vitro, we demonstrate that miRNAs are required for the generation of new neurons, but not astrocytes, in the adult murine hippocampus. Moreover, we identify 11 miRNAs, of which 9 have not been previously characterized in neurogenesis, that determine neurogenic lineage fate choice of aNSCs at the expense of astrogliogenesis. Finally, we propose that the 11 miRNAs sustain adult hippocampal neurogenesis through synergistic modulation of 26 putative targets from different pathways. Dicer depletion in aNSCs impairs neurogenesis and stimulates astrogliogenesis Synergy of 11 miRNAs sustains neuronal fate of aNSCs miRNA converge on multiple targets in different pathways to induce neurogenesis
Collapse
Affiliation(s)
- Meritxell Pons-Espinal
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Emanuela de Luca
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Matteo Jacopo Marzi
- Center for Genomic Science, Istituto Italiano di Tecnologia, IFOM-IEO CAMPUS, Via Adamello 16, 20139 Milan, Italy
| | - Ruth Beckervordersandforth
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Andrea Armirotti
- D3 PharmaChemistry, Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Francesco Nicassio
- Center for Genomic Science, Istituto Italiano di Tecnologia, IFOM-IEO CAMPUS, Via Adamello 16, 20139 Milan, Italy
| | - Klaus Fabel
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Arnoldstraße 18/18b, 01307 Dresden, Germany; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Arnoldstraße 18/18b, 01307 Dresden, Germany; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Davide De Pietri Tonelli
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy.
| |
Collapse
|
405
|
Liu FG, Hu WF, Wang JL, Wang P, Gong Y, Tong LJ, Jiang B, Zhang W, Qin YB, Chen Z, Yang RR, Huang C. Z-Guggulsterone Produces Antidepressant-Like Effects in Mice through Activation of the BDNF Signaling Pathway. Int J Neuropsychopharmacol 2017; 20:485-497. [PMID: 28339691 PMCID: PMC5458345 DOI: 10.1093/ijnp/pyx009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/17/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Z-guggulsterone, an active compound extracted from the gum resin of the tree Commiphora mukul, has been shown to improve animal memory deficits via activating the brain-derived neurotrophic factor signaling pathway. Here, we investigated the antidepressant-like effect of Z-guggulsterone in a chronic unpredictable stress mouse model of depression. METHODS The effects of Z-guggulsterone were assessed in mice with the tail suspension test and forced swimming test. Z-guggulsterone was also investigated in the chronic unpredictable stress model of depression with fluoxetine as the positive control. Changes in hippocampal neurogenesis as well as the brain-derived neurotrophic factor signaling pathway after chronic unpredictable stress/Z-guggulsterone treatment were investigated. The tryptophan hydroxylase inhibitor and the tyrosine kinase B inhibitor were also used to explore the antidepressant-like mechanisms of Z-guggulsterone. RESULTS Z-guggulsterone (10, 30 mg/kg) administration protected the mice against the chronic unpredictable stress-induced increases in the immobile time in the tail suspension test and forced swimming test and also reversed the reduction in sucrose intake in sucrose preference experiment. Z-guggulsterone (10, 30 mg/kg) administration prevented the reductions in brain-derived neurotrophic factor protein expression levels as well as the phosphorylation levels of cAMP response element binding protein, extracellular signal-regulated kinase 1/2, and protein kinase B in the hippocampus and cortex induced by chronic unpredictable stress. Z-guggulsterone (10, 30 mg/kg) treatment also improved hippocampal neurogenesis in chronic unpredictable stress-treated mice. Blockade of the brain-derived neurotrophic factor signal, but not the monoaminergic system, attenuated the antidepressant-like effects of Z-guggulsterone. CONCLUSIONS Z-guggulsterone exhibits antidepressant activity via activation of the brain-derived neurotrophic factor signaling pathway and upregulation of hippocampal neurogenesis.
Collapse
Affiliation(s)
- Feng-Guo Liu
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Wen-Feng Hu
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Ji-Li Wang
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Peng Wang
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Yu Gong
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Li-Juan Tong
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Bo Jiang
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Wei Zhang
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Yi-Bin Qin
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Zhuo Chen
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Rong-Rong Yang
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| | - Chao Huang
- Department of Neurology, Danyang People’s Hospital, Danyang, Jiangsu, China (Mr Liu); Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Drs Qin and Yang); Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Tong, Jiang, and Zhang); Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong, Jiangsu, China (Drs Huang, Hu, J.-L. Wang, P. Wang, Gong, Tong, Jiang, and Zhang); Invasive Technology Department, Nantong First People’s Hospital, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (Dr Chen)
| |
Collapse
|
406
|
Regulation of mRNA splicing by MeCP2 via epigenetic modifications in the brain. Sci Rep 2017; 7:42790. [PMID: 28211484 PMCID: PMC5314398 DOI: 10.1038/srep42790] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 01/17/2017] [Indexed: 02/08/2023] Open
Abstract
Mutations of X-linked gene Methyl CpG binding protein 2 (MECP2) are the major causes of Rett syndrome (RTT), a severe neurodevelopmental disorder. Duplications of MECP2-containing genomic segments lead to severe autistic symptoms in human. MECP2-coding protein methyl-CpG-binding protein 2 (MeCP2) is involved in transcription regulation, microRNA processing and mRNA splicing. However, molecular mechanisms underlying the involvement of MeCP2 in mRNA splicing in neurons remain largely elusive. In this work we found that the majority of MeCP2-associated proteins are involved in mRNA splicing using mass spectrometry analysis with multiple samples from Mecp2-null rat brain, mouse primary neuron and human cell lines. We further showed that Mecp2 knockdown in cultured cortical neurons led to widespread alternations of mRNA alternative splicing. Analysis of ChIP-seq datasets indicated that MeCP2-regulated exons display specific epigenetic signatures, with DNA modification 5-hydroxymethylcytosine (5hmC) and histone modification H3K4me3 are enriched in down-regulated exons, while the H3K36me3 signature is enriched in exons up-regulated in Mecp2-knockdown neurons comparing to un-affected neurons. Functional analysis reveals that genes containing MeCP2-regulated exons are mainly involved in synaptic functions and mRNA splicing. These results suggested that MeCP2 regulated mRNA splicing through interacting with 5hmC and epigenetic changes in histone markers, and provide functional insights of MeCP2-mediated mRNA splicing in the nervous system.
Collapse
|
407
|
Usui N, Co M, Harper M, Rieger MA, Dougherty JD, Konopka G. Sumoylation of FOXP2 Regulates Motor Function and Vocal Communication Through Purkinje Cell Development. Biol Psychiatry 2017; 81:220-230. [PMID: 27009683 PMCID: PMC4983264 DOI: 10.1016/j.biopsych.2016.02.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 01/29/2016] [Accepted: 02/01/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Mutations in the gene encoding the transcription factor forkhead box P2 (FOXP2) result in brain developmental abnormalities, including reduced gray matter in both human patients and rodent models and speech and language deficits. However, neither the region-specific function of FOXP2 in the brain, in particular the cerebellum, nor the effects of any posttranslational modifications of FOXP2 in the brain and disorders have been explored. METHODS We characterized sumoylation of FOXP2 biochemically and analyzed the region-specific function and sumoylation of FOXP2 in the developing mouse cerebellum. Using in utero electroporation to manipulate the sumoylation state of FOXP2 as well as Foxp2 expression levels in Purkinje cells of the cerebellum in vivo, we reduced Foxp2 expression approximately 40% in the mouse cerebellum. Such a reduction approximates the haploinsufficiency observed in human patients who demonstrate speech and language impairments. RESULTS We identified sumoylation of FOXP2 at K674 (K673 in mice) in the cerebellum of neonates. In vitro co-immunoprecipitation and in vivo colocalization experiments suggest that PIAS3 acts as the small ubiquitin-like modifier E3 ligase for FOXP2 sumoylation. This sumoylation modifies transcriptional regulation by FOXP2. We demonstrated that FOXP2 sumoylation is required for regulation of cerebellar motor function and vocal communication, likely through dendritic outgrowth and arborization of Purkinje cells in the mouse cerebellum. CONCLUSIONS Sumoylation of FOXP2 in neonatal mouse cerebellum regulates Purkinje cell development and motor functions and vocal communication, demonstrating evidence for sumoylation in regulating mammalian behaviors.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390-911, USA
| | - Marissa Co
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390-911, USA
| | - Matthew Harper
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390-911, USA
| | - Michael A. Rieger
- Department of Genetics and Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph D. Dougherty
- Department of Genetics and Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Genevieve Konopka
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
408
|
Abstract
The limited regenerative capacity of neuronal cells requires tight orchestration of cell death and survival regulation in the context of longevity, age-associated diseases as well as during the development of the nervous system. Subordinate to genetic networks epigenetic mechanisms like DNA methylation and histone modifications are involved in the regulation of neuronal development, function and aging. DNA methylation by DNA methyltransferases (DNMTs), mostly correlated with gene silencing, is a dynamic and reversible process. In addition to their canonical actions performing cytosine methylation, DNMTs influence gene expression by interactions with histone modifying enzymes or complexes increasing the complexity of epigenetic transcriptional networks. DNMTs are expressed in neuronal progenitors, post-mitotic as well as adult neurons. In this review, we discuss the role and mode of actions of DNMTs including downstream networks in the regulation of neuronal survival in the developing and aging nervous system and its relevance for associated disorders.
Collapse
Affiliation(s)
- Judit Symmank
- Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Geraldine Zimmer
- Institute of Human Genetics, University Hospital Jena, Jena, Germany
| |
Collapse
|
409
|
Gigek CO, Chen ES, Smith MAC. Methyl-CpG-Binding Protein (MBD) Family: Epigenomic Read-Outs Functions and Roles in Tumorigenesis and Psychiatric Diseases. J Cell Biochem 2016. [PMID: 26205787 DOI: 10.1002/jcb.25281] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epigenetics is the study of the heritable changes on gene expression that are responsible for the regulation of development and that have an impact on several diseases. However, it is of equal importance to understand how epigenetic machinery works. DNA methylation is the most studied epigenetic mark and is generally associated with the regulation of gene expression through the repression of promoter activity and by affecting genome stability. Therefore, the ability of the cell to interpret correct methylation marks and/or the correct interpretation of methylation plays a role in many diseases. The major family of proteins that bind methylated DNA is the methyl-CpG binding domain proteins, or the MBDs. Here, we discuss the structure that makes these proteins a family, the main functions and interactions of all protein family members and their role in human disease such as psychiatric disorders and cancer.
Collapse
Affiliation(s)
- Carolina Oliveira Gigek
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 740, Edifício Leitão da Cunha, 1, ° andar, CEP 04023-900, São Paulo, SP, Brazil.,Disciplina de Gastroenterologia Cirúrgica, Departamento de Cirurgia, Universidade Federal de São Paulo (UNIFESP), R. Napoleão de Barros, 715, 2º andar, CEP:04024-002, São Paulo, Brazil
| | - Elizabeth Suchi Chen
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 740, Edifício Leitão da Cunha, 1, ° andar, CEP 04023-900, São Paulo, SP, Brazil
| | - Marilia Arruda Cardoso Smith
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 740, Edifício Leitão da Cunha, 1, ° andar, CEP 04023-900, São Paulo, SP, Brazil
| |
Collapse
|
410
|
Kim R, Sheaffer KL, Choi I, Won KJ, Kaestner KH. Epigenetic regulation of intestinal stem cells by Tet1-mediated DNA hydroxymethylation. Genes Dev 2016; 30:2433-2442. [PMID: 27856615 PMCID: PMC5131782 DOI: 10.1101/gad.288035.116] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/21/2016] [Indexed: 12/02/2022]
Abstract
Here, Kim et al. provide novel insights into the epigenomic changes that occur during intestinal stem cell development and differentiation. They demonstrate that Tet1-mediated DNA hydroxymethylation plays a critical role in the epigenetic regulation of the Wnt pathway in intestinal stem and progenitor cells and consequently in the self-renewal of the intestinal epithelium. Methylated cytosines are associated with gene silencing. The ten-eleven translocation (TET) hydroxylases, which oxidize methylated cytosines to 5-hydroxymethylcytosine (5hmC), are essential for cytosine demethylation. Gene silencing and activation are critical for intestinal stem cell (ISC) maintenance and differentiation, but the potential role of TET hydroxylases in these processes has not yet been examined. Here, we generated genome-wide maps of the 5hmC mark in ISCs and their differentiated progeny. Genes with high levels of hydroxymethylation in ISCs are strongly associated with Wnt signaling and developmental processes. We found Tet1 to be the most abundantly expressed Tet gene in ISCs; therefore, we analyzed intestinal development in Tet1-deficient mice and determined that these mice are growth-retarded, exhibit partial postnatal lethality, and have significantly reduced numbers of proliferative cells in the intestinal epithelium. In addition, the Tet1-deficient intestine displays reduced organoid-forming capacity. In the Tet1-deficient crypt, decreased expression of Wnt target genes such as Axin2 and Lgr5 correlates with lower 5hmC levels at their promoters. These data demonstrate that Tet1-mediated DNA hydroxymethylation plays a critical role in the epigenetic regulation of the Wnt pathway in intestinal stem and progenitor cells and consequently in the self-renewal of the intestinal epithelium.
Collapse
Affiliation(s)
- Rinho Kim
- Department of Genetics, Center for Molecular Studies in Digestive and Liver Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Karyn L Sheaffer
- Department of Genetics, Center for Molecular Studies in Digestive and Liver Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Inchan Choi
- Department of Genetics, Center for Molecular Studies in Digestive and Liver Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kyoung-Jae Won
- Department of Genetics, Center for Molecular Studies in Digestive and Liver Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Klaus H Kaestner
- Department of Genetics, Center for Molecular Studies in Digestive and Liver Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
411
|
Maiese K. Targeting molecules to medicine with mTOR, autophagy and neurodegenerative disorders. Br J Clin Pharmacol 2016; 82:1245-1266. [PMID: 26469771 PMCID: PMC5061806 DOI: 10.1111/bcp.12804] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 10/11/2015] [Accepted: 10/13/2015] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative disorders are significantly increasing in incidence as the age of the global population continues to climb with improved life expectancy. At present, more than 30 million individuals throughout the world are impacted by acute and chronic neurodegenerative disorders with limited treatment strategies. The mechanistic target of rapamycin (mTOR), also known as the mammalian target of rapamycin, is a 289 kDa serine/threonine protein kinase that offers exciting possibilities for novel treatment strategies for a host of neurodegenerative diseases that include Alzheimer's disease, Parkinson's disease, Huntington's disease, epilepsy, stroke and trauma. mTOR governs the programmed cell death pathways of apoptosis and autophagy that can determine neuronal stem cell development, precursor cell differentiation, cell senescence, cell survival and ultimate cell fate. Coupled to the cellular biology of mTOR are a number of considerations for the development of novel treatments involving the fine control of mTOR signalling, tumourigenesis, complexity of the apoptosis and autophagy relationship, functional outcome in the nervous system, and the intimately linked pathways of growth factors, phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), AMP activated protein kinase (AMPK), silent mating type information regulation two homologue one (Saccharomyces cerevisiae) (SIRT1) and others. Effective clinical translation of the cellular signalling mechanisms of mTOR offers provocative avenues for new drug development in the nervous system tempered only by the need to elucidate further the intricacies of the mTOR pathway.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey, 07101, USA.
| |
Collapse
|
412
|
Chromatin Association of Gcn4 Is Limited by Post-translational Modifications Triggered by its DNA-Binding in Saccharomyces cerevisiae. Genetics 2016; 204:1433-1445. [PMID: 27770033 DOI: 10.1534/genetics.116.194134] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/17/2016] [Indexed: 11/18/2022] Open
Abstract
The Saccharomyces cerevisiae transcription factor Gcn4 is expressed during amino acid starvation, and its abundance is controlled by ubiquitin-mediated proteolysis. Cdk8, a kinase component of the RNA polymerase II Mediator complex, phosphorylates Gcn4, which triggers its ubiquitination/proteolysis, and is thought to link Gcn4 degradation with transcription of target genes. In addition to phosphorylation and ubiquitination, we previously showed that Gcn4 becomes sumoylated in a DNA-binding dependent manner, while a nonsumoylatable form of Gcn4 showed increased chromatin occupancy, but only if Cdk8 was present. To further investigate how the association of Gcn4 with chromatin is regulated, here we examine determinants for Gcn4 sumoylation, and how its post-translational modifications are coordinated. Remarkably, artificially targeting Gcn4 that lacks its DNA binding domain to a heterologous DNA site restores sumoylation at its natural modification sites, indicating that DNA binding is sufficient for the modification to occur in vivo Indeed, we find that neither transcription of target genes nor phosphorylation are required for Gcn4 sumoylation, but blocking its sumoylation alters its phosphorylation and ubiquitination patterns, placing Gcn4 sumoylation upstream of these Cdk8-mediated modifications. Strongly supporting a role for sumoylation in limiting its association with chromatin, a hyper-sumoylated form of Gcn4 shows dramatically reduced DNA occupancy and expression of target genes. Importantly, we find that Cdk8 is at least partly responsible for clearing hyper-sumoylated Gcn4 from DNA, further implicating sumoylation as a stimulus for Cdk8-mediated phosphorylation and degradation. These results support a novel function for SUMO in marking the DNA-bound form of a transcription factor, which triggers downstream processes that limit its association with chromatin, thus preventing uncontrolled expression of target genes.
Collapse
|
413
|
Meinke MC, Schanzer S, Lohan SB, Shchatsinin I, Darvin ME, Vollert H, Magnussen B, Köcher W, Helfmann J, Lademann J. Comparison of different cutaneous carotenoid sensors and influence of age, skin type, and kinetic changes subsequent to intake of a vegetable extract. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:107002. [PMID: 27762423 DOI: 10.1117/1.jbo.21.10.107002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/29/2016] [Indexed: 06/06/2023]
Abstract
In the last decade, cutaneous carotenoid measurements have become increasingly popular, as carotenoids were found to be a biomarker of nutrition rich in fruits and vegetables, permitting monitoring of the influence of various stress factors. For such measurements, in addition to the specific and selective resonance Raman spectroscopy (RRS), newly developed low expensive small and mobile sensors that are based on spatially resolved reflectance spectroscopy (SRRS) are used for cutaneous carotenoid measurements. Human volunteers of different age exhibiting skin types I to III were investigated using RRS and two SRRS-based sensors to determine the influence of these parameters on the measuring results. In two studies on volunteers of either the same age or skin type, however, the respective other parameter being varied and no significant influences of age or skin type could be detected. Furthermore, the kinetic changes resulting from the intake and discontinued intake of a vegetable extract rich in carotenoids showed a good correlation among the three sensors and with the detected blood carotenoids. This illustrates that the SRRS-based sensors and RRS device provide reliable cutaneous carotenoid values independent of age and skin types I to III of the volunteers.
Collapse
Affiliation(s)
- Martina C Meinke
- Charité - Universitätsmedizin Berlin, Department of Dermatology, Venerology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Sabine Schanzer
- Charité - Universitätsmedizin Berlin, Department of Dermatology, Venerology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Silke B Lohan
- Charité - Universitätsmedizin Berlin, Department of Dermatology, Venerology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Ihar Shchatsinin
- Laser- und Medizin Technologie GmbH Berlin, Fabeckstr. 60-62, 14195 Berlin, Germany
| | - Maxim E Darvin
- Charité - Universitätsmedizin Berlin, Department of Dermatology, Venerology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Henning Vollert
- BioActive Food GmbH, Am Ihlsee 36a, 23795 Bad Segeberg, Germany
| | - Björn Magnussen
- Opsolution GmbH, Wilhelmshöher Allee 273a, 34131 Kassel, Germany
| | - Wolfang Köcher
- Opsolution GmbH, Wilhelmshöher Allee 273a, 34131 Kassel, Germany
| | - Jürgen Helfmann
- Laser- und Medizin Technologie GmbH Berlin, Fabeckstr. 60-62, 14195 Berlin, Germany
| | - Jürgen Lademann
- Charité - Universitätsmedizin Berlin, Department of Dermatology, Venerology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
414
|
Gupta T, Morgan HR, Bailey JA, Certel SJ. Functional conservation of MBD proteins: MeCP2 and Drosophila MBD proteins alter sleep. GENES BRAIN AND BEHAVIOR 2016; 15:757-774. [PMID: 27489246 DOI: 10.1111/gbb.12314] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 01/01/2023]
Abstract
Proteins containing a methyl-CpG-binding domain (MBD) bind 5mC and convert the methylation pattern information into appropriate functional cellular states. The correct readout of epigenetic marks is of particular importance in the nervous system where abnormal expression or compromised MBD protein function, can lead to disease and developmental disorders. Recent evidence indicates that the genome of Drosophila melanogaster is methylated and two MBD proteins, dMBD2/3 and dMBD-R2, are present. Are Drosophila MBD proteins required for neuronal function, and as MBD-containing proteins have diverged and evolved, does the MBD domain retain the molecular properties required for conserved cellular function across species? To address these questions, we expressed the human MBD-containing protein, hMeCP2, in distinct amine neurons and quantified functional changes in sleep circuitry output using a high throughput assay in Drosophila. hMeCP2 expression resulted in phase-specific sleep loss and sleep fragmentation with the hMeCP2-mediated sleep deficits requiring an intact MBD domain. Reducing endogenous dMBD2/3 and dMBD-R2 levels also generated sleep fragmentation, with an increase in sleep occurring upon dMBD-R2 reduction. To examine if hMeCP2 and dMBD-R2 are targeting common neuronal functions, we reduced dMBD-R2 levels in combination with hMeCP2 expression and observed a complete rescue of sleep deficits. Furthermore, chromosomal binding experiments indicate MBD-R2 and MeCP2 associate on shared genomic loci. Our results provide the first demonstration that Drosophila MBD-containing family members are required for neuronal function and suggest that the MBD domain retains considerable functional conservation at the whole organism level across species.
Collapse
Affiliation(s)
- T Gupta
- Neuroscience Graduate Program
| | - H R Morgan
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, USA
| | - J A Bailey
- Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, USA
| | - S J Certel
- Neuroscience Graduate Program.,Division of Biological Sciences and Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, USA
| |
Collapse
|
415
|
Kurian JR, Louis S, Keen KL, Wolfe A, Terasawa E, Levine JE. The Methylcytosine Dioxygenase Ten-Eleven Translocase-2 (tet2) Enables Elevated GnRH Gene Expression and Maintenance of Male Reproductive Function. Endocrinology 2016; 157:3588-603. [PMID: 27384303 PMCID: PMC5007894 DOI: 10.1210/en.2016-1087] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reproduction depends on the establishment and maintenance of elevated GnRH neurosecretion. The elevation of primate GnRH release is accompanied by epigenetic changes. Specifically, cytosine residues within the GnRH gene promoter are actively demethylated, whereas GnRH mRNA levels and peptide release rise. Whether active DNA demethylation has an impact on GnRH neuron development and consequently reproductive function remains unknown. In this study, we investigated whether ten-eleven translocation (tet) enzymes, which initiate the process of active DNA demethylation, influence neuronal function and reproduction. We found that tet2 expression increases with age in the developing mouse preoptic area-hypothalamus and is substantially higher in a mature (GT1-7) than an immature (GN11) GnRH cell line. GnRH mRNA levels and mean GnRH peptide release elevated after overexpression of tet2 in GN11 cells, whereas CRISPR/cas9-mediated knockdown of tet2 in GT1-7 cells led to a significant decline in GnRH expression. Manipulations of tet2 expression altered tet2 genome binding and histone 3 lysine 4 trimethylation abundance at the GnRH promoter. Mice with selective disruption of tet2 in GnRH neurons (GnRH-specific tet2 knockout mice) exhibited no sign of altered pubertal timing in either sex, although plasma LH levels were significantly lower, and fecundity was altered specifically in adult male GnRH-specific tet2 knockout animals, indicating that tet2 may participate in the maintenance GnRH neuronal function. Exposure to bisphenol A, an environmental contaminant that alters GnRH neuron activity, caused a shift in tet2 subcellular localization and a decrease in histone 3 lysine 4 trimethylation abundance at the GnRH promoter. Finally, evaluation of tet2 protein interactions in GT1-7 cells suggests that the influence of tet2 on neuronal function are not limited to nuclear mechanisms but could depend on mitochondrial function, and RNA metabolism. Together, these studies implicate tet2 in the maintenance of GnRH neuronal function and neuroendocrine control of male reproduction.
Collapse
Affiliation(s)
- Joseph R Kurian
- Department of Obstetrics and Gynecology (J.R.K., S.L.), Southern Illinois University School of Medicine, Springfield, Illinois 62794; St. John's Hospital Carol Jo Vecchie Women and Children's Center (J.R.K.), Springfield, Illinois 62769; Wisconsin National Primate Research Center (K.L.K., E.T., J.E.L.), Madison, Wisconsin 53705; Department of Pediatrics and Physiology (A.W.), The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Departments of Pediatrics (E.T.) and Neuroscience (J.E.L.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Somaja Louis
- Department of Obstetrics and Gynecology (J.R.K., S.L.), Southern Illinois University School of Medicine, Springfield, Illinois 62794; St. John's Hospital Carol Jo Vecchie Women and Children's Center (J.R.K.), Springfield, Illinois 62769; Wisconsin National Primate Research Center (K.L.K., E.T., J.E.L.), Madison, Wisconsin 53705; Department of Pediatrics and Physiology (A.W.), The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Departments of Pediatrics (E.T.) and Neuroscience (J.E.L.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Kim L Keen
- Department of Obstetrics and Gynecology (J.R.K., S.L.), Southern Illinois University School of Medicine, Springfield, Illinois 62794; St. John's Hospital Carol Jo Vecchie Women and Children's Center (J.R.K.), Springfield, Illinois 62769; Wisconsin National Primate Research Center (K.L.K., E.T., J.E.L.), Madison, Wisconsin 53705; Department of Pediatrics and Physiology (A.W.), The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Departments of Pediatrics (E.T.) and Neuroscience (J.E.L.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Andrew Wolfe
- Department of Obstetrics and Gynecology (J.R.K., S.L.), Southern Illinois University School of Medicine, Springfield, Illinois 62794; St. John's Hospital Carol Jo Vecchie Women and Children's Center (J.R.K.), Springfield, Illinois 62769; Wisconsin National Primate Research Center (K.L.K., E.T., J.E.L.), Madison, Wisconsin 53705; Department of Pediatrics and Physiology (A.W.), The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Departments of Pediatrics (E.T.) and Neuroscience (J.E.L.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Ei Terasawa
- Department of Obstetrics and Gynecology (J.R.K., S.L.), Southern Illinois University School of Medicine, Springfield, Illinois 62794; St. John's Hospital Carol Jo Vecchie Women and Children's Center (J.R.K.), Springfield, Illinois 62769; Wisconsin National Primate Research Center (K.L.K., E.T., J.E.L.), Madison, Wisconsin 53705; Department of Pediatrics and Physiology (A.W.), The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Departments of Pediatrics (E.T.) and Neuroscience (J.E.L.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Jon E Levine
- Department of Obstetrics and Gynecology (J.R.K., S.L.), Southern Illinois University School of Medicine, Springfield, Illinois 62794; St. John's Hospital Carol Jo Vecchie Women and Children's Center (J.R.K.), Springfield, Illinois 62769; Wisconsin National Primate Research Center (K.L.K., E.T., J.E.L.), Madison, Wisconsin 53705; Department of Pediatrics and Physiology (A.W.), The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Departments of Pediatrics (E.T.) and Neuroscience (J.E.L.), University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706
| |
Collapse
|
416
|
Maison C, Bailly D, Quivy JP, Almouzni G. The methyltransferase Suv39h1 links the SUMO pathway to HP1α marking at pericentric heterochromatin. Nat Commun 2016; 7:12224. [PMID: 27426629 PMCID: PMC4960310 DOI: 10.1038/ncomms12224] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/10/2016] [Indexed: 02/03/2023] Open
Abstract
The trimethylation of histone H3 on lysine 9 (H3K9me3) – a mark recognized by HP1 that depends on the Suv39h lysine methyltransferases (KMTs) – has provided a basis for the reader/writer model to explain HP1 accumulation at pericentric heterochromatin in mammals. Here, we identify the Suv39h1 paralog, as a unique enhancer of HP1α sumoylation both in vitro and in vivo. The region responsible for promoting HP1α sumoylation (aa1–167) is distinct from the KMT catalytic domain and mediates binding to Ubc9. Tethering the 1–167 domain of Suv39h1 to pericentric heterochromatin, but not mutants unable to bind Ubc9, accelerates the de novo targeting of HP1α to these domains. Our results establish an unexpected feature of Suv39h1, distinct from the KMT activity, with a major role for heterochromatin formation. We discuss how linking Suv39h1 to the SUMO pathway provides conceptual implications for our general view on nuclear domain organization and physiological functions. The Suv39h histone methyltransferases promote trimethylation of histone H3 on lysine 9 (H3K9me3). Here, in the Suv39h1 paralog, the authors identify an enhancer of HP1a sumoylation activity that impacts heterochromatin.
Collapse
Affiliation(s)
- Christèle Maison
- Institut Curie, PSL Research University, CNRS, UMR3664, Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR3664, F-75005 Paris, France
| | - Delphine Bailly
- Institut Curie, PSL Research University, CNRS, UMR3664, Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR3664, F-75005 Paris, France
| | - Jean-Pierre Quivy
- Institut Curie, PSL Research University, CNRS, UMR3664, Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR3664, F-75005 Paris, France
| | - Geneviève Almouzni
- Institut Curie, PSL Research University, CNRS, UMR3664, Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR3664, F-75005 Paris, France
| |
Collapse
|
417
|
Ludwig AK, Zhang P, Cardoso MC. Modifiers and Readers of DNA Modifications and Their Impact on Genome Structure, Expression, and Stability in Disease. Front Genet 2016; 7:115. [PMID: 27446199 PMCID: PMC4914596 DOI: 10.3389/fgene.2016.00115] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 06/06/2016] [Indexed: 12/16/2022] Open
Abstract
Cytosine base modifications in mammals underwent a recent expansion with the addition of several naturally occurring further modifications of methylcytosine in the last years. This expansion was accompanied by the identification of the respective enzymes and proteins reading and translating the different modifications into chromatin higher order organization as well as genome activity and stability, leading to the hypothesis of a cytosine code. Here, we summarize the current state-of-the-art on DNA modifications, the enzyme families setting the cytosine modifications and the protein families reading and translating the different modifications with emphasis on the mouse protein homologs. Throughout this review, we focus on functional and mechanistic studies performed on mammalian cells, corresponding mouse models and associated human diseases.
Collapse
Affiliation(s)
- Anne K Ludwig
- Cell Biology and Epigenetics, Department of Biology, Technische Universität Darmstadt, Darmstadt Germany
| | - Peng Zhang
- Cell Biology and Epigenetics, Department of Biology, Technische Universität Darmstadt, Darmstadt Germany
| | - M C Cardoso
- Cell Biology and Epigenetics, Department of Biology, Technische Universität Darmstadt, Darmstadt Germany
| |
Collapse
|
418
|
Abstract
Oxidative stress has a significant impact on the development and progression of common human pathologies, including cancer, diabetes, hypertension and neurodegenerative diseases. Increasing evidence suggests that oxidative stress globally influences chromatin structure, DNA methylation, enzymatic and non-enzymatic post-translational modifications of histones and DNA-binding proteins. The effects of oxidative stress on these chromatin alterations mediate a number of cellular changes, including modulation of gene expression, cell death, cell survival and mutagenesis, which are disease-driving mechanisms in human pathologies. Targeting oxidative stress-dependent pathways is thus a promising strategy for the prevention and treatment of these diseases. We summarize recent research developments connecting oxidative stress and chromatin regulation.
Collapse
Affiliation(s)
- Sarah Kreuz
- King Abdullah University of Science & Technology (KAUST), Environmental Epigenetics Program, Thuwal 23955-6900, Saudi Arabia
| | - Wolfgang Fischle
- King Abdullah University of Science & Technology (KAUST), Environmental Epigenetics Program, Thuwal 23955-6900, Saudi Arabia
| |
Collapse
|
419
|
Della Ragione F, Vacca M, Fioriniello S, Pepe G, D'Esposito M. MECP2, a multi-talented modulator of chromatin architecture. Brief Funct Genomics 2016; 15:420-431. [PMID: 27296483 DOI: 10.1093/bfgp/elw023] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
It has been a long trip from 1992, the year of the discovery of MECP2, to the present day. What is surprising is that some of the pivotal roles of MeCP2 were already postulated at that time, such as repression of inappropriate expression from repetitive elements and the regulation of pericentric heterochromatin condensation. However, MeCP2 performs many more functions. MeCP2 is a reader of epigenetic information contained in methylated (and hydroxymethylated) DNA, moving from the 'classical' CpG doublet to the more complex view addressed by the non-CpG methylation, which is a feature of the postnatal brain. MECP2 is a transcriptional repressor, although when it forms complexes with the appropriate molecules, it can become a transcriptional activator. For all of these aspects, Rett syndrome, which is caused by MECP2 mutations, is considered a paradigmatic example of a 'chromatin disorder'. Even if the hunt for bona-fide MECP2 target genes is far from concluded today, the role of MeCP2 in the maintenance of chromatin architecture appears to be clearly established. Taking a cue from the non-scientific literature, we can firmly attest that MeCP2 is a player with 'a great future behind it'*.*V. Gassmann 'Un grande avvenire dietro le spalle'. TEA Eds.
Collapse
|
420
|
Zhao H, Han Z, Ji X, Luo Y. Epigenetic Regulation of Oxidative Stress in Ischemic Stroke. Aging Dis 2016; 7:295-306. [PMID: 27330844 PMCID: PMC4898926 DOI: 10.14336/ad.2015.1009] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/09/2015] [Indexed: 12/11/2022] Open
Abstract
The prevalence and incidence of stroke rises with life expectancy. However, except for the use of recombinant tissue-type plasminogen activator, the translation of new therapies for acute stroke from animal models into humans has been relatively unsuccessful. Oxidative DNA and protein damage following stroke is typically associated with cell death. Cause-effect relationships between reactive oxygen species and epigenetic modifications have been established in aging, cancer, acute pancreatitis, and fatty liver disease. In addition, epigenetic regulatory mechanisms during stroke recovery have been reviewed, with focuses mainly on neural apoptosis, necrosis, and neuroplasticity. However, oxidative stress-induced epigenetic regulation in vascular neural networks following stroke has not been sufficiently explored. Improved understanding of the epigenetic regulatory network upon oxidative stress may provide effective antioxidant approaches for treating stroke. In this review, we summarize the epigenetic events, including DNA methylation, histone modification, and microRNAs, that result from oxidative stress following experimental stroke in animal and cell models, and the ways in which epigenetic changes and their crosstalk influence the redox state in neurons, glia, and vascular endothelial cells, helping us to understand the foregone and vicious epigenetic regulation of oxidative stress in the vascular neural network following stroke.
Collapse
Affiliation(s)
- Haiping Zhao
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Ziping Han
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- 22Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yumin Luo
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; 3Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
| |
Collapse
|
421
|
Long-Term Memory in Drosophila Is Influenced by Histone Deacetylase HDAC4 Interacting with SUMO-Conjugating Enzyme Ubc9. Genetics 2016; 203:1249-64. [PMID: 27182943 DOI: 10.1534/genetics.115.183194] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 04/29/2016] [Indexed: 12/28/2022] Open
Abstract
HDAC4 is a potent memory repressor with overexpression of wild type or a nuclear-restricted mutant resulting in memory deficits. Interestingly, reduction of HDAC4 also impairs memory via an as yet unknown mechanism. Although histone deacetylase family members are important mediators of epigenetic mechanisms in neurons, HDAC4 is predominantly cytoplasmic in the brain and there is increasing evidence for interactions with nonhistone proteins, suggesting HDAC4 has roles beyond transcriptional regulation. To that end, we performed a genetic interaction screen in Drosophila and identified 26 genes that interacted with HDAC4, including Ubc9, the sole SUMO E2-conjugating enzyme. RNA interference-induced reduction of Ubc9 in the adult brain impaired long-term memory in the courtship suppression assay, a Drosophila model of associative memory. We also demonstrate that HDAC4 and Ubc9 interact genetically during memory formation, opening new avenues for investigating the mechanisms through which HDAC4 regulates memory formation and other neurological processes.
Collapse
|
422
|
Wehling-Henricks M, Li Z, Lindsey C, Wang Y, Welc SS, Ramos JN, Khanlou N, Kuro-O M, Tidball JG. Klotho gene silencing promotes pathology in the mdx mouse model of Duchenne muscular dystrophy. Hum Mol Genet 2016; 25:2465-2482. [PMID: 27154199 DOI: 10.1093/hmg/ddw111] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/10/2016] [Accepted: 04/08/2016] [Indexed: 11/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal muscle disease involving progressive loss of muscle regenerative capacity and increased fibrosis. We tested whether epigenetic silencing of the klotho gene occurs in the mdx mouse model of DMD and whether klotho silencing is an important feature of the disease. Our findings show that klotho undergoes muscle-specific silencing at the acute onset of mdx pathology. Klotho experiences increased methylation of CpG sites in its promoter region, which is associated with gene silencing, and increases in a repressive histone mark, H3K9me2. Expression of a klotho transgene in mdx mice restored their longevity, reduced muscle wasting, improved function and greatly increased the pool of muscle-resident stem cells required for regeneration. Reductions of fibrosis in late, progressive stages of the mdx pathology achieved by transgene expression were paralleled by reduced expression of Wnt target genes (axin-2), transforming growth factor-beta (TGF-β1) and collagens types 1 and 3, indicating that Klotho inhibition of the profibrotic Wnt/TGFβ axis underlies its anti-fibrotic effect in aging, dystrophic muscle. Thus, epigenetic silencing of klotho during muscular dystrophy contributes substantially to lost regenerative capacity and increased fibrosis of dystrophic muscle during late progressive stages of the disease.
Collapse
Affiliation(s)
| | - Zhenzhi Li
- Department of Integrative Biology and Physiology
| | | | - Ying Wang
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095-1606, USA
| | | | | | - Négar Khanlou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1732, USA
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - James G Tidball
- Department of Integrative Biology and Physiology .,Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095-1606, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1732, USA
| |
Collapse
|
423
|
Linscott ML, Chung WCJ. Fibroblast Growth Factor 8 Expression in GT1-7 GnRH-Secreting Neurons Is Androgen-Independent, but Can Be Upregulated by the Inhibition of DNA Methyltransferases. Front Cell Dev Biol 2016; 4:34. [PMID: 27200347 PMCID: PMC4853385 DOI: 10.3389/fcell.2016.00034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/13/2016] [Indexed: 11/13/2022] Open
Abstract
Fibroblast growth factor 8 (FGF8) is a potent morphogen that regulates the embryonic development of hypothalamic neuroendocrine cells. Indeed, using Fgf8 hypomorphic mice, we showed that reduced Fgf8 mRNA expression completely eliminated the presence of gonadotropin-releasing hormone (GnRH) neurons. These findings suggest that FGF8 signaling is required during the embryonic development of mouse GnRH neurons. Additionally, in situ hybridization studies showed that the embryonic primordial birth place of GnRH neurons, the olfactory placode, is highly enriched for Fgf8 mRNA expression. Taken together these data underscore the importance of FGF8 signaling for GnRH emergence. However, an important question remains unanswered: How is Fgf8 gene expression regulated in the developing embryonic mouse brain? One major candidate is the androgen receptor (AR), which has been shown to upregulate Fgf8 mRNA in 60-70% of newly diagnosed prostate cancers. Therefore, we hypothesized that ARs may be involved in the regulation of Fgf8 transcription in the developing mouse brain. To test this hypothesis, we used chromatin-immunoprecipitation (ChIP) assays to elucidate whether ARs interact with the 5'UTR region upstream of the translational start site of the Fgf8 gene in immortalized mouse GnRH neurons (GT1-7) and nasal explants. Our data showed that while AR interacts with the Fgf8 promoter region, this interaction was androgen-independent, and that androgen treatment did not affect Fgf8 mRNA levels, indicating that androgen signaling does not induce Fgf8 transcription. In contrast, inhibition of DNA methyltransferases (DNMT) significantly upregulated Fgf8 mRNA levels indicating that Fgf8 transcriptional activity may be dependent on DNA methylation status.
Collapse
Affiliation(s)
- Megan L Linscott
- Department of Biological Sciences, Kent State University Kent, OH, USA
| | - Wilson C J Chung
- Department of Biological Sciences, Kent State UniversityKent, OH, USA; School of Biomedical Sciences, Kent State UniversityKent, OH, USA
| |
Collapse
|
424
|
Maiese K. Novel nervous and multi-system regenerative therapeutic strategies for diabetes mellitus with mTOR. Neural Regen Res 2016; 11:372-85. [PMID: 27127460 PMCID: PMC4828986 DOI: 10.4103/1673-5374.179032] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Throughout the globe, diabetes mellitus (DM) is increasing in incidence with limited therapies presently available to prevent or resolve the significant complications of this disorder. DM impacts multiple organs and affects all components of the central and peripheral nervous systems that can range from dementia to diabetic neuropathy. The mechanistic target of rapamycin (mTOR) is a promising agent for the development of novel regenerative strategies for the treatment of DM. mTOR and its related signaling pathways impact multiple metabolic parameters that include cellular metabolic homeostasis, insulin resistance, insulin secretion, stem cell proliferation and differentiation, pancreatic β-cell function, and programmed cell death with apoptosis and autophagy. mTOR is central element for the protein complexes mTOR Complex 1 (mTORC1) and mTOR Complex 2 (mTORC2) and is a critical component for a number of signaling pathways that involve phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), AMP activated protein kinase (AMPK), silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), Wnt1 inducible signaling pathway protein 1 (WISP1), and growth factors. As a result, mTOR represents an exciting target to offer new clinical avenues for the treatment of DM and the complications of this disease. Future studies directed to elucidate the delicate balance mTOR holds over cellular metabolism and the impact of its broad signaling pathways should foster the translation of these targets into effective clinical regimens for DM.
Collapse
|
425
|
Schorova L, Martin S. Sumoylation in Synaptic Function and Dysfunction. Front Synaptic Neurosci 2016; 8:9. [PMID: 27199730 PMCID: PMC4848311 DOI: 10.3389/fnsyn.2016.00009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/08/2016] [Indexed: 12/18/2022] Open
Abstract
Sumoylation has recently emerged as a key post-translational modification involved in many, if not all, biological processes. Small Ubiquitin-like Modifier (SUMO) polypeptides are covalently attached to specific lysine residues of target proteins through a dedicated enzymatic pathway. Disruption of the SUMO enzymatic pathway in the developing brain leads to lethality indicating that this process exerts a central role during embryonic and post-natal development. However, little is still known regarding how this highly dynamic protein modification is regulated in the mammalian brain despite an increasing number of data implicating sumoylated substrates in synapse formation, synaptic communication and plasticity. The aim of this review is therefore to briefly describe the enzymatic SUMO pathway and to give an overview of our current knowledge on the function and dysfunction of protein sumoylation at the mammalian synapse.
Collapse
Affiliation(s)
- Lenka Schorova
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (UMR7275), University of Nice-Sophia-Antipolis, Laboratory of Excellence "Network for Innovation on Signal Transduction, Pathways in Life Sciences" Valbonne, France
| | - Stéphane Martin
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (UMR7275), University of Nice-Sophia-Antipolis, Laboratory of Excellence "Network for Innovation on Signal Transduction, Pathways in Life Sciences" Valbonne, France
| |
Collapse
|
426
|
Bueno D, Garcia-Fernàndez J. Evolutionary development of embryonic cerebrospinal fluid composition and regulation: an open research field with implications for brain development and function. Fluids Barriers CNS 2016; 13:5. [PMID: 26979569 PMCID: PMC4793645 DOI: 10.1186/s12987-016-0029-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/10/2016] [Indexed: 12/29/2022] Open
Abstract
Within the consolidated field of evolutionary development, there is emerging research on evolutionary aspects of central nervous system development and its implications for adult brain structure and function, including behaviour. The central nervous system is one of the most intriguing systems in complex metazoans, as it controls all body and mind functions. Its failure is responsible for a number of severe and largely incurable diseases, including neurological and neurodegenerative ones. Moreover, the evolution of the nervous system is thought to be a critical step in the adaptive radiation of vertebrates. Brain formation is initiated early during development. Most embryological, genetic and evolutionary studies have focused on brain neurogenesis and regionalisation, including the formation and function of organising centres, and the comparison of homolog gene expression and function among model organisms from different taxa. The architecture of the vertebrate brain primordium also reveals the existence of connected internal cavities, the cephalic vesicles, which in fetuses and adults become the ventricular system of the brain. During embryonic and fetal development, brain cavities and ventricles are filled with a complex, protein-rich fluid called cerebrospinal fluid (CSF). However, CSF has not been widely analysed from either an embryological or evolutionary perspective. Recently, it has been demonstrated in higher vertebrates that embryonic cerebrospinal fluid has key functions in delivering diffusible signals and nutrients to the developing brain, thus contributing to the proliferation, differentiation and survival of neural progenitor cells, and to the expansion and patterning of the brain. Moreover, it has been shown that the composition and homeostasis of CSF are tightly controlled in a time-dependent manner from the closure of the anterior neuropore, just before the initiation of primary neurogenesis, up to the formation of functional choroid plexuses. In this review, we draw together existing literature about the formation, function and homeostatic regulation of embryonic cerebrospinal fluid, from the closure of the anterior neuropore to the formation of functional fetal choroid plexuses, from an evolutionary perspective. The relevance of these processes to the normal functions and diseases of adult brain will also be discussed.
Collapse
Affiliation(s)
- David Bueno
- Department of Genetics, Microbiology and Statistics, Unit of Biomedical, Evolutionary and Developmental Genetics, Faculty of Biological Sciences, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Catalonia, Spain.
| | - Jordi Garcia-Fernàndez
- Department of Genetics, Microbiology and Statistics, Unit of Biomedical, Evolutionary and Developmental Genetics, Faculty of Biological Sciences, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Catalonia, Spain
| |
Collapse
|
427
|
Sequential chromatin immunoprecipitation to detect SUMOylated MeCP2 in neurons. Biochem Biophys Rep 2016; 5:374-378. [PMID: 28944302 PMCID: PMC5600420 DOI: 10.1016/j.bbrep.2016.01.014] [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] [Indexed: 11/22/2022] Open
Abstract
The small ubiquitin-like modifier (SUMO) is a short peptide that can be covalently linked to proteins altering their function. SUMOylation is an essential post-translational modification (PTM). Because of its dynamic nature, low abundance levels, and technical limitations, the occupation of endogenous SUMOylated transcription factors at genomic loci is challenging to detect. The chromatin regulator Methyl CpG binding protein 2 (MeCP2) is subjected to PTMs including SUMO. Mutations in MeCP2 lead to Rett syndrome, a severe neurodevelopmental disorder. Here, we present an efficient method to perform sequential chromatin immunoprecipitation (Seq-ChIP) for detecting SUMOylated MeCP2 in neurons. This Seq-ChIP technique is a useful tool to determine the occupancy of SUMOylated transcription and chromatin factors at specific genomic regions. SUMO is a short peptide that can be covalently linked to proteins. SUMOylation is an essential post-translational modification. MeCP2 is a chromatin regulator whose mutations lead to Rett syndrome. Methodology to detect SUMOylated MeCP2 at specific genomic regions in neurons.
Collapse
|
428
|
Tokarz P, Kaarniranta K, Blasiak J. Inhibition of DNA methyltransferase or histone deacetylase protects retinal pigment epithelial cells from DNA damage induced by oxidative stress by the stimulation of antioxidant enzymes. Eur J Pharmacol 2016; 776:167-75. [PMID: 26899469 DOI: 10.1016/j.ejphar.2016.02.049] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/11/2016] [Accepted: 02/15/2016] [Indexed: 12/13/2022]
Abstract
Epigenetic modifications influence DNA damage response (DDR). In this study we explored the role of DNA methylation and histone acetylation in DDR in cells challenged with acute or chronic oxidative stress. We used retinal pigment epithelial cells (ARPE-19), which natively are exposed to oxidative stress due to permanent exposure to light and high blood flow. We employed a DNA methyltransferase inhibitor - RG108 (RG), or a histone deacetylase inhibitor - valproic acid (VA). ARPE-19 cells were exposed to tert-butyl hydroperoxide, an acute oxidative stress inducer, or glucose oxidase, which slowly liberates low-doses of hydrogen peroxide in the presence of glucose, creating chronic conditions. VA and RG reduced level of intracellular reactive oxygen species and DNA damage in ARPE-19 cells in normal condition and in oxidative stress. This protective effect of VA and RG was associated with the up-regulated expression of antioxidant enzyme genes: CAT, GPx1, GPx4, SOD1 and SOD2. RG decreased the number of cells in G2/M checkpoint in response to chronic oxidative stress. Neither RG nor VA changed the DNA repair or apoptosis induced by oxidative stress. Therefore, certain epigenetic manipulations may protect ARPE-19 cells from detrimental effects of oxidative stress by modulation of antioxidative enzyme gene expression, which may be further explored in pharmacological studies on oxidative stress-related eye diseases.
Collapse
Affiliation(s)
- Paulina Tokarz
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland.
| | - Janusz Blasiak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
429
|
Tai DJC, Liu YC, Hsu WL, Ma YL, Cheng SJ, Liu SY, Lee EHY. MeCP2 SUMOylation rescues Mecp2-mutant-induced behavioural deficits in a mouse model of Rett syndrome. Nat Commun 2016; 7:10552. [PMID: 26842955 PMCID: PMC4743023 DOI: 10.1038/ncomms10552] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/24/2015] [Indexed: 11/17/2022] Open
Abstract
The methyl-CpG-binding protein 2 (MeCP2) gene, MECP2, is an X-linked gene encoding the MeCP2 protein, and mutations of MECP2 cause Rett syndrome (RTT). However, the molecular mechanism of MECP2-mutation-caused RTT is less known. Here we find that MeCP2 could be SUMO-modified by the E3 ligase PIAS1 at Lys-412. MeCP2 phosphorylation (at Ser-421 and Thr-308) facilitates MeCP2 SUMOylation, and MeCP2 SUMOylation is induced by NMDA, IGF-1 and CRF in the rat brain. MeCP2 SUMOylation releases CREB from the repressor complex and enhances Bdnf mRNA expression. Several MECP2 mutations identified in RTT patients show decreased MeCP2 SUMOylation. Re-expression of wild-type MeCP2 or SUMO-modified MeCP2 in Mecp2-null neurons rescues the deficits of social interaction, fear memory and LTP observed in Mecp2 conditional knockout (cKO) mice. These results together reveal an important role of MeCP2 SUMOylation in social interaction, memory and synaptic plasticity, and that abnormal MeCP2 SUMOylation is implicated in RTT. Post-translational modifications of methyl-CpG-binding protein 2 (MeCP2) are important for its function and dysfunction in Rett syndrome. Here, Tai et al. show a functional interaction between MeCP2 SUMOylation and phosphorylation in rodent behavior and synaptic plasticity.
Collapse
Affiliation(s)
- Derek J C Tai
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yen C Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Wei L Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yun L Ma
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Sin J Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.,Neuroscience Program in Academia Sinica, Taipei 115, Taiwan
| | - Shau Y Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Eminy H Y Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
430
|
Gene-Stress-Epigenetic Regulation of FKBP5: Clinical and Translational Implications. Neuropsychopharmacology 2016; 41:261-74. [PMID: 26250598 PMCID: PMC4677131 DOI: 10.1038/npp.2015.235] [Citation(s) in RCA: 391] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 12/13/2022]
Abstract
Stress responses and related outcomes vary markedly across individuals. Elucidating the molecular underpinnings of this variability is of great relevance for developing individualized prevention strategies and treatments for stress-related disorders. An important modulator of stress responses is the FK506-binding protein 51 (FKBP5/FKBP51). FKBP5 acts as a co-chaperone that modulates not only glucocorticoid receptor activity in response to stressors but also a multitude of other cellular processes in both the brain and periphery. Notably, the FKBP5 gene is regulated via complex interactions among environmental stressors, FKBP5 genetic variants, and epigenetic modifications of glucocorticoid-responsive genomic sites. These interactions can result in FKBP5 disinhibition that has been shown to contribute to a number of aberrant phenotypes in both rodents and humans. Consequently, FKBP5 blockade may hold promise as treatment intervention for stress-related disorders, and recently developed selective FKBP5 blockers show encouraging results in vitro and in rodent models. Although risk for stress-related disorders is conferred by multiple environmental and genetic factors, the findings related to FKBP5 illustrate how a deeper understanding of the molecular and systemic mechanisms underlying specific gene-environment interactions may provide insights into the pathogenesis of stress-related disorders.
Collapse
|
431
|
Maiese K. Erythropoietin and mTOR: A "One-Two Punch" for Aging-Related Disorders Accompanied by Enhanced Life Expectancy. Curr Neurovasc Res 2016; 13:329-340. [PMID: 27488211 PMCID: PMC5079807 DOI: 10.2174/1567202613666160729164900] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/16/2022]
Abstract
Life expectancy continues to increase throughout the world, but is accompanied by a rise in the incidence of non-communicable diseases. As a result, the benefits of an increased lifespan can be limited by aging-related disorders that necessitate new directives for the development of effective and safe treatment modalities. With this objective, the mechanistic target of rapamycin (mTOR), a 289-kDa serine/threonine protein, and its related pathways of mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), proline rich Akt substrate 40 kDa (PRAS40), AMP activated protein kinase (AMPK), Wnt signaling, and silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), have generated significant excitement for furthering novel therapies applicable to multiple systems of the body. Yet, the biological and clinical outcome of these pathways can be complex especially with oversight of cell death mechanisms that involve apoptosis and autophagy. Growth factors, and in particular erythropoietin (EPO), are one avenue under consideration to implement control over cell death pathways since EPO can offer potential treatment for multiple disease entities and is intimately dependent upon mTOR signaling. In experimental and clinical studies, EPO appears to have significant efficacy in treating several disorders including those involving the developing brain. However, in mature populations that are affected by aging-related disorders, the direction for the use of EPO to treat clinical disease is less clear that may be dependent upon a number of factors including the understanding of mTOR signaling. Continued focus upon the regulatory elements that control EPO and mTOR signaling could generate critical insights for targeting a broad range of clinical maladies.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101, USA.
| |
Collapse
|
432
|
Abstract
Globally, greater than 30 million individuals are afflicted with disorders of the nervous system accompanied by tens of thousands of new cases annually with limited, if any, treatment options. Erythropoietin (EPO) offers an exciting and novel therapeutic strategy to address both acute and chronic neurodegenerative disorders. EPO governs a number of critical protective and regenerative mechanisms that can impact apoptotic and autophagic programmed cell death pathways through protein kinase B (Akt), sirtuins, mammalian forkhead transcription factors, and wingless signaling. Translation of the cytoprotective pathways of EPO into clinically effective treatments for some neurodegenerative disorders has been promising, but additional work is necessary. In particular, development of new treatments with erythropoiesis-stimulating agents such as EPO brings several important challenges that involve detrimental vascular outcomes and tumorigenesis. Future work that can effectively and safely harness the complexity of the signaling pathways of EPO will be vital for the fruitful treatment of disorders of the nervous system.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101
| |
Collapse
|
433
|
van Niekerk EA, Tuszynski MH, Lu P, Dulin JN. Molecular and Cellular Mechanisms of Axonal Regeneration After Spinal Cord Injury. Mol Cell Proteomics 2015; 15:394-408. [PMID: 26695766 DOI: 10.1074/mcp.r115.053751] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Indexed: 12/28/2022] Open
Abstract
Following axotomy, a complex temporal and spatial coordination of molecular events enables regeneration of the peripheral nerve. In contrast, multiple intrinsic and extrinsic factors contribute to the general failure of axonal regeneration in the central nervous system. In this review, we examine the current understanding of differences in protein expression and post-translational modifications, activation of signaling networks, and environmental cues that may underlie the divergent regenerative capacity of central and peripheral axons. We also highlight key experimental strategies to enhance axonal regeneration via modulation of intraneuronal signaling networks and the extracellular milieu. Finally, we explore potential applications of proteomics to fill gaps in the current understanding of molecular mechanisms underlying regeneration, and to provide insight into the development of more effective approaches to promote axonal regeneration following injury to the nervous system.
Collapse
Affiliation(s)
- Erna A van Niekerk
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093;
| | - Mark H Tuszynski
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093; §Veterans Administration Medical Center, San Diego, CA 92161
| | - Paul Lu
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093; §Veterans Administration Medical Center, San Diego, CA 92161
| | - Jennifer N Dulin
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093
| |
Collapse
|
434
|
Autism-Like Behavior and Epigenetic Changes Associated with Autism as Consequences of In Utero Exposure to Environmental Pollutants in a Mouse Model. Behav Neurol 2015; 2015:426263. [PMID: 26586927 PMCID: PMC4637446 DOI: 10.1155/2015/426263] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/01/2015] [Accepted: 10/01/2015] [Indexed: 12/31/2022] Open
Abstract
We tested the hypothesis that in utero exposure to heavy metals increases autism-like behavioral phenotypes in adult animals and induces epigenetic changes in genes that have roles in the etiology of autism. Mouse dams were treated with cadmium, lead, arsenate, manganese, and mercury via drinking water from gestational days (E) 1–10. Valproic acid (VPA) injected intraperitoneally once on (E) 8.5 served as a positive control. Young male offspring were tested for behavioral deficits using four standardized behavioral assays. In this study, in utero exposure to heavy metals resulted in multiple behavioral abnormalities that persisted into adulthood. VPA and manganese induced changes in perseverative/impulsive behavior and social dominance behavior, arsenic caused changes only in perseverative/impulsive behavior, and lead induced abnormalities in social interaction in comparison to the control animals. Brain samples from Mn, Pb, and VPA treated and control animals were evaluated for changes in CpG island methylation in promoter regions and associated changes in gene expression. The Chd7 gene, essential for neural crest cell migration and patterning, was found to be hypomethylated in each experimental animal tested compared to water-treated controls. Furthermore, distinct patterns of CpG island methylation yielded novel candidate genes for further investigation.
Collapse
|
435
|
Mi Y, Gao X, Dai J, Ma Y, Xu L, Jin W. A Novel Function of TET2 in CNS: Sustaining Neuronal Survival. Int J Mol Sci 2015; 16:21846-57. [PMID: 26378518 PMCID: PMC4613284 DOI: 10.3390/ijms160921846] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 08/19/2015] [Accepted: 09/01/2015] [Indexed: 11/16/2022] Open
Abstract
DNA dioxygenases Ten-Eleven Translocation (TET) proteins can catalyze the conversion of 5-methylcytosine (5mC) of DNA to 5-hydroxymethylcytosine (5hmC), and thereby alter the epigenetic state of DNA. The TET family includes TET1, TET2 and TET3 members in mammals. Recently, accumulative research uncovered that TET1-3 occur abundantly in the central nervous system (CNS), and their biological functions have just begun to be investigated. In the present study, we demonstrated that mRNA and protein of TET2 were highly expressed in the cerebral cortex and hippocampus along the whole brain-development process. Further studies showed that TET2 was expressed in various types of cells, especially in most neurons. Subcellular distribution pattern implicated that TET2 is localized in both nucleus and cytoplasm of neurons. Down-regulation of TET2 in cultured cortical neurons with RNA interference implied that TET2 was required for cell survival. In all, our results indicate that neuronal TET2 is positively involved in the regulation of cell survival.
Collapse
Affiliation(s)
- Yajing Mi
- State Key Laboratory of Military Stomatology, Department of Anesthesiology, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China.
- Institute of Basic Medicine Science, Xi'an Medical University, Xi'an 710021, China.
| | - Xingchun Gao
- Institute of Basic Medicine Science, Xi'an Medical University, Xi'an 710021, China.
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jinxiang Dai
- Department of Cell and Developmental Biology, University of Colorado Denver, Denver, CO 80045, USA.
| | - Yue Ma
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Lixian Xu
- State Key Laboratory of Military Stomatology, Department of Anesthesiology, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China.
| | - Weilin Jin
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
436
|
Pirola CJ, Scian R, Gianotti TF, Dopazo H, Rohr C, Martino JS, Castaño GO, Sookoian S. Epigenetic Modifications in the Biology of Nonalcoholic Fatty Liver Disease: The Role of DNA Hydroxymethylation and TET Proteins. Medicine (Baltimore) 2015; 94:e1480. [PMID: 26356709 PMCID: PMC4616643 DOI: 10.1097/md.0000000000001480] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The 5-Hydroxymethylcytosine (5-hmC) is an epigenetic modification whose role in the pathogenesis of metabolic-related complex diseases remains unexplored; 5-hmC appears to be prevalent in the mitochondrial genome. The Ten-Eleven-Translocation (TET) family of proteins is responsible for catalyzing the conversion of 5-methylcytosine to 5-hmC. We hypothesized that epigenetic editing by 5-hmC might be a novel mechanism through which nonalcoholic fatty liver disease (NAFLD)-associated molecular traits could be explained.Hence, we performed an observational study to explore global levels of 5-hmC in fresh liver samples of patients with NAFLD and controls (n = 90) using an enzyme-linked-immunosorbent serologic assay and immunohistochemistry. We also screened for genetic variation in TET 1-3 loci by next generation sequencing to explore its contribution to the disease biology. The study was conducted in 2 stages (discovery and replication) and included 476 participants.We observed that the amount of 5-hmC in the liver of both NAFLD patients and controls was relatively low (up to 0.1%); a significant association was found with liver mitochondrial DNA copy number (R = 0.50, P = 0.000382) and PPARGC1A-mRNA levels (R = -0.57, P = 0.04).We did not observe any significant difference in the 5-hmC nuclear immunostaining score between NAFLD patients and controls; nevertheless, we found that patients with NAFLD (0.4 ± 0.5) had significantly lower nonnuclear-5-hmC staining compared with controls (1.8 ± 0.8), means ± standard deviation, P = 0.028. The missense p.Ile1123Met variant (TET1-rs3998860) was significantly associated with serum levels of caspase-generated CK-18 fragment-cell death biomarker in the discovery and replication stage, and the disease severity (odds ratio: 1.47, 95% confidence interval: 1.10-1.97; P = 0.005). The p.Ile1762Val substitution (TET2-rs2454206) was associated with liver PPARGC1A-methylation and transcriptional levels, and Type 2 diabetes.Our results suggest that 5-hmC might be involved in the pathogenesis of NAFLD by regulating liver mitochondrial biogenesis and PPARGC1A expression. Genetic diversity at TET loci suggests an "epigenetic" regulation of programmed liver-cell death and a TET-mediated fine-tuning of the liver PPARGC1A-transcriptional program.
Collapse
Affiliation(s)
- Carlos J Pirola
- From the Department of Molecular Genetics and Biology of Complex Diseases, Institute of Medical Research A Lanari-IDIM, University of Buenos Aires-National Scientific and Technical Research Council (CONICET), Ciudad Autónoma de Buenos Aires, Argentina (CJP, RS, TFG); Biomedical Genomics and Evolution Laboratory, Ecology, Genetics and Evolution Department, Faculty of Science, IEGEBA, University of Buenos Aires-National Scientific and Technical Research Council (CONICET), Ciudad Autónoma de Buenos Aires, Argentina (HD, CR); Department of Pathology, Hospital Diego Thompson, San Martin, Buenos Aires, Argentina (JSM); Liver Unit, Medicine and Surgery Department, Hospital Abel Zubizarreta, Ciudad Autónoma de Buenos Aires, Argentina (GOC); and Department of Clinical and Molecular Hepatology, Institute of Medical Research A Lanari-IDIM, University of Buenos Aires-National Scientific and Technical Research Council (CONICET), Ciudad Autónoma de Buenos Aires, Argentina (RS, SS)
| | | | | | | | | | | | | | | |
Collapse
|
437
|
Liu H, Shen B, Xing M, Zhang J, Tian B. Vacuum-activated Co2+ and Ti3+ co-modified TiO2 with stable and enhanced photocatalytic activity. RESEARCH ON CHEMICAL INTERMEDIATES 2015. [DOI: 10.1007/s11164-015-2225-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
438
|
Maiese K. Stem cell guidance through the mechanistic target of rapamycin. World J Stem Cells 2015; 7:999-1009. [PMID: 26328016 PMCID: PMC4550632 DOI: 10.4252/wjsc.v7.i7.999] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/29/2015] [Accepted: 07/17/2015] [Indexed: 02/06/2023] Open
Abstract
Stem cells offer great promise for the treatment of multiple disorders throughout the body. Critical to this premise is the ability to govern stem cell pluripotency, proliferation, and differentiation. The mechanistic target of rapamycin (mTOR), 289-kDa serine/threonine protein kinase, that is a vital component of mTOR Complex 1 and mTOR Complex 2 represents a critical pathway for the oversight of stem cell maintenance. mTOR can control the programmed cell death pathways of autophagy and apoptosis that can yield variable outcomes in stem cell survival and be reliant upon proliferative pathways that include Wnt signaling, Wnt1 inducible signaling pathway protein 1 (WISP1), silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), and trophic factors. mTOR also is a necessary component for the early development and establishment of stem cells as well as having a significant impact in the regulation of the maturation of specific cell phenotypes. Yet, as a proliferative agent, mTOR can not only foster cancer stem cell development and tumorigenesis, but also mediate cell senescence under certain conditions to limit invasive cancer growth. mTOR offers an exciting target for the oversight of stem cell therapies but requires careful consideration of the diverse clinical outcomes that can be fueled by mTOR signaling pathways.
Collapse
|
439
|
Hitchcock LN, Lattal KM. Histone-mediated epigenetics in addiction. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 128:51-87. [PMID: 25410541 DOI: 10.1016/b978-0-12-800977-2.00003-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Many of the brain regions, neurotransmitter systems, and behavioral changes that occur after occasional drug use in healthy subjects and after chronic drug abuse in addicted patients are well characterized. An emerging literature suggests that epigenetic processes, those processes that regulate the accessibility of DNA to regulatory proteins within the nucleus, are keys to how addiction develops and how it may be treated. Investigations of the regulation of chromatin, the organizational system of DNA, by histone modification are leading to a new understanding of the cellular and behavioral alterations that occur after drug use. We will describe how, when, and where histone tails are modified and how some of the most recognized histone regulation patterns are involved in the cycle of addiction, including initial and chronic drug intake, withdrawal, abstinence, and relapse. Finally, we consider how an approach that targets histone modifications may promote successful treatment.
Collapse
Affiliation(s)
- Leah N Hitchcock
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
440
|
Wu MV, Sahay A, Duman RS, Hen R. Functional differentiation of adult-born neurons along the septotemporal axis of the dentate gyrus. Cold Spring Harb Perspect Biol 2015; 7:a018978. [PMID: 26238355 DOI: 10.1101/cshperspect.a018978] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Over the past several decades, the proliferation and integration of adult-born neurons into existing hippocampal circuitry has been implicated in a wide range of behaviors, including novelty recognition, pattern separation, spatial learning, anxiety behaviors, and antidepressant response. In this review, we suggest that the diversity in behavioral requirements for new neurons may be partly caused by separate functional roles of individual neurogenic niches. Growing evidence shows that the hippocampal formation can be compartmentalized not only along the classic trisynaptic circuit, but also along a longitudinal septotemporal axis. We suggest that subpopulations of hippocampal adult-born neurons may be specialized for distinct mnemonic- or mood-related behavioral tasks. We will examine the literature supporting a functional and anatomical dissociation of the hippocampus along the longitudinal axis and discuss techniques to functionally dissect the roles of adult-born hippocampal neurons in these distinct subregions.
Collapse
Affiliation(s)
- Melody V Wu
- Department of Psychiatry, Columbia University, New York, New York 10027 Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032
| | - Amar Sahay
- Center for Regenerative Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts 02114 Harvard Stem Cell Institute and Harvard Medical School, Boston, Massachusetts 02115
| | - Ronald S Duman
- Department of Psychiatry, Yale University, New Haven, Connecticut 06520 Department of Neurobiology, Yale University, New Haven, Connecticut 06520
| | - René Hen
- Department of Psychiatry, Columbia University, New York, New York 10027 Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032 Department of Neuroscience, Columbia University, New York, New York 10027 Department of Pharmacology, Columbia University, New York, New York 10027
| |
Collapse
|
441
|
Su M, Hong J, Zhao Y, Liu S, Xue X. MeCP2 controls hippocampal brain-derived neurotrophic factor expression via homeostatic interactions with microRNA‑132 in rats with depression. Mol Med Rep 2015; 12:5399-406. [PMID: 26239616 DOI: 10.3892/mmr.2015.4104] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 06/03/2015] [Indexed: 11/06/2022] Open
Abstract
Major depressive disorder (MDD) is a considerable public health concern, which affects patients worldwide. MDD is associated with psychosocial impairment, poor quality of life, and significant disability, morbidity and mortality. Stress is a major factor in depression, which impairs the structural and functional plasticity of the hippocampus. Previous studies have demonstrated that chronic unpredictable mild stress is able to downregulate the expression of brain‑derived neurotrophic factor (BDNF) and methyl‑CpG‑binding protein 2 (MeCP2), and alter the expression levels of certain microRNAs (miR). The aim of the present study was to investigate the regulatory association between BDNF, MeCP2 and miR-132 in an animal model of chronic stress‑induced depression. ELISA, western blot and qPCR were used to detect the expression levels of BDNF, MeCP2 and miR-132 in the peripheral blood samples of patients with MDD and in the hippocampi of depressed animals. In addition, a dual luciferase reporter gene system was used to determine whether miR-132 directly targets BDNF or MeCP2. The present study demonstrated that, as compared with normal subjects, miR‑132 expression was increased in the peripheral blood samples of patients with MDD, whereas the expression of MeCP2 and BDNF was decreased; thus, the expression levels of MeCP2 and BDNF were negatively correlated with those of miR‑132. In addition, in an animal model of chronic stress‑induced depression, increased expression levels of miR‑132, and decreased levels of MeCP2 and BDNF were detected in the hippocampi. Furthermore, knockdown of MeCP2 expression in primary hippocampal neurons increased the expression of miR‑132 and decreased the expression levels of BDNF. The results of the present study demonstrated that miR‑132 may directly target MeCP2, but not BDNF, and control its expression at the transcriptional and translational level. miR‑132 was also shown to negatively regulate BDNF expression. The reduced expression levels of BDNF, as induced by MeCP2 knockdown, were enhanced by miR‑132 mimics, and were rescued by miR‑132 inhibitors. These results suggested that homeostatic interactions between MeCP2 and miR‑132 may regulate hippocampal BDNF levels, which may have a role in the pathogenesis of MDD.
Collapse
Affiliation(s)
- Meilei Su
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jun Hong
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yongzhi Zhao
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Shuai Liu
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiang Xue
- Department of Psychology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
442
|
In Pursuit of New Imprinting Syndromes by Epimutation Screening in Idiopathic Neurodevelopmental Disorder Patients. BIOMED RESEARCH INTERNATIONAL 2015; 2015:341986. [PMID: 26106604 PMCID: PMC4461700 DOI: 10.1155/2015/341986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 05/04/2015] [Accepted: 05/11/2015] [Indexed: 12/19/2022]
Abstract
Alterations of epigenetic mechanisms, and more specifically imprinting modifications, could be responsible of neurodevelopmental disorders such as intellectual disability (ID) or autism together with other associated clinical features in many cases. Currently only eight imprinting syndromes are defined in spite of the fact that more than 200 genes are known or predicted to be imprinted. Recent publications point out that some epimutations which cause imprinting disorders may affect simultaneously different imprinted loci, suggesting that DNA-methylation may have been altered more globally. Therefore, we hypothesised that the detection of altered methylation patterns in known imprinting loci will indirectly allow identifying new syndromes due to epimutations among patients with unexplained ID. In a screening for imprinting alterations in 412 patients with syndromic ID/autism we found five patients with altered methylation in the four genes studied: MEG3, H19, KCNQ1OT1, and SNRPN. Remarkably, the cases with partial loss of methylation in KCNQ1OT1 and SNRPN present clinical features different to those associated with the corresponding imprinting syndromes, suggesting a multilocus methylation defect in accordance with our initial hypothesis. Consequently, our results are a proof of concept that the identification of epimutations in known loci in patients with clinical features different from those associated with known syndromes will eventually lead to the definition of new imprinting disorders.
Collapse
|
443
|
New Insights for Oxidative Stress and Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:875961. [PMID: 26064426 PMCID: PMC4443788 DOI: 10.1155/2015/875961] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 04/15/2015] [Indexed: 12/12/2022]
Abstract
The release of reactive oxygen species (ROS) and the generation of oxidative stress are considered critical factors for the pathogenesis of diabetes mellitus (DM), a disorder that is growing in prevalence and results in significant economic loss. New therapeutic directions that address the detrimental effects of oxidative stress may be especially warranted to develop effective care for the millions of individuals that currently suffer from DM. The mechanistic target of rapamycin (mTOR), silent mating type information regulation 2 homolog 1 (S. cerevisiae) (SIRT1), and Wnt1 inducible signaling pathway protein 1 (WISP1) are especially justified to be considered treatment targets for DM since these pathways can address the complex relationship between stem cells, trophic factors, impaired glucose tolerance, programmed cell death pathways of apoptosis and autophagy, tissue remodeling, cellular energy homeostasis, and vascular biology that greatly impact the biology and disease progression of DM. The translation and development of these pathways into viable therapies will require detailed understanding of their proliferative nature to maximize clinical efficacy and limit adverse effects that have the potential to lead to unintended consequences.
Collapse
|
444
|
Maiese K. Novel applications of trophic factors, Wnt and WISP for neuronal repair and regeneration in metabolic disease. Neural Regen Res 2015; 10:518-28. [PMID: 26170801 PMCID: PMC4424733 DOI: 10.4103/1673-5374.155427] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2015] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus affects almost 350 million individuals throughout the globe resulting in significant morbidity and mortality. Of further concern is the growing population of individuals that remain undiagnosed but are susceptible to the detrimental outcomes of this disorder. Diabetes mellitus leads to multiple complications in the central and peripheral nervous systems that include cognitive impairment, retinal disease, neuropsychiatric disease, cerebral ischemia, and peripheral nerve degeneration. Although multiple strategies are being considered, novel targeting of trophic factors, Wnt signaling, Wnt1 inducible signaling pathway protein 1, and stem cell tissue regeneration are considered to be exciting prospects to overcome the cellular mechanisms that lead to neuronal injury in diabetes mellitus involving oxidative stress, apoptosis, and autophagy. Pathways that involve insulin-like growth factor-1, fibroblast growth factor, epidermal growth factor, and erythropoietin can govern glucose homeostasis and are intimately tied to Wnt signaling that involves Wnt1 and Wnt1 inducible signaling pathway protein 1 (CCN4) to foster control over stem cell proliferation, wound repair, cognitive decline, β-cell proliferation, vascular regeneration, and programmed cell death. Ultimately, cellular metabolism through Wnt signaling is driven by primary metabolic pathways of the mechanistic target of rapamycin and AMP activated protein kinase. These pathways offer precise biological control of cellular metabolism, but are exquisitely sensitive to the different components of Wnt signaling. As a result, unexpected clinical outcomes can ensue and therefore demand careful translation of the mechanisms that govern neural repair and regeneration in diabetes mellitus.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101, USA
| |
Collapse
|
445
|
Love CE, Prince VE. Rest represses maturation within migrating facial branchiomotor neurons. Dev Biol 2015; 401:220-35. [PMID: 25769695 DOI: 10.1016/j.ydbio.2015.02.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 02/04/2015] [Accepted: 02/28/2015] [Indexed: 10/23/2022]
Abstract
The vertebrate brain arises from the complex organization of millions of neurons. Neurogenesis encompasses not only cell fate specification from neural stem cells, but also the terminal molecular and morphological maturation of neurons at correct positions within the brain. RE1-silencing transcription factor (Rest) is expressed in non-neural tissues and neuronal progenitors where it inhibits the terminal maturation of neurons by repressing hundreds of neuron-specific genes. Here we show that Rest repression of maturation is intimately linked with the migratory capability of zebrafish facial branchiomotor neurons (FBMNs), which undergo a characteristic tangential migration from hindbrain rhombomere (r) 4 to r6/r7 during development. We establish that FBMN migration is increasingly disrupted as Rest is depleted in zebrafish rest mutant embryos, such that around two-thirds of FBMNs fail to complete migration in mutants depleted of both maternal and zygotic Rest. Although Rest is broadly expressed, we show that de-repression or activation of Rest target genes only within FBMNs is sufficient to disrupt their migration. We demonstrate that this migration defect is due to precocious maturation of FBMNs, based on both morphological and molecular criteria. We further show that the Rest target gene and alternative splicing factor srrm4 is a key downstream regulator of maturation; Srrm4 knockdown partially restores the ability of FBMNs to migrate in rest mutants while preventing their precocious morphological maturation. Rest must localize to the nucleus to repress its targets, and its subcellular localization is highly regulated: we show that targeting Rest specifically to FBMN nuclei rescues FBMN migration in Rest-deficient embryos. We conclude that Rest functions in FBMN nuclei to inhibit maturation until the neurons complete their migration.
Collapse
Affiliation(s)
- Crystal E Love
- Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, Chicago, IL 60615, USA
| | - Victoria E Prince
- Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, Chicago, IL 60615, USA; Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
446
|
Maiese K. Programming apoptosis and autophagy with novel approaches for diabetes mellitus. Curr Neurovasc Res 2015; 12:173-88. [PMID: 25742566 PMCID: PMC4380829 DOI: 10.2174/1567202612666150305110929] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/16/2015] [Accepted: 02/19/2015] [Indexed: 12/13/2022]
Abstract
According to the World Health Organization, diabetes mellitus (DM) in the year 2030 will be ranked the seventh leading cause of death in the world. DM impacts all systems of the body with oxidant stress controlling cell fate through endoplasmic reticulum stress, mitochondrial dysfunction, alterations in uncoupling proteins, and the induction of apoptosis and autophagy. Multiple treatment approaches are being entertained for DM with Wnt1 inducible signaling pathway protein 1 (WISP1), mechanistic target of rapamycin (mTOR), and silent mating type information regulation 2 homolog) 1 (S. cerevisiae) (SIRT1) generating significant interest as target pathways that can address maintenance of glucose homeostasis as well as prevention of cellular pathology by controlling insulin resistance, stem cell proliferation, and the programmed cell death pathways of apoptosis and autophagy. WISP1, mTOR, and SIRT1 can rely upon similar pathways such as AMP activated protein kinase as well as govern cellular metabolism through cytokines such as EPO and oral hypoglycemics such as metformin. Yet, these pathways require precise biological control to exclude potentially detrimental clinical outcomes. Further elucidation of the ability to translate the roles of WISP1, mTOR, and SIRT1 into effective clinical avenues offers compelling prospects for new therapies against DM that can benefit hundreds of millions of individuals throughout the globe.
Collapse
Affiliation(s)
- Kenneth Maiese
- MD, Cellular and Molecular Signaling, Newark, New Jersey 07101, USA.
| |
Collapse
|
447
|
Dempster EL, Wong CC, Lester KJ, Burrage J, Gregory AM, Mill J, Eley TC. Genome-wide methylomic analysis of monozygotic twins discordant for adolescent depression. Biol Psychiatry 2014; 76:977-83. [PMID: 24929637 PMCID: PMC4252163 DOI: 10.1016/j.biopsych.2014.04.013] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/14/2014] [Accepted: 04/13/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Adolescent depression is a common neuropsychiatric disorder that often continues into adulthood and is associated with a wide range of poor outcomes including suicide. Although numerous studies have looked at genetic markers associated with depression, the role of epigenetic variation remains relatively unexplored. METHODS Monozygotic (MZ) twins were selected from an adolescent twin study designed to investigate the interplay of genetic and environmental factors in the development of emotional and behavioral difficulties. There were 18 pairs of MZ twins identified in which one member scored consistently higher (group mean within the clinically significant range) on self-rated depression than the other. We assessed genome-wide patterns of DNA methylation in twin buccal cell DNA using the Infinium HumanMethylation450 BeadChip from Illumina. Quality control and data preprocessing was undertaken using the wateRmelon package. Differentially methylated probes (DMPs) were identified using an analysis strategy taking into account both the significance and the magnitude of DNA methylation differences. The top differentially methylated DMP was successfully validated by bisulfite-pyrosequencing, and identified DMPs were tested in postmortem brain samples obtained from patients with major depressive disorder (n = 14) and matched control subjects (n = 15). RESULTS Two reproducible depression-associated DMPs were identified, including the top-ranked DMP that was located within STK32C, which encodes a serine/threonine kinase, of unknown function. CONCLUSIONS Our data indicate that DNA methylation differences are apparent in MZ twins discordant for adolescent depression and that some of the disease-associated variation observed in buccal cell DNA is mirrored in adult brain tissue obtained from individuals with clinical depression.
Collapse
Affiliation(s)
- Emma L. Dempster
- University of Exeter Medical School, Exeter University, Exeter,Address correspondence to Emma L. Dempster, Ph.D., University of Exeter Medical School, Exeter University, RILD-Medical Research, Level 4, Royal Devon and Exeter Hospital, Barrack Rd, Exeter EX2 5DW, United Kingdom
| | - Chloe C.Y. Wong
- Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King’s College London
| | - Kathryn J. Lester
- Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King’s College London
| | - Joe Burrage
- University of Exeter Medical School, Exeter University, Exeter
| | - Alice M. Gregory
- Department of Psychology, Goldsmiths, University of London, London, United Kingdom
| | - Jonathan Mill
- University of Exeter Medical School, Exeter University, Exeter,Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King’s College London
| | - Thalia C. Eley
- Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King’s College London
| |
Collapse
|
448
|
Fortress AM, Frick KM. Epigenetic regulation of estrogen-dependent memory. Front Neuroendocrinol 2014; 35:530-49. [PMID: 24878494 PMCID: PMC4174980 DOI: 10.1016/j.yfrne.2014.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 02/09/2023]
Abstract
Hippocampal memory formation is highly regulated by post-translational histone modifications and DNA methylation. Accordingly, these epigenetic processes play a major role in the effects of modulatory factors, such as sex steroid hormones, on hippocampal memory. Our laboratory recently demonstrated that the ability of the potent estrogen 17β-estradiol (E2) to enhance hippocampal-dependent novel object recognition memory in ovariectomized female mice requires ERK-dependent histone H3 acetylation and DNA methylation in the dorsal hippocampus. Although these data provide valuable insight into the chromatin modifications that mediate the memory-enhancing effects of E2, epigenetic regulation of gene expression is enormously complex. Therefore, more research is needed to fully understand how E2 and other hormones employ epigenetic alterations to shape behavior. This review discusses the epigenetic alterations shown thus far to regulate hippocampal memory, briefly reviews the effects of E2 on hippocampal function, and describes in detail our work on epigenetic regulation of estrogenic memory enhancement.
Collapse
Affiliation(s)
- Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
449
|
Henley JM, Craig TJ, Wilkinson KA. Neuronal SUMOylation: mechanisms, physiology, and roles in neuronal dysfunction. Physiol Rev 2014; 94:1249-85. [PMID: 25287864 PMCID: PMC4187031 DOI: 10.1152/physrev.00008.2014] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Protein SUMOylation is a critically important posttranslational protein modification that participates in nearly all aspects of cellular physiology. In the nearly 20 years since its discovery, SUMOylation has emerged as a major regulator of nuclear function, and more recently, it has become clear that SUMOylation has key roles in the regulation of protein trafficking and function outside of the nucleus. In neurons, SUMOylation participates in cellular processes ranging from neuronal differentiation and control of synapse formation to regulation of synaptic transmission and cell survival. It is a highly dynamic and usually transient modification that enhances or hinders interactions between proteins, and its consequences are extremely diverse. Hundreds of different proteins are SUMO substrates, and dysfunction of protein SUMOylation is implicated in a many different diseases. Here we briefly outline core aspects of the SUMO system and provide a detailed overview of the current understanding of the roles of SUMOylation in healthy and diseased neurons.
Collapse
Affiliation(s)
- Jeremy M Henley
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Tim J Craig
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Kevin A Wilkinson
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
450
|
Berbel P, Navarro D, Román GC. An evo-devo approach to thyroid hormones in cerebral and cerebellar cortical development: etiological implications for autism. Front Endocrinol (Lausanne) 2014; 5:146. [PMID: 25250016 PMCID: PMC4158880 DOI: 10.3389/fendo.2014.00146] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 08/25/2014] [Indexed: 12/11/2022] Open
Abstract
The morphological alterations of cortical lamination observed in mouse models of developmental hypothyroidism prompted the recognition that these experimental changes resembled the brain lesions of children with autism; this led to recent studies showing that maternal thyroid hormone deficiency increases fourfold the risk of autism spectrum disorders (ASD), offering for the first time the possibility of prevention of some forms of ASD. For ethical reasons, the role of thyroid hormones on brain development is currently studied using animal models, usually mice and rats. Although mammals have in common many basic developmental principles regulating brain development, as well as fundamental basic mechanisms that are controlled by similar metabolic pathway activated genes, there are also important differences. For instance, the rodent cerebral cortex is basically a primary cortex, whereas the primary sensory areas in humans account for a very small surface in the cerebral cortex when compared to the associative and frontal areas that are more extensive. Associative and frontal areas in humans are involved in many neurological disorders, including ASD, attention deficit-hyperactive disorder, and dyslexia, among others. Therefore, an evo-devo approach to neocortical evolution among species is fundamental to understand not only the role of thyroid hormones and environmental thyroid disruptors on evolution, development, and organization of the cerebral cortex in mammals but also their role in neurological diseases associated to thyroid dysfunction.
Collapse
Affiliation(s)
- Pere Berbel
- Departamento de Histología y Anatomía, Facultad de Medicina, Universidad Miguel Hernández, Alicante, Spain
| | - Daniela Navarro
- Departamento de Histología y Anatomía, Facultad de Medicina, Universidad Miguel Hernández, Alicante, Spain
| | - Gustavo C. Román
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Methodist Neurological Institute, Houston, TX, USA
| |
Collapse
|