1
|
Joshi RS, Rigau M, García-Prieto CA, Castro de Moura M, Piñeyro D, Moran S, Davalos V, Carrión P, Ferrando-Bernal M, Olalde I, Lalueza-Fox C, Navarro A, Fernández-Tena C, Aspandi D, Sukno FM, Binefa X, Valencia A, Esteller M. Look-alike humans identified by facial recognition algorithms show genetic similarities. Cell Rep 2022; 40:111257. [PMID: 36001980 DOI: 10.1016/j.celrep.2022.111257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 06/05/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022] Open
Abstract
The human face is one of the most visible features of our unique identity as individuals. Interestingly, monozygotic twins share almost identical facial traits and the same DNA sequence but could exhibit differences in other biometrical parameters. The expansion of the world wide web and the possibility to exchange pictures of humans across the planet has increased the number of people identified online as virtual twins or doubles that are not family related. Herein, we have characterized in detail a set of "look-alike" humans, defined by facial recognition algorithms, for their multiomics landscape. We report that these individuals share similar genotypes and differ in their DNA methylation and microbiome landscape. These results not only provide insights about the genetics that determine our face but also might have implications for the establishment of other human anthropometric properties and even personality characteristics.
Collapse
Affiliation(s)
- Ricky S Joshi
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain
| | - Maria Rigau
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
| | - Carlos A García-Prieto
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain; Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
| | | | - David Piñeyro
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
| | - Sebastian Moran
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain
| | - Veronica Davalos
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain
| | - Pablo Carrión
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), 08003 Barcelona, Spain
| | - Manuel Ferrando-Bernal
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), 08003 Barcelona, Spain
| | - Iñigo Olalde
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), 08003 Barcelona, Spain
| | - Carles Lalueza-Fox
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), 08003 Barcelona, Spain
| | - Arcadi Navarro
- Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), 08003 Barcelona, Spain; Centre for Genomic Regulation (CNAG-CRG), 08003 Barcelona, Catalonia, Spain; Institucio Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | | | - Decky Aspandi
- Departament de Tecnologies de la Informació i les Comunicaciones (DTIC), Universitat Pompeu Fabra (UPF), 08018 Barcelona, Spain
| | - Federico M Sukno
- Departament de Tecnologies de la Informació i les Comunicaciones (DTIC), Universitat Pompeu Fabra (UPF), 08018 Barcelona, Spain
| | - Xavier Binefa
- Departament de Tecnologies de la Informació i les Comunicaciones (DTIC), Universitat Pompeu Fabra (UPF), 08018 Barcelona, Spain
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain; Institucio Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916 Barcelona, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain; Institucio Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), L'Hospitalet, 08907 Barcelona, Spain.
| |
Collapse
|
2
|
Zheng K, Yan J, Deng J, Wu W, Wen Y. Modification of Experimental Design and Statistical Method for Mapping Imprinted QTLs Based on Immortalized F2 Population. Front Genet 2020; 11:589047. [PMID: 33329733 PMCID: PMC7714927 DOI: 10.3389/fgene.2020.589047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/29/2020] [Indexed: 11/20/2022] Open
Abstract
Genomic imprinting is an epigenetic phenomenon, which plays important roles in the growth and development of animals and plants. Immortalized F2 (imF2) populations generated by random cross between recombinant inbred (RI) or doubled haploid (DH) lines have been proved to have significant advantages for mapping imprinted quantitative trait loci (iQTLs), and statistical methods for this purpose have been proposed. In this paper, we propose a special type of imF2 population (R-imF2) for iQTL mapping, which is developed by random reciprocal cross between RI/DH lines. We also propose two modified iQTL mapping methods: two-step point mapping (PM-2) and two-step composite point mapping (CPM-2). Simulation studies indicated that: (i) R-imF2 cannot improve the results of iQTL mapping, but the experimental design can probably reduce the workload of population construction; (ii) PM-2 can increase the precision of estimating the position and effects of a single iQTL; and (iii) CPM-2 can precisely map not only iQTLs, but also non-imprinted QTLs. The modified experimental design and statistical methods will facilitate and promote the study of iQTL mapping.
Collapse
Affiliation(s)
- Kehui Zheng
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Computer and Information Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jiqiang Yan
- College of Computer and Information Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jiacong Deng
- School of Ocean and Biochemical Engineering, Fuqing Branch of Fujian Normal University, Fuzhou, China
| | - Weiren Wu
- Fujian Provincial Key Laboratory of Crop Breeding by Design, Fujian Agriculture and Forestry University, Fuzhou, China
- Key Laboratory of Genetics, Breeding and Multiple Utilization of Crops, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou, China
- *Correspondence: Weiren Wu,
| | - Yongxian Wen
- College of Computer and Information Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Yongxian Wen,
| |
Collapse
|
3
|
Gholami D, Salman Yazdi R, Jami MS, Ghasemi S, Sadighi Gilani MA, Sadeghinia S, Teimori H. The expression of Cysteine-Rich Secretory Protein 2 (CRISP2) and miR-582-5p in seminal plasma fluid and spermatozoa of infertile men. Gene 2019; 730:144261. [PMID: 31778754 DOI: 10.1016/j.gene.2019.144261] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/19/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022]
Abstract
Cysteine-Rich Secretory Protein 2 (CRISP2) plays an important role in the morphology and motion of male ejaculated spermatozoa. The association of its expression with some miRNAs is also well known. The aim of this study was to determine the expression of CRISP2 and mir-582 in the seminal plasma fluid and spermatozoa of three groups of infertile men and the possible association of their expressions. In this experimental study, the expression of CRISP2 in seminal plasma fluid and spermatozoa of 17 men with asthenozoospermia, 15 men with teratozoospermia, 17 men with teratoasthenozoospermia, and 18 infertile individuals with normozoospermia were measured using western blotting. Then by using bioinformatics studies, miR-582-5p was nominated as a CRISP2-associated miRNA, and its expression was evaluated by means of Real-Time PCR. Comparison of expression of CRISP2 and miRNA-582 in the studied groups was analyzed by t-test and Mann-Whitney U test. The expression of CRISP2 showed a significant reduction in the spermatozoa and seminal plasma fluid of all three groups, (p < 0.05). MiR-582-5p expression significantly increased in teratozoospermia patients (<0.05), and significantly decreased in teratoasthenozoospermia patients (p < 0.05). Meanwhile, changes in the expression of miR-582-5p in teratoasthenozoospermia individuals was associated with a decrease in the expression of CRISP2, which could represent the potential role of miR-582-5p in regulation of CRISP2 expression in teratoasthenozoospermia individuals.
Collapse
Affiliation(s)
- Delnya Gholami
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Reza Salman Yazdi
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mohammad-Saeid Jami
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Neurology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Sorayya Ghasemi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad-Ali Sadighi Gilani
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Shaghayegh Sadeghinia
- College of Medical Veterinary and Life Sciences School of Molecular Cell and Systems Biology, University of Glasgow, Scotland
| | - Hossien Teimori
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
4
|
Prats-Puig A, Xargay-Torrent S, Carreras-Badosa G, Mas-Parés B, Bassols J, Petry CJ, Girardot M, D E Zegher F, Ibáñez L, Dunger DB, Feil R, López-Bermejo A. Methylation of the C19MC microRNA locus in the placenta: association with maternal and chilhood body size. Int J Obes (Lond) 2019; 44:13-22. [PMID: 31554916 DOI: 10.1038/s41366-019-0450-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/18/2019] [Accepted: 07/07/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To study DNA methylation at the C19MC locus in the placenta and its association with (1) parental body size, (2) transmission of haplotypes for the C19MC rs55765443 SNP, and (3) offspring's body size and/or body composition at birth and in childhood. SUBJECTS AND METHODS Seventy-two pregnant women-infant pairs and 63 fathers were included in the study. Weight and height of mothers, fathers and newborns were registered during pregnancy or at birth (n = 72). Placental DNA methylation at the C19MC imprinting control region (ICR) was quantified by bisulfite pyrosequencing. Genotyping of the SNP was performed using restriction fragment length polymorphisms. The children's body size and composition were reassessed at age 6 years (n = 32). RESULTS Lower levels of placental C19MC methylation were associated with increased body size of mother, specifically with higher pregestational and predelivery weights and height of the mother (β from -0.294 to -0.371; R2 from 0.04 to 0.10 and all p < 0.019), and with higher weight, height, waist and hip circumferences, and fat mass of the child (β from -0.428 to -0.552; R2 from 0.33 to 0.56 and all p < 0.009). Parental transmission of the SNP did not correlate with an altered placental methylation status at the C19MC ICR. CONCLUSIONS Increased maternal size is associated with reduced placental C19MC methylation, which, in turn, relate to larger body size of the child.
Collapse
Affiliation(s)
- Anna Prats-Puig
- Department of Pediatrics, Girona Institute for Biomedical Research (IDIBGI), Salt, Spain.,Department of Physical Therapy, EUSES University of Girona, Salt Girona, Spain
| | - Sílvia Xargay-Torrent
- Department of Pediatrics, Girona Institute for Biomedical Research (IDIBGI), Salt, Spain
| | - Gemma Carreras-Badosa
- Department of Pediatrics, Girona Institute for Biomedical Research (IDIBGI), Salt, Spain
| | - Berta Mas-Parés
- Department of Pediatrics, Girona Institute for Biomedical Research (IDIBGI), Salt, Spain
| | - Judit Bassols
- Department of Pediatrics, Girona Institute for Biomedical Research (IDIBGI), Salt, Spain
| | - Clive J Petry
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Michael Girardot
- Institute of Molecular Genetics (IGMM), CNRS, University of Montpellier, Montpellier, France
| | - Francis D E Zegher
- Department of Development & Regeneration, University of Leuven, Leuven, Belgium
| | - Lourdes Ibáñez
- Endocrinology, Institut de Recerca Pediàtrica Sant Joan de Déu, University of Barcelona, Esplugues, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - David B Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Robert Feil
- Institute of Molecular Genetics (IGMM), CNRS, University of Montpellier, Montpellier, France
| | - Abel López-Bermejo
- Department of Pediatrics, Girona Institute for Biomedical Research (IDIBGI), Salt, Spain. .,Department of Pediatrics, Dr. Josep Trueta Hospital, Girona, Spain.
| |
Collapse
|
5
|
Artiles KL, Fire AZ, Frøkjær-Jensen C. Assessment and Maintenance of Unigametic Germline Inheritance for C. elegans. Dev Cell 2019; 48:827-839.e9. [PMID: 30799227 PMCID: PMC6435406 DOI: 10.1016/j.devcel.2019.01.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 11/06/2018] [Accepted: 01/21/2019] [Indexed: 12/22/2022]
Abstract
The recent work of Besseling and Bringmann (2016) identified a molecular intervention for C. elegans in which premature segregation of maternal and paternal chromosomes in the fertilized oocyte can produce viable animals exhibiting a non-Mendelian inheritance pattern. Overexpression in embryos of a single protein regulating chromosome segregation (GPR-1) provides a germline derived clonally from a single parental gamete. We present a collection of strains and cytological assays to consistently generate and track non-Mendelian inheritance. These tools allow reproducible and high-frequency (>80%) production of non-Mendelian inheritance, the facile and simultaneous homozygosis for all nuclear chromosomes in a single generation, the precise exchange of nuclear and mitochondrial genomes between strains, and the assessments of non-canonical mitosis events. We show the utility of these strains by demonstrating a rapid assessment of cell lineage requirements (AB versus P1) for a set of genes (lin-2, lin-3, lin-12, and lin-31) with roles in C. elegans vulval development.
Collapse
Affiliation(s)
- Karen L Artiles
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew Z Fire
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Christian Frøkjær-Jensen
- King Abdullah University of Science and Technology, Biological and Environmental Science and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal 23955-6900, Saudi Arabia.
| |
Collapse
|
6
|
Cuellar Partida G, Laurin C, Ring SM, Gaunt TR, McRae AF, Visscher PM, Montgomery GW, Martin NG, Hemani G, Suderman M, Relton CL, Davey Smith G, Evans DM. Genome-wide survey of parent-of-origin effects on DNA methylation identifies candidate imprinted loci in humans. Hum Mol Genet 2018; 27:2927-2939. [PMID: 29860447 PMCID: PMC6077796 DOI: 10.1093/hmg/ddy206] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/23/2018] [Indexed: 12/14/2022] Open
Abstract
Genomic imprinting is an epigenetic mechanism leading to parent-of-origin silencing of alleles. So far, the precise number of imprinted regions in humans is uncertain. In this study, we leveraged genome-wide DNA methylation in whole blood measured longitudinally at three time points (birth, childhood and adolescence) and genome-wide association studies (GWAS) data in 740 mother-child duos from the Avon Longitudinal Study of parents and children to identify candidate imprinted loci. We reasoned that cis-meQTLs at genomic regions that were imprinted would show strong evidence of parent-of-origin associations with DNA methylation, enabling the detection of imprinted regions. Using this approach, we identified genome-wide significant cis-meQTLs that exhibited parent-of-origin effects (POEs) at 82 loci, 34 novel and 48 regions previously implicated in imprinting (3.7-10<P < 10-300). Using an independent dataset from the Brisbane Systems Genetic Study, we replicated 76 out of the 82 identified loci. POEs were remarkably consistent across time points and were so strong at some loci that methylation levels enabled good discrimination of parental transmissions at these and surrounding genomic regions. The implication is that parental allelic transmissions could be modelled at many imprinted (and linked) loci in GWAS of unrelated individuals given a combination of genetic and methylation data. Novel regions showing parent of origin effects on methylation will require replication using a different technology and further functional experiments to confirm that such effects arise through a genomic imprinting mechanism.
Collapse
Affiliation(s)
- Gabriel Cuellar Partida
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Charles Laurin
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Susan M Ring
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tom R Gaunt
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Allan F McRae
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Peter M Visscher
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia.,Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Grant W Montgomery
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia.,Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | | | - Gibran Hemani
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Matthew Suderman
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Caroline L Relton
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - George Davey Smith
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - David M Evans
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia.,Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
7
|
Jiao C, Zhang C, Dai R, Xia Y, Wang K, Giase G, Chen C, Liu C. Positional effects revealed in Illumina methylation array and the impact on analysis. Epigenomics 2018; 10:643-659. [PMID: 29469594 PMCID: PMC6021926 DOI: 10.2217/epi-2017-0105] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 01/17/2018] [Indexed: 12/18/2022] Open
Abstract
AIM We aimed to prove the existence of positional effects in the Illumina methylation beadchip data and to find an optimal correction method. MATERIALS & METHODS Three HumanMethylation450, three HumanMethylation27 datasets and two EPIC datasets were analyzed. ComBat, linear regression, functional normalization and single-sample Noob were used for minimizing positional effects. The corrected results were evaluated by four methods. RESULTS We detected 52,988 CpG loci significantly associated with sample positions, 112 remained after ComBat correction in the primary dataset. The pre- and postcorrection comparisons indicate the positional effects could alter the measured methylation values and downstream analysis results. CONCLUSION Positional effects exist in the Illumina methylation array and may bias the analyses. Using ComBat to correct positional effects is recommended.
Collapse
Affiliation(s)
- Chuan Jiao
- Center for Medical Genetics, Central South University, Changsha, Hunan 410012, PR China
| | - Chunling Zhang
- Department of Neurology and Physiology, SUNY Upstate Medical University, Syracuse, NY 13201, USA
| | - Rujia Dai
- Center for Medical Genetics, Central South University, Changsha, Hunan 410012, PR China
| | - Yan Xia
- Center for Medical Genetics, Central South University, Changsha, Hunan 410012, PR China
| | - Kangli Wang
- Center for Medical Genetics, Central South University, Changsha, Hunan 410012, PR China
| | - Gina Giase
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Chao Chen
- Center for Medical Genetics, Central South University, Changsha, Hunan 410012, PR China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan 410012, PR China
| | - Chunyu Liu
- Center for Medical Genetics, Central South University, Changsha, Hunan 410012, PR China
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13201, USA
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Epigenetics is defined as mitotically heritable changes in gene expression that do not directly alter the DNA sequence. By implication, such epigenetic changes are non-genetically determined, although they can be affected by inherited genetic variation. Extensive evidence indicates that autoimmune diseases including type 1 diabetes are determined by the interaction of genetic and non-genetic factors. Much is known of the genetic causes of these diseases, but the non-genetic effects are less clear-cut. Further, it remains unclear how they interact to cause the destructive autoimmune process. This review identifies the key issues in the genetic/non-genetic interaction, examining the most recent evidence of the role of non-genetic effects in the disease process, including the impact of epigenetic effects on key pathways. RECENT FINDINGS Recent research indicates that these pathways likely involve immune effector cells both of the innate and adaptive immune response. Specifically, there is evidence of cell type-specific enrichment in altered DNA methylation, changes which were temporally stable and enriched at gene regulatory elements. Epigenomics remains in its infancy, and we anticipate further studies will define how the interaction of genetic and non-genetic effects induces tissue-specific destruction and enhances our ability to predict, and possibly even modify that process.
Collapse
Affiliation(s)
| | - Mary N. Dang
- Queen Mary University of London, Mile End Rd, London, E1 4NS UK
| | - R. David Leslie
- Queen Mary University of London, Mile End Rd, London, E1 4NS UK
- The Blizard Institute, London, UK
| |
Collapse
|
9
|
Zhang H, Gelernter J. Review: DNA methylation and alcohol use disorders: Progress and challenges. Am J Addict 2016; 26:502-515. [PMID: 27759945 DOI: 10.1111/ajad.12465] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 09/17/2016] [Accepted: 10/02/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Risk for alcohol use disorders (AUDs) is influenced by gene-environment interactions. Environmental factors can affect gene expression through epigenetic mechanisms such as DNA methylation. This review outlines the findings regarding the association of DNA methylation and AUDs. METHODS We searched PubMed (by April 2016) and identified 29 studies that examined the association of DNA methylation and AUDs. We also evaluated the methods used in these studies. RESULTS Two studies demonstrated elevated global (repetitive element) DNA methylation levels in AUD subjects. Fifteen candidate gene studies showed hypermethylation of promoter regions of six genes (AVP, DNMT3B, HERP, HTR3A, OPRM1, and SNCA) or hypomethylation of the GDAP1 promoter region in AUD subjects. Five genome-wide DNA methylation studies demonstrated widespread DNA methylation changes across the genome in AUD subjects. Six studies showed significant correlations of DNA methylation with gene expression in AUD subjects. Three studies revealed interactive effects of genetic variation and DNA methylation on susceptibility to AUDs. Most studies analyzed AUD-associated DNA methylation changes in the peripheral blood; a few studies examined DNA methylation changes in postmortem brains of AUD subjects. DISCUSSION AND CONCLUSIONS Chronic alcohol consumption may result in DNA methylation changes, leading to neuroadaptations that may underlie some of the mechanisms of AUD risk and persistence. Future studies are needed to confirm the few existing results, and then to elucidate whether DNA methylation changes are the cause or consequence of AUDs. SCIENTIFIC SIGNIFICANCE DNA methylation profiles may be used to assess AUD status or monitor AUD treatment response. (Am J Addict 2017;26:502-515).
Collapse
Affiliation(s)
- Huiping Zhang
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,VA Connecticut Healthcare System, West Haven, Connecticut
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,VA Connecticut Healthcare System, West Haven, Connecticut.,Department of Genetics, Yale University School of Medicine, New Haven, Connecticut.,Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
10
|
Tsanov N, Samacoits A, Chouaib R, Traboulsi AM, Gostan T, Weber C, Zimmer C, Zibara K, Walter T, Peter M, Bertrand E, Mueller F. smiFISH and FISH-quant - a flexible single RNA detection approach with super-resolution capability. Nucleic Acids Res 2016; 44:e165. [PMID: 27599845 PMCID: PMC5159540 DOI: 10.1093/nar/gkw784] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/26/2016] [Indexed: 11/12/2022] Open
Abstract
Single molecule FISH (smFISH) allows studying transcription and RNA localization by imaging individual mRNAs in single cells. We present smiFISH (single molecule inexpensive FISH), an easy to use and flexible RNA visualization and quantification approach that uses unlabelled primary probes and a fluorescently labelled secondary detector oligonucleotide. The gene-specific probes are unlabelled and can therefore be synthesized at low cost, thus allowing to use more probes per mRNA resulting in a substantial increase in detection efficiency. smiFISH is also flexible since differently labelled secondary detector probes can be used with the same primary probes. We demonstrate that this flexibility allows multicolor labelling without the need to synthesize new probe sets. We further demonstrate that the use of a specific acrydite detector oligonucleotide allows smiFISH to be combined with expansion microscopy, enabling the resolution of transcripts in 3D below the diffraction limit on a standard microscope. Lastly, we provide improved, fully automated software tools from probe-design to quantitative analysis of smFISH images. In short, we provide a complete workflow to obtain automatically counts of individual RNA molecules in single cells.
Collapse
Affiliation(s)
- Nikolay Tsanov
- Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France.,Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France
| | - Aubin Samacoits
- Unité Imagerie et Modélisation, Institut Pasteur and CNRS UMR 3691, 28 rue du Docteur Roux, 75015 Paris, France.,C3BI, USR 3756 IP CNRS - Paris, France
| | - Racha Chouaib
- Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France.,Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France.,ER045, Laboratory of Stem Cells, DSST, PRASE, Lebanese University, Beirut, Lebanon
| | - Abdel-Meneem Traboulsi
- Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France.,Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| | - Thierry Gostan
- Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France.,Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| | - Christian Weber
- Unité Imagerie et Modélisation, Institut Pasteur and CNRS UMR 3691, 28 rue du Docteur Roux, 75015 Paris, France.,C3BI, USR 3756 IP CNRS - Paris, France
| | - Christophe Zimmer
- Unité Imagerie et Modélisation, Institut Pasteur and CNRS UMR 3691, 28 rue du Docteur Roux, 75015 Paris, France.,C3BI, USR 3756 IP CNRS - Paris, France
| | - Kazem Zibara
- ER045, Laboratory of Stem Cells, DSST, PRASE, Lebanese University, Beirut, Lebanon.,Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Thomas Walter
- MINES ParisTech, PSL-Research University, CBIO-Centre for Computational Biology, 77300 Fontainebleau, France.,Institut Curie, 75248 Paris Cedex, France.,INSERM, U900, 75248 Paris Cedex, France
| | - Marion Peter
- Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France .,Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| | - Edouard Bertrand
- Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France .,Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| | - Florian Mueller
- Unité Imagerie et Modélisation, Institut Pasteur and CNRS UMR 3691, 28 rue du Docteur Roux, 75015 Paris, France .,C3BI, USR 3756 IP CNRS - Paris, France
| |
Collapse
|
11
|
Li N, Ding YU, Yu T, Li J, Shen Y, Wang X, Fu Q, Shen Y, Huang X, Wang J. Causal variants screened by whole exome sequencing in a patient with maternal uniparental isodisomy of chromosome 10 and a complicated phenotype. Exp Ther Med 2016; 11:2247-2253. [PMID: 27284308 PMCID: PMC4887894 DOI: 10.3892/etm.2016.3241] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 02/11/2016] [Indexed: 11/18/2022] Open
Abstract
Uniparental disomy (UPD), which is the abnormal situation in which both copies of a chromosomal pair have been inherited from one parent, may cause clinical abnormalities by affecting genomic imprinting or causing autosomal recessive variation. Whole Exome Sequencing (WES) and chromosomal microarray analysis (CMA) are powerful technologies used to search for underlying causal variants. In the present study, WES was used to screen for candidate causal variants in the genome of a Chinese pediatric patient, who had been shown by CMA to have maternal uniparental isodisomy of chromosome 10. This was associated with numerous severe medical problems, including bilateral deafness, binocular blindness, stunted growth and leukoderma. A total of 13 rare homozygous variants of these genes were identified on chromosome 10. These included a classical splice variant in the HPS1 gene (c.398+5G>A), which causes Hermansky-Pudlak syndrome type 1 and may explain the patient's ocular and dermal disorders. In addition, six likely pathogenic genes on other chromosomes were found to be associated with the subject's ocular and aural disorders by phenotypic analysis. The results of the present study demonstrated that WES and CMA may be successfully combined in order to identify candidate causal genes. Furthermore, a connection between phenotype and genotype was established in this patient.
Collapse
Affiliation(s)
- Niu Li
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| | - Y U Ding
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| | - Tingting Yu
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| | - Juan Li
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| | - Yongnian Shen
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| | - Xiumin Wang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| | - Qihua Fu
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China; Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| | - Yiping Shen
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China; Boston Children's Hospital, Boston, MA 02115, USA
| | - Xiaodong Huang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| | - Jian Wang
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China; Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| |
Collapse
|
12
|
Cacabelos R, Torrellas C. Epigenetics of Aging and Alzheimer's Disease: Implications for Pharmacogenomics and Drug Response. Int J Mol Sci 2015; 16:30483-543. [PMID: 26703582 PMCID: PMC4691177 DOI: 10.3390/ijms161226236] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/16/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023] Open
Abstract
Epigenetic variability (DNA methylation/demethylation, histone modifications, microRNA regulation) is common in physiological and pathological conditions. Epigenetic alterations are present in different tissues along the aging process and in neurodegenerative disorders, such as Alzheimer’s disease (AD). Epigenetics affect life span and longevity. AD-related genes exhibit epigenetic changes, indicating that epigenetics might exert a pathogenic role in dementia. Epigenetic modifications are reversible and can potentially be targeted by pharmacological intervention. Epigenetic drugs may be useful for the treatment of major problems of health (e.g., cancer, cardiovascular disorders, brain disorders). The efficacy and safety of these and other medications depend upon the efficiency of the pharmacogenetic process in which different clusters of genes (pathogenic, mechanistic, metabolic, transporter, pleiotropic) are involved. Most of these genes are also under the influence of the epigenetic machinery. The information available on the pharmacoepigenomics of most drugs is very limited; however, growing evidence indicates that epigenetic changes are determinant in the pathogenesis of many medical conditions and in drug response and drug resistance. Consequently, pharmacoepigenetic studies should be incorporated in drug development and personalized treatments.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, 15165-Bergondo, Corunna, Spain.
- Chair of Genomic Medicine, Camilo José Cela University, 28692-Madrid, Spain.
| | - Clara Torrellas
- EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, 15165-Bergondo, Corunna, Spain.
- Chair of Genomic Medicine, Camilo José Cela University, 28692-Madrid, Spain.
| |
Collapse
|
13
|
Foley DL, Mackinnon A, Morgan VA, Watts GF, Castle DJ, Waterreus A, Galletly CA. Effect of age, family history of diabetes, and antipsychotic drug treatment on risk of diabetes in people with psychosis: a population-based cross-sectional study. Lancet Psychiatry 2015; 2:1092-8. [PMID: 26477242 DOI: 10.1016/s2215-0366(15)00276-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/09/2015] [Accepted: 06/09/2015] [Indexed: 01/25/2023]
Abstract
BACKGROUND Psychosis is associated with an increased risk of diabetes mellitus. A positive synergy between antipsychotic drug effects and a pre-existing liability to diabetes mellitus might explain the especially high relative risk of diabetes mellitus in young adults with psychosis. We aimed to assess the individual and joint effect of age, family history of diabetes mellitus, and currently prescribed antipsychotic drug treatment on risk for diabetes mellitus. METHODS In this study, we used data from the 2010 Australian National Survey of Psychosis-an observational study done at seven sites in five Australian states. We included data from 1155 people with psychosis aged 18-64 years who were in contact with psychiatric services and who gave a fasting blood sample to test for current diabetes mellitus. Using logistic regression, we modelled the association of diabetes mellitus with age, family history of diabetes mellitus, and current antipsychotic drug treatment. We compared model fit with and without two-way and three-way interaction terms and used likelihood ratio tests to establish which terms to include in the final model. FINDINGS After adjustment for older age, which was an independent risk factor, compared with not taking antipsychotic drugs, antipsychotic drug treatment was associated with diabetes mellitus only in those without a family history of diabetes mellitus (clozapine adjusted odds ratio [OR] 7·22, 95% CI 1·62-32·20, p=0·01; quetiapine 5·91, 1·33-26·30, p=0·02; aripiprazole 5·06, 0·86-29·64, p=0·07; risperidone 4·17, 0·90-19·24, p=0·07; and olanzapine 2·23, 0·45-11·06, p=0·32). Antipsychotic drug treatment was not associated with additional risk of diabetes mellitus in those with a family history (clozapine adjusted OR 1·51, 95% CI 0·64-3·54, p=0·34; quetiapine 1·09, 0·49-2·43, p=0·82; aripiprazole 0·43, 0·12-1·49, p=0·18; risperidone 1·12, 0·48-2·63, p=0·79; and olanzapine 0·67, 0·26-1·71, p=0·39). INTERPRETATION People with psychosis are at increased risk of diabetes mellitus if they have a family history of diabetes mellitus or if they have no family history of diabetes mellitus but are taking antipsychotic drugs. Increasing age increases risk but independently of family history or antipsychotic drug treatment. Clinicians should not think the absence of a family history of diabetes mellitus protects their patients from the diabetic side-effects of antipsychotics. FUNDING Australian Federal Government and Orygen.
Collapse
Affiliation(s)
- Debra L Foley
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia; Centre for Youth Mental Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, VIC, Australia.
| | - Andrew Mackinnon
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia; Centre for Youth Mental Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, VIC, Australia
| | - Vera A Morgan
- Neuropsychiatric Epidemiology Research Unit, School of Psychiatry and Clinical Neurosciences, University of Western Australia, WA, Australia
| | - Gerald F Watts
- Lipid Disorders Clinic, Metabolic Research Centre and Department of Internal Medicine, Royal Perth Hospital and School of Medicine and Pharmacology, University of Western Australia, WA, Australia
| | - David J Castle
- Department of Psychiatry, University of Melbourne, VIC, Australia; St Vincent's Hospital, Melbourne, VIC, Australia
| | - Anna Waterreus
- Neuropsychiatric Epidemiology Research Unit, School of Psychiatry and Clinical Neurosciences, University of Western Australia, WA, Australia
| | - Cherrie A Galletly
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia; Ramsay Health Care SA, Adelaide, SA, Australia; Mental Health Services, Adelaide, SA, Australia; Northern Adelaide Local Health Network, SA, Australia
| |
Collapse
|
14
|
Isles AR. Neural and behavioral epigenetics; what it is, and what is hype. GENES BRAIN AND BEHAVIOR 2015; 14:64-72. [PMID: 25346298 DOI: 10.1111/gbb.12184] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/24/2014] [Accepted: 10/24/2014] [Indexed: 12/13/2022]
Abstract
The ability to examine epigenetic mechanisms in the brain has become readily available over the last 20 years. This has led to an explosion of research and interest in neural and behavioral epigenetics. Of particular interest to researchers, and indeed the lay public, is the possibility that epigenetic processes, such as changes in DNA-methylation and histone modification, may provide a biochemical record of environmental effects. This has led to some fascinating insights into how molecular changes in the brain can control behavior. However, some of this research has also attracted controversy and, as is dealt with here, some overblown claims. This latter problem is partly linked to the shifting sands of what is defined as 'epigenetics'. In this review, I provide an overview of what exactly epigenetics is, and what is hype, with the aim of opening up a debate as to how this exciting field moves forward.
Collapse
Affiliation(s)
- A R Isles
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, UK
| |
Collapse
|
15
|
Lalevée S, Feil R. Long noncoding RNAs in human disease: emerging mechanisms and therapeutic strategies. Epigenomics 2015; 7:877-9. [PMID: 26418705 DOI: 10.2217/epi.15.55] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Sébastien Lalevée
- Institute of Molecular Genetics (IGMM), Centre National de Recherche Scientifique (CNRS), 1919 Route de Mende, 34293 Montpellier, France.,The University of Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| | - Robert Feil
- Institute of Molecular Genetics (IGMM), Centre National de Recherche Scientifique (CNRS), 1919 Route de Mende, 34293 Montpellier, France.,The University of Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| |
Collapse
|
16
|
Molecular insights into transgenerational non-genetic inheritance of acquired behaviours. Nat Rev Genet 2015; 16:641-52. [PMID: 26416311 DOI: 10.1038/nrg3964] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Behavioural traits in mammals are influenced by environmental factors, which can interact with the genome and modulate its activity by complex molecular interplay. Environmental experiences can modify social, emotional and cognitive behaviours during an individual's lifetime, and result in acquired behavioural traits that can be transmitted to subsequent generations. This Review discusses the concept of, and experimental support for, non-genetic transgenerational inheritance of acquired traits involving the germ line in mammals. Possible mechanisms of induction and maintenance during development and adulthood are considered along with an interpretation of recent findings showing the involvement of epigenetic modifications and non-coding RNAs in male germ cells.
Collapse
|
17
|
Krefft M, Frydecka D, Adamowski T, Misiak B. From Prader-Willi syndrome to psychosis: translating parent-of-origin effects into schizophrenia research. Epigenomics 2015; 6:677-88. [PMID: 25531260 DOI: 10.2217/epi.14.52] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Prader-Willi syndrome (PWS) is a relatively rare disorder that originates from paternally inherited deletions and maternal disomy (mUPD) within the 15q11-q13 region or alterations in the PWS imprinting center. Evidence is accumulating that mUPD underlies high prevalence of psychosis among PWS patients. Several genes involved in differentiation and survival of neurons as well as neurotransmission known to act in the development of PWS have been also implicated in schizophrenia. In this article, we provide an overview of genetic and epigenetic underpinnings of psychosis in PWS indicating overlapping points in the molecular background of PWS and schizophrenia. Simultaneously, we highlight the need for studies investigating genetic and epigenetic makeup of the 15q11-q13 in schizophrenia indicating promising candidate genes.
Collapse
Affiliation(s)
- Maja Krefft
- Department of Psychiatry, 10 Pasteur Street, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | | | | | | |
Collapse
|
18
|
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
|
19
|
Cacabelos R. Epigenomic networking in drug development: from pathogenic mechanisms to pharmacogenomics. Drug Dev Res 2015; 75:348-65. [PMID: 25195579 DOI: 10.1002/ddr.21219] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Different epigenetic alterations (DNA methylation, histone modifications, chromatin remodeling, noncoding RNA dysregulation) are associated with the phenotypic expression of complex disorders in which genomic, epigenomic, proteomic, and metabolomic changes, in conjunction with environmental factors, are involved. As epigenetic modifications are reversible and can be potentially targeted by pharmacological and dietary interventions, a series of epigenetic drugs have been developed, including DNA methyltransferase inhibitors (nucleoside analogs, small molecules, bioproducts, antisense oligonucleotides, miRNAs), histone deacetylase inhibitors (short-chain fatty acids, hydroxamic acids, cyclic peptides, benzamides, ketones, sirtuin inhibitors, sirtuin activators), histone acetyltransferase modulators, histone methyltransferase inhibitors, histone demethylase inhibitors, and noncoding RNAs (miRNAs), with potential effects against myelodysplastic syndromes, different types of cancer, and neurodegenerative disorders. Pharmacogenetic and pharmacoepigenetic studies are required for the proper evaluation of efficacy and safety issues in clinical trials with epigenetic drugs.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Genomic Medicine, Camilo José Cela University, Madrid, 28692, Spain; EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, Corunna, 15165, Spain
| |
Collapse
|
20
|
Van Soom A, Peelman L, Holt WV, Fazeli A. An introduction to epigenetics as the link between genotype and environment: a personal view. Reprod Domest Anim 2015; 49 Suppl 3:2-10. [PMID: 25220743 DOI: 10.1111/rda.12341] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2014] [Indexed: 12/18/2022]
Abstract
Lamarck was one of the first scientists who attempted to explain evolution, and he is especially well known for formulating the concept that acquired characteristics can be transmitted to future generations and may therefore steer evolution. Although Lamarckism fell out of favour soon after the publication of Darwin's work on natural selection and evolution, the concept of transmission of acquired characteristics has recently gained renewed attention and has led to some rethinking of the standard evolutionary model. Epigenetics, or the study of heritable (mitotically and/or meiotically) changes in gene activity that are not brought about by changes in the DNA sequence, can explain some types of ill health in offspring, which have been exposed to stressors during early development, when DNA is most susceptible to such epigenetic influences. In this review, we explain briefly the history of epigenetics and we propose some examples of epigenetic and transgenerational effects demonstrated in humans and animals. Growing evidence is available that the health and phenotype of a given individual is already shaped shortly before and after the time of conception. Some evidence suggests that epigenetic markings, which have been established around conception, can also be transmitted to future generations. This knowledge can possibly be used to revolutionize animal breeding and to increase human and animal health worldwide.
Collapse
Affiliation(s)
- A Van Soom
- Faculty of Veterinary Medicine, Department of Reproduction, Obstetrics and Herd Health, Ghent University, Merelbeke, Belgium
| | | | | | | |
Collapse
|
21
|
Genome-wide methylation analysis in Silver-Russell syndrome patients. Hum Genet 2015; 134:317-332. [PMID: 25563730 DOI: 10.1007/s00439-014-1526-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/29/2014] [Indexed: 01/09/2023]
Abstract
Silver-Russell syndrome (SRS) is a clinically heterogeneous disorder characterised by severe in utero growth restriction and poor postnatal growth, body asymmetry, irregular craniofacial features and several additional minor malformations. The aetiology of SRS is complex and current evidence strongly implicates imprinted genes. Approximately, half of all patients exhibit DNA hypomethylation at the H19/IGF2 imprinted domain, and around 10% have maternal uniparental disomy of chromosome 7. We measured DNA methylation in 18 SRS patients at >485,000 CpG sites using DNA methylation microarrays. Using a novel bioinformatics methodology specifically designed to identify subsets of patients with a shared epimutation, we analysed methylation changes genome-wide as well as at known imprinted regions to identify SRS-associated epimutations. Our analysis identifies epimutations at the previously characterised domains of H19/IGF2 and at imprinted regions on chromosome 7, providing proof of principle that our methodology can detect DNA methylation changes at imprinted loci. In addition, we discovered two novel epimutations associated with SRS and located at imprinted loci previously linked to relevant mouse and human phenotypes. We identify RB1 as an additional imprinted locus associated with SRS, with a region near the RB1 differentially methylated region hypermethylated in 13/18 (~70%) patients. We also report 6/18 (~33%) patients were hypermethylated at a CpG island near the ANKRD11 gene. We do not observe consistent co-occurrence of epimutations at multiple imprinted loci in single SRS individuals. SRS is clinically heterogeneous and the absence of multiple imprinted loci epimutations reflects the heterogeneity at the molecular level. Further stratification of SRS patients by molecular phenotypes might aid the identification of disease causes.
Collapse
|
22
|
Abstract
Genetic studies have been well established for identifying sequence variants associated with phenotypes. With the expanding field of epigenetics, and the growing understanding of epigenetic regulation of gene expression, similar studies can be undertaken to also define associations between epigenetic variation and phenotypes. Of particular interest are imprinted genes, which have parent-of-origin specific regulation and expression, and are key regulators of early development. Herein, we describe methods for examining epigenetic regulation by the two major hallmarks of imprinted genes: differentially methylated regions (DMRs), regulatory DNA sequences with allele specific methylation; and monoallelic expression, the silencing and transcription of opposite alleles in a parent-of-origin specific manner.
Collapse
Affiliation(s)
- David A Skaar
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA.
| | - Randy L Jirtle
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, 53706, USA.,Department of Sport and Exercise Sciences, Institute of Sport and Physical Activity Research (ISPAR), University of Bedfordshire, Bedford, Bedfordshire, MK41 9EA, UK
| |
Collapse
|
23
|
Zhou JH, Zhou QZ, Lyu XM, Zhu T, Chen ZJ, Chen MK, Xia H, Wang CY, Qi T, Li X, Liu CD. The Expression of Cysteine-Rich Secretory Protein 2 (CRISP2) and Its Specific Regulator miR-27b in the Spermatozoa of Patients with Asthenozoospermia1. Biol Reprod 2015; 92:28. [DOI: 10.1095/biolreprod.114.124487] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
24
|
Eggermann T, Binder G, Brioude F, Maher ER, Lapunzina P, Cubellis MV, Bergadá I, Prawitt D, Begemann M. CDKN1C mutations: two sides of the same coin. Trends Mol Med 2014; 20:614-22. [PMID: 25262539 DOI: 10.1016/j.molmed.2014.09.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/13/2014] [Accepted: 09/02/2014] [Indexed: 01/03/2023]
Abstract
Cyclin-dependent kinase (CDK)-inhibitor 1C (CDKN1C) negatively regulates cellular proliferation and it has been shown that loss-of-function mutations in the imprinted CDKN1C gene (11p15.5) are associated with the overgrowth disorder Beckwith-Wiedemann syndrome (BWS). With recent reports of gain-of-function mutations of the PCNA domain of CDKN1C in growth-retarded patients with IMAGe syndrome or Silver-Russell syndrome (SRS), its key role for growth has been confirmed. Thereby, the last gap in the spectrum of molecular alterations in 11p15.5 in growth-retardation and overgrowth syndromes could be closed. Recent functional studies explain the strict association of CDKN1C mutations with clinically opposite phenotypes and thereby contribute to our understanding of the function and regulation of the gene in particular and epigenetic regulation in general.
Collapse
Affiliation(s)
- Thomas Eggermann
- Institute of Human Genetics, University Hospital, Technical University Aachen, Aachen, Germany.
| | - Gerhard Binder
- University Children's Hospital, Paediatric Endocrinology, University of Tübingen, Tübingen, Germany
| | - Frédéric Brioude
- AP-HP, Hôpital Armand Trousseau, Explorations Fonctionnelles Endocriniennes, Paris, France
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, Cambridge, UK; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Pablo Lapunzina
- INGEMM, Instituto de Genética Médica y Molecular, Hospital Universitario La Paz, IdiPAZ, CIBERER-ISCIII, Madrid, Spain
| | | | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Dirk Prawitt
- Molekulare Pädiatrie, Zentrum für Kinder- und Jugendmedizin, Universitätsmedizin Mainz, Mainz, Germany
| | - Matthias Begemann
- Institute of Human Genetics, University Hospital, Technical University Aachen, Aachen, Germany
| |
Collapse
|
25
|
Grybek V, Aubry L, Maupetit-Méhouas S, Le Stunff C, Denis C, Girard M, Linglart A, Silve C. Methylation and transcripts expression at the imprinted GNAS locus in human embryonic and induced pluripotent stem cells and their derivatives. Stem Cell Reports 2014; 3:432-43. [PMID: 25241742 PMCID: PMC4266011 DOI: 10.1016/j.stemcr.2014.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 07/04/2014] [Accepted: 07/07/2014] [Indexed: 02/06/2023] Open
Abstract
Data from the literature indicate that genomic imprint marks are disturbed in human pluripotent stem cells (PSCs). GNAS is an imprinted locus that produces one biallelic (Gsα) and four monoallelic (NESP55, GNAS-AS1, XLsα, and A/B) transcripts due to differential methylation of their promoters (DMR). To document imprinting at the GNAS locus in PSCs, we studied GNAS locus DMR methylation and transcript (NESP55, XLsα, and A/B) expression in human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) derived from two human fibroblasts and their progenies. Results showed that (1) methylation at the GNAS locus DMRs is DMR and cell line specific, (2) changes in allelic transcript expression can be independent of a change in allele-specific DNA methylation, and (3) interestingly, methylation at A/B DMR is correlated with A/B transcript expression. These results indicate that these models are valuable to study the mechanisms controlling GNAS methylation, factors involved in transcript expression, and possibly mechanisms involved in the pathophysiology of pseudohypoparathyroidism type 1B. GNAS locus methylation is DMR and cell line specific in human pluripotent stem cells Allelic transcript expression can be independent of allele-specific DNA methylation A/B transcript expression, a key for PHP1B, is correlated with A/B DMR methylation
Collapse
Affiliation(s)
- Virginie Grybek
- INSERM U986, Hôpital Bicêtre, Le Kremlin Bicêtre 94276, France
| | - Laetitia Aubry
- UEVE UMR 861, I-Stem, AFM, Evry 91030, France; INSERM UMR 861, I-Stem, AFM, Evry 91030, France
| | | | | | - Cécile Denis
- CECS, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry 91030, France
| | - Mathilde Girard
- CECS, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry 91030, France
| | - Agnès Linglart
- INSERM U986, Hôpital Bicêtre, Le Kremlin Bicêtre 94276, France; Service d'Endocrinologie Pédiatrique, Hôpital Bicêtre-AP-HP, Le Kremlin Bicêtre 94276, France; Centre de Référence des Maladies Rares du Métabolisme Phospho-Calcique Hôpital Bicêtre, Le Kremlin Bicêtre 94276, France
| | - Caroline Silve
- INSERM U986, Hôpital Bicêtre, Le Kremlin Bicêtre 94276, France; Centre de Référence des Maladies Rares du Métabolisme Phospho-Calcique Hôpital Bicêtre, Le Kremlin Bicêtre 94276, France; Laboratoire de Biochimie Hormonale et Génétique, Hôpital Bichat Claude Bernard-AP-HP, Paris 75018, France.
| |
Collapse
|
26
|
[Clinical profile of a cohort of Silver-Russell syndrome patients followed at the Hospital Infantil de México Federico Gómez from 1998 to 2012]. BOLETIN MEDICO DEL HOSPITAL INFANTIL DE MEXICO 2014; 71:218-226. [PMID: 29421254 DOI: 10.1016/j.bmhimx.2014.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 07/09/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Patients with Silver-Russell syndrome suffer from severe intrauterine and postnatal growth retardation, relative macrocephaly and body asymmetry, among other characteristics. It is caused by several genetic and epigenetic mechanisms in 11p15.5 in 40% of the cases and maternal uniparental disomy of chromosome 7 in 10%. METHODS Twenty patients with a diagnosis of Silver-Russell syndrome who were seen at the HIMFG from 1998 to 2012, were evaluated according to international clinical criteria confirming the diagnosis in nine of the subjects. RESULTS All patients showed intrauterine and postnatal growth retardation and short stature, both considered as major criteria of Silver-Russell syndrome. Relative macrocephaly was present in 78% of the patients and asymmetry in 33%. Other characteristics such as renal tubular acidosis were present > 50% of the cases. CONCLUSIONS The clinical diagnosis of Silver-Russell syndrome is complex. Short stature is the main reason for seeking medical attention and is helpful in the identification of a differential diagnosis. This situation underlines the importance of growth and development evaluation of all patients and particularly in those with short stature to identify those cases that may require molecular studies, with implications in management, prognosis and genetic counseling.
Collapse
|
27
|
Amarasekera M, Martino D, Ashley S, Harb H, Kesper D, Strickland D, Saffery R, Prescott SL. Genome-wide DNA methylation profiling identifies a folate-sensitive region of differential methylation upstream of ZFP57-imprinting regulator in humans. FASEB J 2014; 28:4068-76. [PMID: 24891518 DOI: 10.1096/fj.13-249029] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/27/2014] [Indexed: 12/21/2022]
Abstract
Folate intake during pregnancy may affect the regulation of DNA methylation during fetal development. The genomic regions in the offspring that may be sensitive to folate exposure during in utero development have not been characterized. Using genome-scale profiling, we investigated DNA methylation in 2 immune cell types (CD4(+) and antigen-presenting cells) isolated from neonatal cord blood, selected on the basis of in utero folate exposure. High-folate (HF; n=11) and low-folate (LF; n=12) groups were selected from opposite extremes of maternal serum folate levels measured in the last trimester of pregnancy. A comparison of these groups revealed differential methylation at 7 regions across the genome. By far, the biggest effect observed was hypomethylation of a 923 bp region 3 kb upstream of the ZFP57 transcript, a regulator of DNA methylation during development, observed in both cell types. Levels of H3/H4 acetylation at ZFP57 promoter and ZFP57 mRNA expression were higher in CD4(+) cells in the HF group relative to the LF group. Hypomethylation at this region was replicated in an independent sample set. These data suggest that exposure to folate has effects on the regulation of DNA methylation during fetal development, and this may be important for health and disease.
Collapse
Affiliation(s)
- Manori Amarasekera
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| | - David Martino
- Cancer and Disease Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia; Centre for Food and Allergy Research, Melbourne, Victoria, Australia
| | - Sarah Ashley
- Cancer and Disease Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Institute for Medical Research, Monash University, Melbourne, Victoria, Australia
| | - Hani Harb
- Institute of Laboratory Medicine, Pathobiochemistry, and Molecular Diagnostics, Philipps University, Marburg, Germany; and
| | - Dörthe Kesper
- Institute of Laboratory Medicine, Pathobiochemistry, and Molecular Diagnostics, Philipps University, Marburg, Germany; and
| | - Deborah Strickland
- Telethon Institute for Child Health Research, Perth, Western Australia, Australia
| | - Richard Saffery
- Cancer and Disease Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Susan L Prescott
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia;
| |
Collapse
|
28
|
Wang HD, Hou QF, Guo QN, Li T, Wu D, Zhang XP, Chu Y, He M, Xiao H, Guo LJ, Yang K, Liao SX, Zhu BF. DNA methylation study of fetus genome through a genome-wide analysis. BMC Med Genomics 2014; 7:18. [PMID: 24731722 PMCID: PMC3996908 DOI: 10.1186/1755-8794-7-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 04/10/2014] [Indexed: 01/15/2023] Open
Abstract
Background DNA methylation is a crucial epigenetic modification of the genome which is involved in embryonic development, transcription, chromatin structure, X chromosome inactivation, genomic imprinting and chromosome stability. Consistent with these important roles, DNA methylation has been demonstrated to be required for vertebrate early embryogenesis and essential for regulating temporal and spatial expression of genes controlling cell fate and differentiation. Further studies have shown that abnormal DNA methylation is associated with human diseases including the embryonic development diseases. We attempt to study the DNA methylation status of CpG islands in fetus related to fetus growth and development. Methods GeneChip® Human Tiling 2.0R Array set is used for analysis of methylated DNA in a whole-genome wide in 8 pairs amniotic fluid and maternal blood DNA samples. Results We found 1 fetus hypermethylation DNA markers and 4 fetus hypomethylation DNA markers though a Genome-wide analysis. These DNA markers all found to be associated with the critical genes for fetus growth and development (SH2D3C gene, EML3 gene, TRIM71 gene, HOXA3 gene and HOXA5 gene). Conclusions These genes can be used as a biomarker for association studying of embryonic development, pathological pregnancy and so on. The present study has provided new and fundamental insights into the roles that DNA methylation has in embryonic development and in the pathological pregnancy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Shi-Xiu Liao
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, PR China.
| | | |
Collapse
|