1
|
Dastgheib SA, Bahrami R, Golshan-Tafti M, Danaei M, Azizi S, Shahbazi A, Yeganegi M, Shiri A, Masoudi A, Neamatzadeh H. Decoding bronchopulmonary dysplasia in premature infants through an epigenetic lens. Front Med (Lausanne) 2025; 12:1531169. [PMID: 40248086 PMCID: PMC12003331 DOI: 10.3389/fmed.2025.1531169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/05/2025] [Indexed: 04/19/2025] Open
Abstract
This review provides a comprehensive overview of the evolving insights into the epigenetic mechanisms associated with bronchopulmonary dysplasia (BPD). It specifically highlights the roles of DNA methylation, histone modifications, and RNA regulation in the development of BPD in premature infants. BPD results from complex interactions among genetic factors, environmental exposures, and neonatal stressors. Key findings suggest that intrauterine hypoxia, hyperoxia, and nutrition can lead to epigenetic alterations, affecting gene expression and methylation, which may serve as biomarkers for early BPD detection. RUNX3 is identified as a critical transcription factor influencing lung development and inflammation, while changes in DNA methylation and histone dynamics in cord blood are linked to immune dysregulation associated with BPD. The role of m6A RNA methylation regulators from the IGF2BP family affects mRNA stability and gene expression relevant to BPD. Additionally, specific histones and microRNAs, particularly from the miR-17∼92 cluster, are implicated in pulmonary development and vascular regulation. Long non-coding RNAs (lncRNAs), such as MALAT1, also play a role in gene regulation via competitive endogenous RNA networks, indicating their potential as biomarkers and therapeutic targets. The interplay of these epigenetic mechanisms underscores the need for further research to develop targeted interventions aimed at reducing BPD severity and enhancing health outcomes for at-risk neonates.
Collapse
Affiliation(s)
- Seyed Alireza Dastgheib
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Bahrami
- Neonatal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mahsa Danaei
- Department of Obstetrics and Gynecology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Azizi
- Shahid Akbarabadi Clinical Research Development Unit, Iran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Yeganegi
- Department of Obstetrics and Gynecology, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Amirmasoud Shiri
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Masoudi
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
2
|
Inkster AM, Matthews AM, Phung TN, Plaisier SB, Wilson MA, Brown CJ, Robinson WP. Breaking rules: the complex relationship between DNA methylation and X-chromosome inactivation in the human placenta. Biol Sex Differ 2025; 16:18. [PMID: 40038810 DOI: 10.1186/s13293-025-00696-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/28/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The human placenta is distinct from most organs due to its uniquely low-methylated genome. DNA methylation (DNAme) is particularly depleted in the placenta at partially methylated domains and on the inactive X chromosome (Xi) in XX samples. While Xi DNAme is known to be critical for X-chromosome inactivation (XCI) in other tissues, its role in the placenta remains unclear. Understanding X-linked DNAme variation in the placenta may provide insights into XCI and have implications for prenatal development and phenotypic sex differences. METHODS DNAme data were analyzed from over 350 human placental (chorionic villus) samples, along with samples from cord blood, amnion and chorion placental membranes, and fetal somatic tissues. We characterized X chromosome DNAme variation in the placenta relative to sample variables including cell composition, ancestry, maternal age, placental weight, and fetal birth weight, and compared these patterns to other tissues. We also evaluated the relationship between X-linked DNAme and previously reported XCI gene expression status in placenta. RESULTS Our findings confirm that the placenta exhibits significant depletion of DNAme on the Xi compared to other tissues. Additionally, we observe that X chromosome DNAme profiles in the placenta are influenced by cell composition, particularly trophoblast proportion, with minimal DNAme variation across gestation. Notably, low promoter DNAme is observed at most genes on the Xi regardless of XCI status, challenging known associations in somatic tissues between low promoter DNAme and escape from XCI. CONCLUSIONS This study provides evidence that the human placenta has a distinct Xi DNAme landscape, which may inform our understanding of sex differences during prenatal development. Future research should explore the mechanisms underlying the placenta's unique X-linked DNAme profile, and the factors involved in placental XCI maintenance.
Collapse
Affiliation(s)
- Amy M Inkster
- BC Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V6H 3N1, Canada.
- Department of Medical Genetics, University of British Columbia, 4500 Oak St, Vancouver, BC, V6H 3N1, Canada.
| | - Allison M Matthews
- BC Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V6H 3N1, Canada
- Department of Pathology & Laboratory Medicine, University of British Columbia, 221 Wesbrook Mall, Vancouver, BC, V6T 1Z7, Canada
| | - Tanya N Phung
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ, 85281, USA
| | - Seema B Plaisier
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ, 85281, USA
| | - Melissa A Wilson
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ, 85281, USA
| | - Carolyn J Brown
- Department of Medical Genetics, University of British Columbia, 4500 Oak St, Vancouver, BC, V6H 3N1, Canada
| | - Wendy P Robinson
- BC Children's Hospital Research Institute, 950 W 28th Ave, Vancouver, BC, V6H 3N1, Canada
- Department of Medical Genetics, University of British Columbia, 4500 Oak St, Vancouver, BC, V6H 3N1, Canada
| |
Collapse
|
3
|
Jones MJ, Konwar C, Asiimwe R, Dinh L, Razzaghian HR, de Goede O, MacIsaac JL, Morin AM, Kolsun KP, Gervin K, Lyle R, Ng RT, Koestler DC, Felix JF, Lavoie PM, Robinson WP, Mostafavi S, Kobor MS. DNA methylation differences between cord and adult white blood cells reflect postnatal immune cell maturation. Commun Biol 2025; 8:237. [PMID: 39953282 PMCID: PMC11828917 DOI: 10.1038/s42003-025-07661-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Abstract
Epigenetic modifications such as DNA methylation are both cell type and developmental age specific. Here, we show that the immunological maturation of blood cell types influences DNA methylation changes from naive cord blood to fully functional adult blood. Lymphoid cells in adult blood showed more variability than in cord blood suggesting an antigen-dependent maturation of DNA methylation in lymphoid cells throughout the lifespan. Fewer DNA methylation changes between cord and adult blood were observed in myeloid cells, particularly in monocytes, which demonstrated the least number of DNA methylation changes between cord and adult blood. We also noted differences in epigenetic ages by immune cell types within the same individuals, specifically in cord blood where monocytes were epigenetically oldest compared to the other cell types. In addition, we provide a publicly available resource to the community as an R Shiny web application to interactively explore epigenetic patterns between naive cord white blood cells and fully functional adult white blood cells for six immune cell types.
Collapse
Affiliation(s)
- Meaghan J Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Chaini Konwar
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Rebecca Asiimwe
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Louie Dinh
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Hamid Reza Razzaghian
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Olivia de Goede
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Julia L MacIsaac
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Alexander M Morin
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kurt P Kolsun
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Kristina Gervin
- Division of Clinical Neuroscience, Department of Research and Innovation, Oslo University Hospital, Oslo, Norway
| | - Robert Lyle
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Raymond T Ng
- Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Janine F Felix
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Pascal M Lavoie
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Wendy P Robinson
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Sara Mostafavi
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- Canadian Institute for Advanced Research, Toronto, ON, Canada
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Michael S Kobor
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
- BC Children's Hospital Research Institute, Vancouver, BC, Canada.
- Canadian Institute for Advanced Research, Toronto, ON, Canada.
- Edwin S.H. Leong Centre for Healthy Aging, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Yasumizu Y, Hagiwara M, Umezu Y, Fuji H, Iwaisako K, Asagiri M, Uemoto S, Nakamura Y, Thul S, Ueyama A, Yokoi K, Tanemura A, Nose Y, Saito T, Wada H, Kakuda M, Kohara M, Nojima S, Morii E, Doki Y, Sakaguchi S, Ohkura N. Neural-net-based cell deconvolution from DNA methylation reveals tumor microenvironment associated with cancer prognosis. NAR Cancer 2024; 6:zcae022. [PMID: 38751935 PMCID: PMC11094754 DOI: 10.1093/narcan/zcae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/18/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
DNA methylation is a pivotal epigenetic modification that defines cellular identity. While cell deconvolution utilizing this information is considered useful for clinical practice, current methods for deconvolution are limited in their accuracy and resolution. In this study, we collected DNA methylation data from 945 human samples derived from various tissues and tumor-infiltrating immune cells and trained a neural network model with them. The model, termed MEnet, predicted abundance of cell population together with the detailed immune cell status from bulk DNA methylation data, and showed consistency to those of flow cytometry and histochemistry. MEnet was superior to the existing methods in the accuracy, speed, and detectable cell diversity, and could be applicable for peripheral blood, tumors, cell-free DNA, and formalin-fixed paraffin-embedded sections. Furthermore, by applying MEnet to 72 intrahepatic cholangiocarcinoma samples, we identified immune cell profiles associated with cancer prognosis. We believe that cell deconvolution by MEnet has the potential for use in clinical settings.
Collapse
Affiliation(s)
- Yoshiaki Yasumizu
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| | - Masaki Hagiwara
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Basic Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Yuto Umezu
- Faculty of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hiroaki Fuji
- Department of Hepato-Biliary-Pancreatic Surgery, Hyogo Medical University, Nishinomiya, Hyogo, Japan
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Keiko Iwaisako
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Masataka Asagiri
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Shinji Uemoto
- Shiga University Medical Science, Otsu, Shiga, Japan
| | - Yamami Nakamura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Sophia Thul
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Azumi Ueyama
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kazunori Yokoi
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yohei Nose
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Mamoru Kakuda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masaharu Kohara
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shimon Sakaguchi
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Experimental Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto, Japan
| | - Naganari Ohkura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Basic Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
5
|
Song AY, Bulka CM, Niemiec SS, Kechris K, Boyle KE, Marsit CJ, O’Shea TM, Fry RC, Lyall K, Fallin MD, Volk HE, Ladd-Acosta C. Accelerated epigenetic age at birth and child emotional and behavioura development in early childhood: a meta-analysis of four prospective cohort studies in ECHO. Epigenetics 2023; 18:2254971. [PMID: 37691382 PMCID: PMC10496525 DOI: 10.1080/15592294.2023.2254971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/16/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023] Open
Abstract
Background: 'Epigenetic clocks' have been developed to accurately predict chronologic gestational age and have been associated with child health outcomes in prior work.Methods: We meta-analysed results from four prospective U.S cohorts investigating the association between epigenetic age acceleration estimated using blood DNA methylation collected at birth and preschool age Childhood Behavior Checklist (CBCL) scores.Results: Epigenetic ageing was not significantly associated with CBCL total problem scores (β = 0.33, 95% CI: -0.95, 0.28) and DSM-oriented pervasive development problem scores (β = -0.23, 95% CI: -0.61, 0.15). No associations were observed for other DSM-oriented subscales.Conclusions: The meta-analysis results suggest that epigenetic gestational age acceleration is not associated with child emotional and behavioural functioning for preschool age group. These findings may relate to our study population, which includes two cohorts enriched for ASD and one preterm birth cohort.; future work should address the role of epigenetic age in child health in other study populations.Abbreviations: DNAm: DNA methylation; CBCL: Child Behavioral Checklist; ECHO: Environmental Influences on Child Health Outcomes; EARLI: Early Autism Risk Longitudinal Investigation; MARBLES: Markers of Autism Risk in Babies - Learning Early Signs; ELGAN: Extremely Low Gestational Age Newborns; ASD: autism spectrum disorder; BMI: body mass index; DSM: Diagnostic and Statistical Manual of Mental Disorders.
Collapse
Affiliation(s)
- Ashley Y. Song
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Sierra S. Niemiec
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristen E. Boyle
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carmen J. Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - T. Michael O’Shea
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristen Lyall
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | | | - Heather E. Volk
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christine Ladd-Acosta
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
6
|
Mouat JS, Li S, Myint SS, Laufer BI, Lupo PJ, Schraw JM, Woodhouse JP, de Smith AJ, LaSalle JM. Epigenomic signature of major congenital heart defects in newborns with Down syndrome. Hum Genomics 2023; 17:92. [PMID: 37803336 PMCID: PMC10559462 DOI: 10.1186/s40246-023-00540-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Congenital heart defects (CHDs) affect approximately half of individuals with Down syndrome (DS), but the molecular reasons for incomplete penetrance are unknown. Previous studies have largely focused on identifying genetic risk factors associated with CHDs in individuals with DS, but comprehensive studies of the contribution of epigenetic marks are lacking. We aimed to identify and characterize DNA methylation differences from newborn dried blood spots (NDBS) of DS individuals with major CHDs compared to DS individuals without CHDs. METHODS We used the Illumina EPIC array and whole-genome bisulfite sequencing (WGBS) to quantitate DNA methylation for 86 NDBS samples from the California Biobank Program: (1) 45 DS-CHD (27 female, 18 male) and (2) 41 DS non-CHD (27 female, 14 male). We analyzed global CpG methylation and identified differentially methylated regions (DMRs) in DS-CHD versus DS non-CHD comparisons (both sex-combined and sex-stratified) corrected for sex, age of blood collection, and cell-type proportions. CHD DMRs were analyzed for enrichment in CpG and genic contexts, chromatin states, and histone modifications by genomic coordinates and for gene ontology enrichment by gene mapping. DMRs were also tested in a replication dataset and compared to methylation levels in DS versus typical development (TD) WGBS NDBS samples. RESULTS We found global CpG hypomethylation in DS-CHD males compared to DS non-CHD males, which was attributable to elevated levels of nucleated red blood cells and not seen in females. At a regional level, we identified 58, 341, and 3938 CHD-associated DMRs in the Sex Combined, Females Only, and Males Only groups, respectively, and used machine learning algorithms to select 19 Males Only loci that could distinguish CHD from non-CHD. DMRs in all comparisons were enriched for gene exons, CpG islands, and bivalent chromatin and mapped to genes enriched for terms related to cardiac and immune functions. Lastly, a greater percentage of CHD-associated DMRs than background regions were differentially methylated in DS versus TD samples. CONCLUSIONS A sex-specific signature of DNA methylation was detected in NDBS of DS-CHD compared to DS non-CHD individuals. This supports the hypothesis that epigenetics can reflect the variability of phenotypes in DS, particularly CHDs.
Collapse
Affiliation(s)
- Julia S Mouat
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, USA
- Perinatal Origins of Disparities Center, University of California, Davis, CA, USA
- Genome Center, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Shaobo Li
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Swe Swe Myint
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin I Laufer
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, USA
- Perinatal Origins of Disparities Center, University of California, Davis, CA, USA
- Genome Center, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Philip J Lupo
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jeremy M Schraw
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - John P Woodhouse
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Adam J de Smith
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, USA.
- Perinatal Origins of Disparities Center, University of California, Davis, CA, USA.
- Genome Center, University of California, Davis, CA, USA.
- MIND Institute, University of California, Davis, CA, USA.
| |
Collapse
|
7
|
Mouat JS, Li S, Myint SS, Laufer BI, Lupo PJ, Schraw JM, Woodhouse JP, de Smith AJ, LaSalle JM. Epigenomic signature of major congenital heart defects in newborns with Down syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.02.23289417. [PMID: 37205408 PMCID: PMC10187438 DOI: 10.1101/2023.05.02.23289417] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Background Congenital heart defects (CHDs) affect approximately half of individuals with Down syndrome (DS) but the molecular reasons for incomplete penetrance are unknown. Previous studies have largely focused on identifying genetic risk factors associated with CHDs in individuals with DS, but comprehensive studies of the contribution of epigenetic marks are lacking. We aimed to identify and characterize DNA methylation differences from newborn dried blood spots (NDBS) of DS individuals with major CHDs compared to DS individuals without CHDs. Methods We used the Illumina EPIC array and whole-genome bisulfite sequencing (WGBS) to quantitate DNA methylation for 86 NDBS samples from the California Biobank Program: 1) 45 DS-CHD (27 female, 18 male) and 2) 41 DS non-CHD (27 female, 14 male). We analyzed global CpG methylation and identified differentially methylated regions (DMRs) in DS-CHD vs DS non-CHD comparisons (both sex-combined and sex-stratified) corrected for sex, age of blood collection, and cell type proportions. CHD DMRs were analyzed for enrichment in CpG and genic contexts, chromatin states, and histone modifications by genomic coordinates and for gene ontology enrichment by gene mapping. DMRs were also tested in a replication dataset and compared to methylation levels in DS vs typical development (TD) WGBS NDBS samples. Results We found global CpG hypomethylation in DS-CHD males compared to DS non-CHD males, which was attributable to elevated levels of nucleated red blood cells and not seen in females. At a regional level, we identified 58, 341, and 3,938 CHD-associated DMRs in the Sex Combined, Females Only, and Males Only groups, respectively, and used machine learning algorithms to select 19 Males Only loci that could distinguish CHD from non-CHD. DMRs in all comparisons were enriched for gene exons, CpG islands, and bivalent chromatin and mapped to genes enriched for terms related to cardiac and immune functions. Lastly, a greater percentage of CHD-associated DMRs than background regions were differentially methylated in DS vs TD samples. Conclusions A sex-specific signature of DNA methylation was detected in NDBS of DS-CHD compared to DS non-CHD individuals. This supports the hypothesis that epigenetics can reflect the variability of phenotypes in DS, particularly CHDs.
Collapse
Affiliation(s)
- Julia S Mouat
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA USA
- Perinatal Origins of Disparities Center, University of California, Davis, CA USA
- Genome Center, University of California, Davis, CA USA
- MIND Institute, University of California, Davis, CA USA
| | - Shaobo Li
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, CA USA
| | - Swe Swe Myint
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, CA USA
| | - Benjamin I Laufer
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA USA
- Perinatal Origins of Disparities Center, University of California, Davis, CA USA
- Genome Center, University of California, Davis, CA USA
- MIND Institute, University of California, Davis, CA USA
| | - Philip J Lupo
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX USA
| | - Jeremy M Schraw
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX USA
| | - John P Woodhouse
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX USA
| | - Adam J de Smith
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, CA USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA USA
- Perinatal Origins of Disparities Center, University of California, Davis, CA USA
- Genome Center, University of California, Davis, CA USA
- MIND Institute, University of California, Davis, CA USA
| |
Collapse
|
8
|
Staley JR, Windmeijer F, Suderman M, Lyon MS, Davey Smith G, Tilling K. A robust mean and variance test with application to high-dimensional phenotypes. Eur J Epidemiol 2022; 37:377-387. [PMID: 34651232 PMCID: PMC9187575 DOI: 10.1007/s10654-021-00805-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 09/06/2021] [Indexed: 12/01/2022]
Abstract
Most studies of continuous health-related outcomes examine differences in mean levels (location) of the outcome by exposure. However, identifying effects on the variability (scale) of an outcome, and combining tests of mean and variability (location-and-scale), could provide additional insights into biological mechanisms. A joint test could improve power for studies of high-dimensional phenotypes, such as epigenome-wide association studies of DNA methylation at CpG sites. One possible cause of heterogeneity of variance is a variable interacting with exposure in its effect on outcome, so a joint test of mean and variability could help in the identification of effect modifiers. Here, we review a scale test, based on the Brown-Forsythe test, for analysing variability of a continuous outcome with respect to both categorical and continuous exposures, and develop a novel joint location-and-scale score (JLSsc) test. These tests were compared to alternatives in simulations and used to test associations of mean and variability of DNA methylation with gender and gestational age using data from the Accessible Resource for Integrated Epigenomics Studies (ARIES). In simulations, the Brown-Forsythe and JLSsc tests retained correct type I error rates when the outcome was not normally distributed in contrast to the other approaches tested which all had inflated type I error rates. These tests also identified > 7500 CpG sites for which either mean or variability in cord blood methylation differed according to gender or gestational age. The Brown-Forsythe test and JLSsc are robust tests that can be used to detect associations not solely driven by a mean effect.
Collapse
Affiliation(s)
- James R Staley
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Frank Windmeijer
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
- Department of Statistics and Nuffield College, University of Oxford, Oxford, UK
| | - Matthew Suderman
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Matthew S Lyon
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
- National Institute for Health Research Bristol Biomedical Research Centre, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Kate Tilling
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK.
| |
Collapse
|
9
|
Kim B, Sasaki A, Murphy K, Matthews SG. DNA methylation signatures in human neonatal blood following maternal antenatal corticosteroid treatment. Transl Psychiatry 2022; 12:132. [PMID: 35354798 PMCID: PMC8967826 DOI: 10.1038/s41398-022-01902-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 11/09/2022] Open
Abstract
Antenatal corticosteroids (ACS) are used to treat women at risk of preterm birth to improve neonatal survival. Though affected children may be at long-term risk of neurobehavioural disorders, the driving mechanisms remain unknown. Animal studies have shown that ACS exposure can lead to overlapping changes in DNA methylation between the blood and the brain, identifying gene pathways for neurodevelopment, which highlights the potential to examine peripheral blood as a surrogate for inaccessible human brain tissue. We hypothesized that differential methylation will be identified in blood of term-born neonates following ACS. Mother-infant dyads that received ACS were retrospectively identified through the Ontario Birth Study at Sinai Health Complex and matched to untreated controls for maternal age, BMI, parity and foetal sex (n = 14/group). Genome-wide methylation differences were examined at single-nucleotide resolution in DNA extracted from dried bloodspot cards using reduced representative bisulfite sequencing approaches. 505 differentially methylated CpG sites (DMCs) were identified, wherein 231 were hypermethylated and 274 were hypomethylated. These sites were annotated to 219 genes, of which USP48, SH3PXD2A, NTM, CAMK2N2, MAP6D1 were five of the top ten genes with known neurological function. Collectively, the set of hypermethylated genes were enriched for pathways of transcription regulation, while pathways of proteasome activity were enriched among the set of hypomethylated genes. This study is the first to identify DNA methylation changes in human neonatal blood following ACS. Understanding the epigenetic changes that occur in response to ACS will support future investigations to delineate the effects of prenatal glucocorticoid exposure on human development.
Collapse
Affiliation(s)
- Bona Kim
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.
| | - Aya Sasaki
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Kellie Murphy
- Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada
| | - Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Sammallahti S, Cortes Hidalgo AP, Tuominen S, Malmberg A, Mulder RH, Brunst KJ, Alemany S, McBride NS, Yousefi P, Heiss JA, McRae N, Page CM, Jin J, Pesce G, Caramaschi D, Rifas-Shiman SL, Koen N, Adams CD, Magnus MC, Baïz N, Ratanatharathorn A, Czamara D, Håberg SE, Colicino E, Baccarelli AA, Cardenas A, DeMeo DL, Lawlor DA, Relton CL, Felix JF, van IJzendoorn MH, Bakermans-Kranenburg MJ, Kajantie E, Räikkönen K, Sunyer J, Sharp GC, Houtepen LC, Nohr EA, Sørensen TIA, Téllez-Rojo MM, Wright RO, Annesi-Maesano I, Wright J, Hivert MF, Wright RJ, Zar HJ, Stein DJ, London SJ, Cecil CAM, Tiemeier H, Lahti J. Maternal anxiety during pregnancy and newborn epigenome-wide DNA methylation. Mol Psychiatry 2021; 26:1832-1845. [PMID: 33414500 PMCID: PMC8595870 DOI: 10.1038/s41380-020-00976-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/31/2020] [Accepted: 11/30/2020] [Indexed: 01/29/2023]
Abstract
Maternal anxiety during pregnancy is associated with adverse foetal, neonatal, and child outcomes, but biological mechanisms remain unclear. Altered foetal DNA methylation (DNAm) has been proposed as a potential underlying mechanism. In the current study, we performed a meta-analysis to examine the associations between maternal anxiety, measured prospectively during pregnancy, and genome-wide DNAm from umbilical cord blood. Sixteen non-overlapping cohorts from 12 independent longitudinal studies of the Pregnancy And Childhood Epigenetics Consortium participated, resulting in a combined dataset of 7243 mother-child dyads. We examined prenatal anxiety in relation to genome-wide DNAm and differentially methylated regions. We observed no association between the general symptoms of anxiety during pregnancy or pregnancy-related anxiety, and DNAm at any of the CpG sites, after multiple-testing correction. Furthermore, we identify no differentially methylated regions associated with maternal anxiety. At the cohort-level, of the 21 associations observed in individual cohorts, none replicated consistently in the other cohorts. In conclusion, contrary to some previous studies proposing cord blood DNAm as a promising potential mechanism explaining the link between maternal anxiety during pregnancy and adverse outcomes in offspring, we found no consistent evidence for any robust associations between maternal anxiety and DNAm in cord blood. Larger studies and analysis of DNAm in other tissues may be needed to establish subtle or subgroup-specific associations between maternal anxiety and the foetal epigenome.
Collapse
Affiliation(s)
- Sara Sammallahti
- Erasmus MC, University Medical Center Rotterdam, Department of Adolescent and Child Psychiatry and Psychology, Rotterdam, The Netherlands
- Erasmus MC, University Medical Center Rotterdam, Generation R Study Group, Rotterdam, The Netherlands
- Harvard T.H. Chan School of Public Health, Department of Social and Behavioral Science, Boston, MA, USA
- University of Helsinki, Department of Psychology and Logopedics, Helsinki, Finland
| | - Andrea P Cortes Hidalgo
- Erasmus MC, University Medical Center Rotterdam, Department of Adolescent and Child Psychiatry and Psychology, Rotterdam, The Netherlands
- Erasmus MC, University Medical Center Rotterdam, Generation R Study Group, Rotterdam, The Netherlands
| | - Samuli Tuominen
- University of Helsinki, Department of Psychology and Logopedics, Helsinki, Finland
| | - Anni Malmberg
- University of Helsinki, Department of Psychology and Logopedics, Helsinki, Finland
| | - Rosa H Mulder
- Erasmus MC, University Medical Center Rotterdam, Department of Adolescent and Child Psychiatry and Psychology, Rotterdam, The Netherlands
- Erasmus MC, University Medical Center Rotterdam, Generation R Study Group, Rotterdam, The Netherlands
- Leiden University, Institute of Education and Child Studies, Leiden, The Netherlands
| | - Kelly J Brunst
- University of Cincinnati, College of Medicine, Department of Environmental Health, Cincinnati, OH, USA
| | - Silvia Alemany
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Nancy S McBride
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Paul Yousefi
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Jonathan A Heiss
- Icahn School of Medicine at Mount Sinai, Department of Environmental Medicine and Public Health, New York, NY, USA
| | - Nia McRae
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christian M Page
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | | | - Giancarlo Pesce
- INSERM UMR-S 1136, EPAR, Saint-Antoine Medical School, Paris, France
- Sorbonne Université, Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Pierre Louis Institute of Epidemiology and Public Health (IPLESP), Paris, France
| | - Doretta Caramaschi
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Sheryl L Rifas-Shiman
- Harvard Medical School, Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Nastassja Koen
- University of Cape Town, Department of Psychiatry and Mental Health, Cape Town, South Africa
- South African Medical Research Council (SAMRC) Unit on Risk and Resilience in Mental Disorders, Cape Town, South Africa
- University of Cape Town, Neuroscience Institute, Cape Town, South Africa
| | - Charleen D Adams
- Beckman Research Institute of City of Hope, Department of Population Sciences, Duarte, CA, USA
| | - Maria C Magnus
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Nour Baïz
- INSERM UMR-S 1136, EPAR, Saint-Antoine Medical School, Paris, France
- Sorbonne Université, Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Pierre Louis Institute of Epidemiology and Public Health (IPLESP), Paris, France
| | - Andrew Ratanatharathorn
- Columbia University, Department of Epidemiology, New York City, NY, USA
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA
| | - Darina Czamara
- Max-Planck-Institute of Psychiatry, Department of Translational Research in Psychiatry, Munich, Germany
| | - Siri E Håberg
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Elena Colicino
- Icahn School of Medicine at Mount Sinai, Department of Environmental Medicine and Public Health, New York, NY, USA
| | - Andrea A Baccarelli
- Columbia University Mailman School of Public Health, Precision Environmental Health Lab, New York, NY, USA
| | - Andres Cardenas
- University of California, Division of Environmental Health Sciences, School of Public Health, Berkeley, CA, USA
| | - Dawn L DeMeo
- Brigham and Women's Hospital, Channing Division of Network Medicine, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Deborah A Lawlor
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Caroline L Relton
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Janine F Felix
- Erasmus MC, University Medical Center Rotterdam, Generation R Study Group, Rotterdam, The Netherlands
- Erasmus MC, University Medical Center Rotterdam, Department of Pediatrics, Rotterdam, The Netherlands
| | - Marinus H van IJzendoorn
- Erasmus University Rotterdam, Department of Psychology, Education, and Child Studies, Rotterdam, The Netherlands
- University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Marian J Bakermans-Kranenburg
- Leiden University, Leiden Institute for Brain and Cognition, Leiden, The Netherlands
- Vrije Universiteit Amsterdam, Clinical Child & Family Studies, Amsterdam, The Netherlands
| | - Eero Kajantie
- Finnish Institute for Health and Welfare, Helsinki, Finland
- Oulu University Hospital and University of Oulu, PEDEGO Research Unit, MRC Oulu, Oulu, Finland
- Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Norwegian University of Science and Technology, Department of Clinical and Molecular Medicine, Trondheim, Norway
| | - Katri Räikkönen
- University of Helsinki, Department of Psychology and Logopedics, Helsinki, Finland
| | - Jordi Sunyer
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Gemma C Sharp
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Lotte C Houtepen
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Ellen A Nohr
- University of Southern Denmark, Institute of Clinical Research and Department of Gynaecology and Obstetrics, Odense, Denmark
| | - Thorkild I A Sørensen
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
- University of Copenhagen, Faculty of Health and Medical Sciences, Department of Public Health, Copenhagen, Denmark
- University of Copenhagen, Faculty of Medical and Health Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Martha M Téllez-Rojo
- National Institute of Public Health, Center for Nutrition and Health Research, Cuernavaca, Mor, Mexico
| | | | - Isabella Annesi-Maesano
- INSERM UMR-S 1136, EPAR, Saint-Antoine Medical School, Paris, France
- Sorbonne Université, Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Pierre Louis Institute of Epidemiology and Public Health (IPLESP), Paris, France
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals, NHS Foundation Trust, Bradford, UK
| | - Marie-France Hivert
- Harvard Medical School, Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Massachusetts General Hospital, Diabetes Unit, Boston, MA, USA
| | - Rosalind J Wright
- Icahn School of Medicine at Mount Sinai, Environmental Medicine & Public Health, Institute for Exposomic Research, New York, NY, USA
| | - Heather J Zar
- University of Cape Town, Department of Paediatrics and Child Health, Cape Town, South Africa
- South African Medical Research Council (SAMRC) Unit on Child and Adolescent Health, Cape Town, Cape Town, South Africa
| | - Dan J Stein
- University of Cape Town, Department of Psychiatry and Mental Health, Cape Town, South Africa
- South African Medical Research Council (SAMRC) Unit on Risk and Resilience in Mental Disorders, Cape Town, South Africa
- University of Cape Town, Neuroscience Institute, Cape Town, South Africa
| | - Stephanie J London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Charlotte A M Cecil
- Erasmus MC, University Medical Center Rotterdam, Department of Adolescent and Child Psychiatry and Psychology, Rotterdam, The Netherlands
- Erasmus MC, University Medical Center Rotterdam, Generation R Study Group, Rotterdam, The Netherlands
- Erasmus MC, University Medical Center Rotterdam, Department of Epidemiology, Rotterdam, The Netherlands
- Leiden University Medical Center, Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden, The Netherlands
| | - Henning Tiemeier
- Erasmus MC, University Medical Center Rotterdam, Department of Adolescent and Child Psychiatry and Psychology, Rotterdam, The Netherlands.
- Erasmus MC, University Medical Center Rotterdam, Generation R Study Group, Rotterdam, The Netherlands.
- Harvard T.H. Chan School of Public Health, Department of Social and Behavioral Science, Boston, MA, USA.
| | - Jari Lahti
- University of Helsinki, Department of Psychology and Logopedics, Helsinki, Finland
| |
Collapse
|
11
|
Coit P, Schollaert KL, Mirizio EM, Torok KS, Sawalha AH. DNA methylation patterns in juvenile systemic sclerosis and localized scleroderma. Clin Immunol 2021; 228:108756. [PMID: 33992755 DOI: 10.1016/j.clim.2021.108756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/12/2021] [Indexed: 12/16/2022]
Abstract
Scleroderma refers to a group of chronic fibrotic immune-mediated diseases of unknown etiology. Characterizing epigenetic changes in childhood-onset scleroderma, systemic sclerosis or localized scleroderma, has not been previously performed. The aim of this study was to assess DNA methylation differences and similarities between juvenile systemic sclerosis (jSSc) and juvenile localized scleroderma (jLS) compared to matched healthy controls. Genome-wide DNA methylation changes in peripheral blood mononuclear cell samples were assessed using the MethylationEPIC array followed by bioinformatic analysis and limited functional assessment. We identified a total of 105 and 144 differentially methylated sites compared to healthy controls in jSSc and jLS, respectively. The majority of differentially methylated sites and genes represented were unique to either jSSc or jLS suggesting a different underlying epigenetic pattern in both diseases. Among shared differentially methylated genes, methylation levels in a CpG site in FGFR2 can distinguish between LS and healthy PBMCs with a high accuracy. Canonical pathway analysis revealed that inflammatory pathways were enriched in genes differentially methylated in jSSc, including STAT3, NF-κB, and IL-15 pathways. In contrast, the HIPPO signaling pathway was enriched in jLS. Our data also suggest a potential role for NOTCH3 in both jSSc and jLS, and revealed a number of transcription factors unique to each of the two diseases. In summary, our data revealed important insights into jSSc and jLS and suggest a potentially novel epigenetic diagnostic biomarker for LS.
Collapse
Affiliation(s)
- Patrick Coit
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Kaila L Schollaert
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emily M Mirizio
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathryn S Torok
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA; University of Pittsburgh Systemic Sclerosis Center of Research Translation, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Aure MR, Fleischer T, Bjørklund S, Ankill J, Castro-Mondragon JA, Børresen-Dale AL, Tost J, Sahlberg KK, Mathelier A, Tekpli X, Kristensen VN. Crosstalk between microRNA expression and DNA methylation drives the hormone-dependent phenotype of breast cancer. Genome Med 2021; 13:72. [PMID: 33926515 PMCID: PMC8086068 DOI: 10.1186/s13073-021-00880-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 03/26/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Abnormal DNA methylation is observed as an early event in breast carcinogenesis. However, how such alterations arise is still poorly understood. microRNAs (miRNAs) regulate gene expression at the post-transcriptional level and play key roles in various biological processes. Here, we integrate miRNA expression and DNA methylation at CpGs to study how miRNAs may affect the breast cancer methylome and how DNA methylation may regulate miRNA expression. METHODS miRNA expression and DNA methylation data from two breast cancer cohorts, Oslo2 (n = 297) and The Cancer Genome Atlas (n = 439), were integrated through a correlation approach that we term miRNA-methylation Quantitative Trait Loci (mimQTL) analysis. Hierarchical clustering was used to identify clusters of miRNAs and CpGs that were further characterized through analysis of mRNA/protein expression, clinicopathological features, in silico deconvolution, chromatin state and accessibility, transcription factor binding, and long-range interaction data. RESULTS Clustering of the significant mimQTLs identified distinct groups of miRNAs and CpGs that reflect important biological processes associated with breast cancer pathogenesis. Notably, two major miRNA clusters were related to immune or fibroblast infiltration, hence identifying miRNAs associated with cells of the tumor microenvironment, while another large cluster was related to estrogen receptor (ER) signaling. Studying the chromatin landscape surrounding CpGs associated with the estrogen signaling cluster, we found that miRNAs from this cluster are likely to be regulated through DNA methylation of enhancers bound by FOXA1, GATA2, and ER-alpha. Further, at the hub of the estrogen cluster, we identified hsa-miR-29c-5p as negatively correlated with the mRNA and protein expression of DNA methyltransferase DNMT3A, a key enzyme regulating DNA methylation. We found deregulation of hsa-miR-29c-5p already present in pre-invasive breast lesions and postulate that hsa-miR-29c-5p may trigger early event abnormal DNA methylation in ER-positive breast cancer. CONCLUSIONS We describe how miRNA expression and DNA methylation interact and associate with distinct breast cancer phenotypes.
Collapse
Affiliation(s)
- Miriam Ragle Aure
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Thomas Fleischer
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Sunniva Bjørklund
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Jørgen Ankill
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Jaime A. Castro-Mondragon
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jörg Tost
- Laboratory for Epigenetics and Environment, Centre National de Recherche en Génomique Humaine, CEA–Institut de Biologie François Jacob, University Paris-Saclay, Evry, France
| | - Kristine K. Sahlberg
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
- Department of Research, Vestre Viken Hospital Trust, Drammen, Norway
| | - Anthony Mathelier
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Xavier Tekpli
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Vessela N. Kristensen
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
- Department of Clinical Molecular Biology and Laboratory Science (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
13
|
Solomon O, Macisaac JL, Tindula G, Kobor MS, Eskenazi B, Holland N. 5-Hydroxymethylcytosine in cord blood and associations of DNA methylation with sex in newborns. Mutagenesis 2020; 34:315-322. [PMID: 31587037 DOI: 10.1093/mutage/gez023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 08/20/2019] [Indexed: 11/14/2022] Open
Abstract
DNA methylation has been widely studied for associations with exposures and health outcomes. Both 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) are epigenetic marks that may function differently to impact gene expression; however, the most commonly used technology to assess methylation for population studies in blood use are the Illumina 450K and EPIC BeadChips, for which the traditional bisulfite conversion does not differentiate 5mC and 5hmC marks. We used a modified protocol originally developed by Stewart et al. to analyse oxidative bisulfite-converted and conventional bisulfite-converted DNA for the same subject in parallel by the EPIC chip, allowing us to isolate the two measures. We measured 5mC and 5hmC in cord blood of 41 newborn participants of the Center for Health Assessment of Mothers and Children of Salinas (CHAMACOS) birth cohort and investigated differential methylation of 5mC + 5hmC, isolated 5mC and isolated 5hmC with sex at birth as an example of a biological variable previously associated with DNA methylation. Results showed low levels of 5hmC throughout the epigenome in the cord blood samples in comparison to 5mC. The concordance of autosomal hits between 5mC + 5hmC and exclusive 5mC analyses were low (25%); however, overlap was larger with increased effect size difference. There were 43 autosomal cytosine nucleotide followed by a guanine nucleotide (CpG) sites where 5hmC was associated with sex, 21 of which were unique to 5hmC after adjustment for cell composition. 5hmC only accounts for a small portion of overall methylation in cord blood; however, it has the potential to impact interpretation of combined 5hmC + 5mC studies in cord blood, especially given that effect sizes of differential methylation analyses are often small. Several significant CpG sites were unique to 5hmC, suggesting some functions distinct from 5mC. More studies of genome-wide 5hmC in children are warranted.
Collapse
Affiliation(s)
- Olivia Solomon
- School of Public Health, Center for Environmental Research and Children's Health (CERCH), University of California, Berkeley, Berkeley, CA, USA
| | - Julia L Macisaac
- University of British Columbia, BC Children's Hospital Research, Vancouver, BC, Canada
| | - Gwen Tindula
- School of Public Health, Center for Environmental Research and Children's Health (CERCH), University of California, Berkeley, Berkeley, CA, USA
| | - Michael S Kobor
- University of British Columbia, BC Children's Hospital Research, Vancouver, BC, Canada
| | - Brenda Eskenazi
- School of Public Health, Center for Environmental Research and Children's Health (CERCH), University of California, Berkeley, Berkeley, CA, USA
| | - Nina Holland
- School of Public Health, Center for Environmental Research and Children's Health (CERCH), University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
14
|
Heiss JA, Téllez-Rojo MM, Estrada-Gutiérrez G, Schnaas L, Amarasiriwardena C, Baccarelli AA, Wright RO, Just AC. Prenatal lead exposure and cord blood DNA methylation in PROGRESS: an epigenome-wide association study. ENVIRONMENTAL EPIGENETICS 2020; 6:dvaa014. [PMID: 33324494 PMCID: PMC7722799 DOI: 10.1093/eep/dvaa014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 05/03/2023]
Abstract
The effects of prenatal lead exposure on child development include impaired growth and cognitive function. DNA methylation might be involved in the underlying mechanisms and previous epigenome-wide association studies reported associations between lead exposure during pregnancy and cord blood methylation levels. However, it is unclear during which developmental stage lead exposure is most harmful. Cord blood methylation levels were assayed in 420 children from a Mexican pre-birth cohort using the Illumina Infinium MethylationEPIC microarray. Lead concentrations were measured in umbilical cord blood as well as in blood samples from the mothers collected at 2nd and 3rd trimester and delivery using inductively coupled plasma-mass spectrometry. In addition, maternal bone lead levels were measured in tibia and patella using X-ray fluorescence. Comprehensive quality control and preprocessing of microarray data was followed by an unbiased restriction to methylation sites with substantial variance. Methylation levels at 202 111 cytosine-phosphate-guanine sites were regressed on each exposure adjusting for child sex, leukocyte composition, batch variables, gestational age, birthweight-for-gestational-age, maternal age, maternal education and mode of delivery. We find no association between prenatal lead exposure and cord blood methylation. This null result is strengthened by a sensitivity analysis showing that in the same dataset known biomarkers for birthweight-for-gestational-age can be recovered and the fact that phenotypic associations with lead exposure have been described in the same cohort.
Collapse
Affiliation(s)
- Jonathan A Heiss
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1057, New York, NY 10029, USA
| | - Martha M Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, University No. 655 Colonia Santa María Ahuacatitlán, Closed Los Pinos and Caminera. Cuernavaca, Morelos, Mexico
| | | | - Lourdes Schnaas
- National Institute of Perinatology, Calle Montes Urales 800, Lomas de Virreyes, Mexico City, Mexico
| | - Chitra Amarasiriwardena
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1057, New York, NY 10029, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, 722 West 168th St., New York, NY, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1057, New York, NY 10029, USA
| | - Allan C Just
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1057, New York, NY 10029, USA
- Correspondence address. 17 East 102 Street Floor 3 Room 131 New York, NY 10029.
| |
Collapse
|
15
|
Timms JA, Relton CL, Sharp GC, Rankin J, Strathdee G, McKay JA. Exploring a potential mechanistic role of DNA methylation in the relationship between in utero and post-natal environmental exposures and risk of childhood acute lymphoblastic leukaemia. Int J Cancer 2019; 145:2933-2943. [PMID: 30740682 PMCID: PMC6790139 DOI: 10.1002/ijc.32203] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 01/09/2019] [Accepted: 01/23/2019] [Indexed: 12/11/2022]
Abstract
The aetiology of childhood acute lymphoblastic leukaemia (ALL) is unclear. Genetic abnormalities have been identified in a number of ALL cases, although these alone are not sufficient for leukaemic transformation. Various in utero and post-natal environmental exposures have been suggested to alter risk of childhood ALL. DNA methylation patterns can be influenced by environmental exposures, and are reported to be altered in ALL, suggesting a potential mediating mechanism between environment and ALL disease risk. To investigate this, we used a 'meet in the middle' approach, investigating the overlap between exposure-associated and disease-associated methylation change. Genome-wide DNA methylation changes in response to possible ALL-risk exposures (i.e. breast feeding, infection history, day care attendance, maternal smoking, alcohol, caffeine, folic acid, iron and radiation exposure) were investigated in a sub-population of the Avon Longitudinal Study of Parents and Children (ALSPAC) cohort using an epigenome-wide association study (EWAS) approach (n = 861-927), and compared to a list of ALL disease-associated methylation changes compiled from published data. Hypergeometric probability tests suggested that the number of directionally concordant gene methylation changes observed in ALL disease and in response to the following exposures; maternal radiation exposure (p = 0.001), alcohol intake (p = 0.006); sugary caffeinated drink intake during pregnancy (p = 0.045); and infant day care attendance (p = 0.003), were not due to chance. Data presented suggests that DNA methylation may be one mediating mechanism in the multiple hit pathway needed for ALL disease manifestation.
Collapse
Affiliation(s)
- Jessica A Timms
- Institute of Health & Society, Newcastle University, Newcastle, United Kingdom
- Research Oncology, King's College London, Guy's Hospital, London
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Gemma C Sharp
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Judith Rankin
- Research Oncology, King's College London, Guy's Hospital, London
| | - Gordon Strathdee
- Northern Institute for Cancer Research, Newcastle University, United Kingdom
| | - Jill A McKay
- Institute of Health & Society, Newcastle University, Newcastle, United Kingdom
- Department of Applied Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
16
|
Heiss JA, Brennan KJ, Baccarelli AA, Téllez-Rojo MM, Estrada-Gutiérrez G, Wright RO, Just AC. Battle of epigenetic proportions: comparing Illumina's EPIC methylation microarrays and TruSeq targeted bisulfite sequencing. Epigenetics 2019; 15:174-182. [PMID: 31538540 PMCID: PMC6961683 DOI: 10.1080/15592294.2019.1656159] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
DNA methylation microarrays have been the platform of choice for epigenome-wide association studies in epidemiology, but declining costs have rendered targeted bisulphite sequencing a feasible alternative. Nonetheless, the literature for researchers seeking guidance on which platform to choose is sparse. To fill this gap, we conducted a comparison study in which we processed cord blood samples from four newborns in duplicates using both the Illumina HumanMethylationEPIC BeadChip and the Illumina TruSeq Methyl Capture EPIC Kit, and evaluated both platforms in regard to coverage, reproducibility, and identification of differential methylation. We conclude that with current analytic goals microarrays still outperform bisulphite sequencing for precise quantification of DNA methylation.
Collapse
Affiliation(s)
- Jonathan A Heiss
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kasey J Brennan
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Martha M Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
| | | | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Allan C Just
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
17
|
Gervin K, Salas LA, Bakulski KM, van Zelm MC, Koestler DC, Wiencke JK, Duijts L, Moll HA, Kelsey KT, Kobor MS, Lyle R, Christensen BC, Felix JF, Jones MJ. Systematic evaluation and validation of reference and library selection methods for deconvolution of cord blood DNA methylation data. Clin Epigenetics 2019; 11:125. [PMID: 31455416 PMCID: PMC6712867 DOI: 10.1186/s13148-019-0717-y] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Umbilical cord blood (UCB) is commonly used in epigenome-wide association studies of prenatal exposures. Accounting for cell type composition is critical in such studies as it reduces confounding due to the cell specificity of DNA methylation (DNAm). In the absence of cell sorting information, statistical methods can be applied to deconvolve heterogeneous cell mixtures. Among these methods, reference-based approaches leverage age-appropriate cell-specific DNAm profiles to estimate cellular composition. In UCB, four reference datasets comprising DNAm signatures profiled in purified cell populations have been published using the Illumina 450 K and EPIC arrays. These datasets are biologically and technically different, and currently, there is no consensus on how to best apply them. Here, we systematically evaluate and compare these datasets and provide recommendations for reference-based UCB deconvolution. RESULTS We first evaluated the four reference datasets to ascertain both the purity of the samples and the potential cell cross-contamination. We filtered samples and combined datasets to obtain a joint UCB reference. We selected deconvolution libraries using two different approaches: automatic selection using the top differentially methylated probes from the function pickCompProbes in minfi and a standardized library selected using the IDOL (Identifying Optimal Libraries) iterative algorithm. We compared the performance of each reference separately and in combination, using the two approaches for reference library selection, and validated the results in an independent cohort (Generation R Study, n = 191) with matched Fluorescence-Activated Cell Sorting measured cell counts. Strict filtering and combination of the references significantly improved the accuracy and efficiency of cell type estimates. Ultimately, the IDOL library outperformed the library from the automatic selection method implemented in pickCompProbes. CONCLUSION These results have important implications for epigenetic studies in UCB as implementing this method will optimally reduce confounding due to cellular heterogeneity. This work provides guidelines for future reference-based UCB deconvolution and establishes a framework for combining reference datasets in other tissues.
Collapse
Affiliation(s)
- Kristina Gervin
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, School of Pharmacy, University of Oslo, Oslo, Norway
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, USA
| | - Kelly M Bakulski
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, Australia
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Devin C Koestler
- Department of Biostatistics, University of Kansas Medical Center, Kansas, KS, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Liesbeth Duijts
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Henriëtte A Moll
- Department of Pediatrics, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Karl T Kelsey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
- Department of Epidemiology, Brown University, Providence, RI, USA
| | - Michael S Kobor
- Department of Medical Genetics, University of British Columbia, and BC Children's Hospital Research Institute, Vancouver, Canada
| | - Robert Lyle
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, USA
- Department of Molecular and Systems Biology, and Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, USA
- Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, USA
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba, and Children's Hospital Research Institute of Manitoba, Winnipeg, Canada.
| |
Collapse
|
18
|
Massrali A, Brunel H, Hannon E, Wong C, Baron-Cohen S, Warrier V. Integrated genetic and methylomic analyses identify shared biology between autism and autistic traits. Mol Autism 2019; 10:31. [PMID: 31346403 PMCID: PMC6637466 DOI: 10.1186/s13229-019-0279-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/29/2019] [Indexed: 02/07/2023] Open
Abstract
Previous studies have identified differences in DNA methylation in autistic individuals compared to neurotypical individuals. Yet, it is unclear if this extends to autistic traits-subclinical manifestation of autism features in the general population. Here, we investigate the association between DNA methylation at birth (cord blood), and scores on the Social and Communication Disorders Checklist (SCDC), a measure of autistic traits, in 701 8-year-olds, by conducting a methylome-wide association study (MWAS). We did not identify significant CpGs associated with SCDC. The most significant CpG site was cg14379490, on chromosome 9 (MWAS beta = - 1.78 ± 0.35, p value = 5.34 × 10 -7 ). Using methylation data for autism in peripheral tissues, we did not identify a significant concordance in effect direction of CpGs with p value < 10-4 in the SCDC MWAS (binomial sign test, p value > 0.5). In contrast, using methylation data for autism from post-mortem brain tissues, we identify a significant concordance in effect direction of CpGs with a p value < 10-4 in the SCDC MWAS (binomial sign test, p value = 0.004). Supporting this, we observe an enrichment for genes that are dysregulated in the post-mortem autism brain (one-sided Wilcoxon rank-sum test, p value = 6.22 × 10-5). Finally, integrating genome-wide association study (GWAS) data for autism (n = 46,350) with mQTL maps from cord-blood (n = 771), we demonstrate that mQTLs of CpGs associated with SCDC scores at p value thresholds of 0.01 and 0.005 are significantly shifted toward lower p values in the GWAS for autism (p < 5 × 10-3). We provide additional support for this using a GWAS of SCDC, and demonstrate a lack of enrichment in a GWAS of Alzheimer's disease. Our results highlight the shared cross-tissue methylation architecture of autism and autistic traits, and demonstrate that mQTLs associated with differences in DNA methylation associated with childhood autistic traits are enriched for common genetic variants associated with autism and autistic traits.
Collapse
Affiliation(s)
- Aicha Massrali
- Autism Research Centre, University of Cambridge, Cambridge, UK
| | - Helena Brunel
- Autism Research Centre, University of Cambridge, Cambridge, UK
| | - Eilis Hannon
- University of Exeter Medical School, University of Exeter, RILD Building, Level 4, Barrack Rd, Exeter, UK
| | - Chloe Wong
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, London, UK
| | - iPSYCH-MINERvA Epigenetics Group
- Autism Research Centre, University of Cambridge, Cambridge, UK
- University of Exeter Medical School, University of Exeter, RILD Building, Level 4, Barrack Rd, Exeter, UK
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, London, UK
- Cambridgeshire and Peterborough National Health Service Foundation Trust, Cambridge, UK
| | - Simon Baron-Cohen
- Autism Research Centre, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough National Health Service Foundation Trust, Cambridge, UK
| | - Varun Warrier
- Autism Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
19
|
Matthews SG, McGowan PO. Developmental programming of the HPA axis and related behaviours: epigenetic mechanisms. J Endocrinol 2019; 242:T69-T79. [PMID: 30917340 DOI: 10.1530/joe-19-0057] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 12/22/2022]
Abstract
It has been approximately 30 years since the seminal discoveries of David Barker and his colleagues, and research is beginning to unravel the mechanisms that underlie developmental programming. The early environment of the embryo, foetus and newborn have been clearly linked to altered hypothalamic-pituitary-adrenal (HPA) function and related behaviours through the juvenile period and into adulthood. A number of recent studies have shown that these effects can pass across multiple generations. The HPA axis is highly responsive to the environment, impacts both central and peripheral systems and is critical to health in a wide variety of contexts. Mechanistic studies in animals are linking early exposures to adversity with changes in gene regulatory mechanisms, including modifications of DNA methylation and altered levels of miRNA. Similar associations are emerging from recent human studies. These findings suggest that epigenetic mechanisms represent a fundamental link between adverse early environments and developmental programming of later disease. The underlying biological mechanisms that connect the perinatal environment with modified long-term health outcomes represent an intensive area of research. Indeed, opportunities for early interventions must identify the relevant environmental factors and their molecular targets. This new knowledge will likely assist in the identification of individuals who are at risk of developing poor outcomes and for whom early intervention is most effective.
Collapse
Affiliation(s)
- Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Departments of Obstetrics & Gynaecology and Medicine, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Patrick O McGowan
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, University of Toronto, Scarborough, Ontario, Canada
- Department of Cell and Systems Biology, Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Bergens MA, Pittman GS, Thompson IJB, Campbell MR, Wang X, Hoyo C, Bell DA. Smoking-associated AHRR demethylation in cord blood DNA: impact of CD235a+ nucleated red blood cells. Clin Epigenetics 2019; 11:87. [PMID: 31182156 PMCID: PMC6558773 DOI: 10.1186/s13148-019-0686-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/24/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Numerous studies have demonstrated that DNA methylation levels in the aryl hydrocarbon receptor repressor (AHRR) gene measured in cord blood are significantly associated with prenatal tobacco smoke exposure and can be used as a fetal exposure biomarker. The mechanism driving this demethylation has not been determined and it is unclear if all cord blood cell types are impacted. Nucleated red blood cells (nRBCs/CD235a+ cells) are developmentally immature RBCs that display genome-wide hypomethylation and are observed at increased frequency in the cord blood of smoking mothers. We tested if AHRR methylation levels in CD235a+ nRBCs or nRBC counts influenced AHRR methylation in whole cord blood. METHODS Cord blood was collected from smoking (n = 34) and nonsmoking (n = 19) mothers and DNA was prepared from whole cord blood, isolated CD235a+ nRBCs, and CD14+ monocytes. AHRR methylation in cord blood DNA was measured using Illumina 850K arrays (cg05575921, chr5:373378). Pyrosequencing was used to compare methylation levels among cord blood, CD235a+, and CD14+ cells. We measured nRBC percentages using conventional complete blood counts and estimated percent nRBCs by a deconvolution model. RESULTS Methylation levels in AHRR were significantly lower in nRBCs relative to whole cord blood and CD14+ monocytes. While AHRR methylation levels in the cell types were significantly correlated across all subjects, methylation values at the chr5:373378 CpG averaged 14.6% lower in nRBCs (range 0.4 to 24.8%; p = 3.8E-13) relative to CD14+, with nonsmokers showing a significantly greater hypomethylation (- 4.1%, p = 1.8E-02). Methylation level at the AHRR chr5:373378 CpG was strongly associated with self-reported smoking in both CD14+ monocytes (t test p = 5.7E-09) and nRBCs (p = 4.8E-08), as well as cotinine levels (regression p = 1.1E-07 and p = 3.6E-04, respectively). For subjects with whole blood 850K data, robust linear regression models adjusting for estimated cell type composition, either including nRBCs counts or estimates, modestly increased the association between smoking and cg05575921 methylation. CONCLUSIONS Prenatal smoke exposure was highly significantly associated with AHRR methylation in cord blood, CD14+ monocytes, and CD235a+ nRBCs. AHRR methylation levels in nRBCs and nRBC counts had minimal effect on cord blood methylation measurements. However, regression models using estimated nRBCs or actual nRBC counts outperformed those lacking these covariates.
Collapse
Affiliation(s)
- Matthew A. Bergens
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709 USA
| | - Gary S. Pittman
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709 USA
| | - Isabel J. B. Thompson
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709 USA
| | - Michelle R. Campbell
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709 USA
| | - Xuting Wang
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709 USA
| | - Cathrine Hoyo
- Epidemiology and Environmental Epigenomics Laboratory, North Carolina State University, Raleigh, NC 27695 USA
| | - Douglas A. Bell
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709 USA
| |
Collapse
|
21
|
Heiss JA, Just AC. Improved filtering of DNA methylation microarray data by detection p values and its impact on downstream analyses. Clin Epigenetics 2019; 11:15. [PMID: 30678737 PMCID: PMC6346546 DOI: 10.1186/s13148-019-0615-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/11/2019] [Indexed: 12/21/2022] Open
Abstract
Background DNA methylation microarrays are popular for epigenome-wide association studies (EWAS), but spurious values complicate downstream analysis and threaten replication. Conventional cut-offs for detection p values for filtering out undetected probes were demonstrated in a single previous study as insufficient leading to many apparent methylation calls in samples from females in probes targeting the Y-chromosome. We present an alternative approach to calculate more accurate detection p values utilizing non-specific background fluorescence. We evaluate and compare our proposed approach of filtering observations with conventional ones by assessing the detection of Y-chromosome probes among males and females in 2755 samples from 17 studies on the 450K microarray and masking of large outliers between technical replicates and their impact downstream via an EWAS reanalysis. Results In contrast to conventional approaches, ours marks most Y-chromosome probes in females as undetected while removing a median of only 0.14% of the data per sample, catches more (30% vs. 6%) of large outliers (more than 20 percentage point difference between technical replicates), and helps to identify strong associations previously obfuscated by outliers between whole blood DNA methylation and chronological age in a well-powered EWAS (n = 729). Conclusions We provide guidance for filtering both 450K and EPIC microarrays as an essential preprocessing step to reduce spurious values. An implementation (including a function compatible with objects from the popular minfi package) was added to ewastools, an R package for comprehensive quality control of DNA methylation microarrays. Scripts to reproduce all analyses are available at doi.org/10.5281/zenodo.1443561.
Collapse
Affiliation(s)
- Jonathan A Heiss
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA.
| | - Allan C Just
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1057, New York, NY, 10029, USA
| |
Collapse
|
22
|
Min JL, Hemani G, Davey Smith G, Relton C, Suderman M. Meffil: efficient normalization and analysis of very large DNA methylation datasets. Bioinformatics 2018; 34:3983-3989. [PMID: 29931280 PMCID: PMC6247925 DOI: 10.1093/bioinformatics/bty476] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/18/2018] [Indexed: 12/11/2022] Open
Abstract
Motivation DNA methylation datasets are growing ever larger both in sample size and genome coverage. Novel computational solutions are required to efficiently handle these data. Results We have developed meffil, an R package designed for efficient quality control, normalization and epigenome-wide association studies of large samples of Illumina Methylation BeadChip microarrays. A complete re-implementation of functional normalization minimizes computational memory without increasing running time. Incorporating fixed and random effects within functional normalization, and automated estimation of functional normalization parameters reduces technical variation in DNA methylation levels, thus reducing false positive rates and improving power. Support for normalization of datasets distributed across physically different locations without needing to share biologically-based individual-level data means that meffil can be used to reduce heterogeneity in meta-analyses of epigenome-wide association studies. Availability and implementation https://github.com/perishky/meffil/. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- J L Min
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Bristol Medical School, University of Bristol, Bristol, UK
| | - G Hemani
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Bristol Medical School, University of Bristol, Bristol, UK
| | - G Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Bristol Medical School, University of Bristol, Bristol, UK
| | - C Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Bristol Medical School, University of Bristol, Bristol, UK
| | - M Suderman
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
23
|
An epigenetic pathway approach to investigating associations between prenatal exposure to maternal mood disorder and newborn neurobehavior. Dev Psychopathol 2018; 30:881-890. [PMID: 30068429 DOI: 10.1017/s0954579418000688] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Following recent advances in behavioral and psychiatric epigenetics, researchers are increasingly using epigenetic methods to study prenatal exposure to maternal mood disorder and its effects on fetal and newborn neurobehavior. Despite notable progress, various methodological limitations continue to obscure our understanding of the epigenetic mechanisms underpinning prenatal exposure to maternal mood disorder on newborn neurobehavioral development. Here we detail this problem, discussing limitations of the currently dominant analytical approaches (i.e., candidate epigenetic and epigenome-wide association studies), then present a solution that retains many benefits of existing methods while minimizing their shortcomings: epigenetic pathway analysis. We argue that the application of pathway-based epigenetic approaches that target DNA methylation at transcription factor binding sites could substantially deepen our mechanistic understanding of how prenatal exposures influence newborn neurobehavior.
Collapse
|
24
|
Lin X, Tan JYL, Teh AL, Lim IY, Liew SJ, MacIsaac JL, Chong YS, Gluckman PD, Kobor MS, Cheong CY, Karnani N. Cell type-specific DNA methylation in neonatal cord tissue and cord blood: a 850K-reference panel and comparison of cell types. Epigenetics 2018; 13:941-958. [PMID: 30232931 PMCID: PMC6284779 DOI: 10.1080/15592294.2018.1522929] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Accounting for cellular heterogeneity is essential in neonatal epigenome-wide association studies (EWAS) performed on heterogeneous tissues, such as umbilical cord tissue (CT) or cord blood (CB). Using a reference-panel-based statistical approach, the cell type composition of heterogeneous tissues can be estimated by comparison of whole tissue DNA methylation profiles with cell type-specific DNA methylation signatures. Currently, there is no adequate DNA methylation reference panel for CT, and existing CB panels have been generated on lower coverage Infinium HumanMethylation450 arrays. In this study, we generate a reference panel for CT and improve available CB panels by using the higher coverage Infinium MethylationEPIC arrays. We performed DNA methylation profiling of 9 cell types isolated from CT and CB samples from 14 neonates. In addition to these cell types, we profiled DNA methylation of unfractionated CT and CB. Cell type composition of these unfractionated tissue samples, as estimated by our reference panels, was in agreement with that obtained by flow cytometry. Expectedly, DNA methylation profiles from CT and CB were distinct, reflecting their mesenchymal and hematopoietic stem cell origins. Variable CpGs from both unfractionated CT and its isolated cell types were more likely to be located in open seas and intronic regions than those in CB. Cell type specific CpGs in CT were enriched in intercellular matrix pathways, while those from CB were enriched in immune-related pathways. This study provides an open source reference panel for estimation and adjustment of cellular heterogeneity in CT and CB, and broadens the scope of tissue utilization assessed in future neonatal EWAS studies.
Collapse
Affiliation(s)
- Xinyi Lin
- a Singapore Institute for Clinical Sciences , A*STAR , Singapore.,b Duke-NUS Medical School , Singapore
| | - Jane Yi Lin Tan
- a Singapore Institute for Clinical Sciences , A*STAR , Singapore
| | - Ai Ling Teh
- a Singapore Institute for Clinical Sciences , A*STAR , Singapore
| | - Ives Yubin Lim
- a Singapore Institute for Clinical Sciences , A*STAR , Singapore
| | - Samantha J Liew
- a Singapore Institute for Clinical Sciences , A*STAR , Singapore
| | - Julia L MacIsaac
- c Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics , University of British Columbia , Vancouver , BC , Canada
| | - Yap Seng Chong
- a Singapore Institute for Clinical Sciences , A*STAR , Singapore.,d Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine , National University of Singapore , Singapore
| | - Peter D Gluckman
- a Singapore Institute for Clinical Sciences , A*STAR , Singapore.,e Centre for Human Evolution, Adaptation and Disease, Liggins Institute , University of Auckland , Auckland , New Zealand
| | - Michael S Kobor
- c Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics , University of British Columbia , Vancouver , BC , Canada
| | | | - Neerja Karnani
- a Singapore Institute for Clinical Sciences , A*STAR , Singapore.,f Department of Biochemistry, Yong Loo Lin School of Medicine , National University of Singapore , Singapore
| |
Collapse
|
25
|
Population DNA methylation studies in the Developmental Origins of Health and Disease (DOHaD) framework. J Dev Orig Health Dis 2018; 10:306-313. [PMID: 30101736 DOI: 10.1017/s2040174418000442] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Epigenetic changes represent a potential mechanism underlying associations of early-life exposures and later life health outcomes. Population-based cohort studies starting in early life are an attractive framework to study the role of such changes. DNA methylation is the most studied epigenetic mechanism in population research. We discuss the application of DNA methylation in early-life population studies, some recent findings, key challenges and recommendations for future research. Studies into DNA methylation within the Developmental Origins of Health and Disease framework generally either explore associations between prenatal exposures and offspring DNA methylation or associations between offspring DNA methylation in early life and later health outcomes. Only a few studies to date have integrated prospective exposure, epigenetic and phenotypic data in order to explicitly test the role of DNA methylation as a potential biological mediator of environmental effects on health outcomes. Population epigenetics is an emerging field which has challenges in terms of methodology and interpretation of the data. Key challenges include tissue specificity, cell type adjustment, issues of power and comparability of findings, genetic influences, and exploring causality and functional consequences. Ongoing studies are working on addressing these issues. Large collaborative efforts of prospective cohorts are emerging, with clear benefits in terms of optimizing power and use of resources, and in advancing methodology. In the future, multidisciplinary approaches, within and beyond longitudinal birth and preconception cohorts will advance this complex, but highly promising, the field of research.
Collapse
|
26
|
Salas LA, Wiencke JK, Koestler DC, Zhang Z, Christensen BC, Kelsey KT. Tracing human stem cell lineage during development using DNA methylation. Genome Res 2018; 28:1285-1295. [PMID: 30072366 PMCID: PMC6120629 DOI: 10.1101/gr.233213.117] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 07/27/2018] [Indexed: 12/22/2022]
Abstract
Stem cell maturation is a fundamental, yet poorly understood aspect of human development. We devised a DNA methylation signature deeply reminiscent of embryonic stem cells (a fetal cell origin signature, FCO) to interrogate the evolving character of multiple human tissues. The cell fraction displaying this FCO signature was highly dependent upon developmental stage (fetal versus adult), and in leukocytes, it described a dynamic transition during the first 5 yr of life. Significant individual variation in the FCO signature of leukocytes was evident at birth, in childhood, and throughout adult life. The genes characterizing the signature included transcription factors and proteins intimately involved in embryonic development. We defined and applied a DNA methylation signature common among human fetal hematopoietic progenitor cells and have shown that this signature traces the lineage of cells and informs the study of stem cell heterogeneity in humans under homeostatic conditions.
Collapse
Affiliation(s)
- Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03756, USA
| | - John K Wiencke
- Department of Neurological Surgery, Institute for Human Genetics, University of California San Francisco, San Francisco, California 94158, USA
| | - Devin C Koestler
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Ze Zhang
- Department of Epidemiology, Brown University, Providence, Rhode Island 02912, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03756, USA.,Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03756, USA.,Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire 03756, USA
| | - Karl T Kelsey
- Department of Epidemiology, Brown University, Providence, Rhode Island 02912, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912, USA
| |
Collapse
|
27
|
Salas LA, Koestler DC, Butler RA, Hansen HM, Wiencke JK, Kelsey KT, Christensen BC. An optimized library for reference-based deconvolution of whole-blood biospecimens assayed using the Illumina HumanMethylationEPIC BeadArray. Genome Biol 2018; 19:64. [PMID: 29843789 PMCID: PMC5975716 DOI: 10.1186/s13059-018-1448-7] [Citation(s) in RCA: 248] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/08/2018] [Indexed: 11/23/2022] Open
Abstract
Genome-wide methylation arrays are powerful tools for assessing cell composition of complex mixtures. We compare three approaches to select reference libraries for deconvoluting neutrophil, monocyte, B-lymphocyte, natural killer, and CD4+ and CD8+ T-cell fractions based on blood-derived DNA methylation signatures assayed using the Illumina HumanMethylationEPIC array. The IDOL algorithm identifies a library of 450 CpGs, resulting in an average R2 = 99.2 across cell types when applied to EPIC methylation data collected on artificial mixtures constructed from the above cell types. Of the 450 CpGs, 69% are unique to EPIC. This library has the potential to reduce unintended technical differences across array platforms.
Collapse
Affiliation(s)
- Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Devin C Koestler
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Rondi A Butler
- Departments of Epidemiology and Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Helen M Hansen
- Department of Neurological Surgery, Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - John K Wiencke
- Department of Neurological Surgery, Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Karl T Kelsey
- Departments of Epidemiology and Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA.
- Departments of Molecular and Systems Biology, and Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA.
| |
Collapse
|
28
|
Titus AJ, Gallimore RM, Salas LA, Christensen BC. Cell-type deconvolution from DNA methylation: a review of recent applications. Hum Mol Genet 2018; 26:R216-R224. [PMID: 28977446 PMCID: PMC5886462 DOI: 10.1093/hmg/ddx275] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023] Open
Abstract
Recent advances in cell-type deconvolution approaches are adding to our understanding of the biology underlying disease development and progression. DNA methylation (DNAm) can be used as a biomarker of cell types, and through deconvolution approaches, to infer underlying cell type proportions. Cell-type deconvolution algorithms have two main categories: reference-based and reference-free. Reference-based algorithms are supervised methods that determine the underlying composition of cell types within a sample by leveraging differentially methylated regions (DMRs) specific to cell type, identified from DNAm measures of purified cell populations. Reference-free algorithms are unsupervised methods for use when cell-type specific DMRs are not available, allowing scientists to estimate putative cellular proportions or control for potential confounding from cell type. Reference-based deconvolution is typically applied to blood samples and has potentiated our understanding of the relation between immune profiles and disease by allowing estimation of immune cell proportions from archival DNA. Bioinformatic analyses using DNAm to infer immune cell proportions, part of a new field known as Immunomethylomics, provides a new direction for consideration in epigenome wide association studies (EWAS).
Collapse
Affiliation(s)
- Alexander J Titus
- Program in Quantitative Biomedical Sciences.,Department of Epidemiology
| | | | | | - Brock C Christensen
- Department of Epidemiology.,Department of Molecular and Systems Biology.,Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
29
|
Dou J, Schmidt RJ, Benke KS, Newschaffer C, Hertz-Picciotto I, Croen LA, Iosif AM, LaSalle JM, Fallin MD, Bakulski KM. Cord blood buffy coat DNA methylation is comparable to whole cord blood methylation. Epigenetics 2018; 13:108-116. [PMID: 29451060 PMCID: PMC5836975 DOI: 10.1080/15592294.2017.1417710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/29/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022] Open
Abstract
Cord blood DNA methylation is associated with numerous health outcomes and environmental exposures. Whole cord blood DNA reflects all nucleated blood cell types, while centrifuging whole blood separates red blood cells, generating a white blood cell buffy coat. Both sample types are used in DNA methylation studies. Cell types have unique methylation patterns and processing can impact cell distributions, which may influence comparability. We evaluated differences in cell composition and DNA methylation between cord blood buffy coat and whole cord blood samples. Cord blood DNA methylation was measured with the Infinium EPIC BeadChip (Illumina) in eight individuals, each contributing buffy coat and whole blood samples. We analyzed principal components (PC) of methylation, performed hierarchical clustering, and computed correlations of mean-centered methylation between pairs. We conducted moderated t-tests on single sites and estimated cell composition. DNA methylation PCs were associated with individual (PPC1 = 1.4 × 10-9; PPC2 = 2.9 × 10-5; PPC3 = 3.8 × 10-5; PPC4 = 4.2 × 10-6; PPC5 = 9.9 × 10-13, PPC6 = 1.3 × 10-11) and not with sample type (PPC1-6>0.7). Samples hierarchically clustered by individual. Pearson correlations of mean-centered methylation between paired samples ranged from r = 0.66 to r = 0.87. No individual site significantly differed between buffy coat and whole cord blood when adjusting for multiple comparisons (five sites had unadjusted P<10-5). Estimated cell type proportions did not differ by sample type (P = 0.46), and estimated proportions were highly correlated between paired samples (r = 0.99). Differences in methylation and cell composition between buffy coat and whole cord blood are much lower than inter-individual variation, demonstrating that both sample preparation types can be analytically combined and compared.
Collapse
Affiliation(s)
- John Dou
- Department of Epidemiology, School of Public Health, University of Michigan
| | - Rebecca J. Schmidt
- Department of Public Health Sciences, University of California Davis
- University of California Davis, MIND Institute
| | - Kelly S. Benke
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University
| | - Craig Newschaffer
- Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University
- A.J. Drexel Autism Institute, Drexel University
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, University of California Davis
- University of California Davis, MIND Institute
| | | | - Ana-Maria Iosif
- Department of Public Health Sciences, University of California Davis
- University of California Davis, MIND Institute
| | - Janine M. LaSalle
- University of California Davis, MIND Institute
- Department of Medical Microbiology and Immunology, Genome Center, University of California Davis
| | - M. Daniele Fallin
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins University
| | - Kelly M. Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan
| |
Collapse
|
30
|
DNA methylation at enhancers identifies distinct breast cancer lineages. Nat Commun 2017; 8:1379. [PMID: 29123100 PMCID: PMC5680222 DOI: 10.1038/s41467-017-00510-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 06/27/2017] [Indexed: 02/06/2023] Open
Abstract
Breast cancers exhibit genome-wide aberrant DNA methylation patterns. To investigate how these affect the transcriptome and which changes are linked to transformation or progression, we apply genome-wide expression-methylation quantitative trait loci (emQTL) analysis between DNA methylation and gene expression. On a whole genome scale, in cis and in trans, DNA methylation and gene expression have remarkably and reproducibly conserved patterns of association in three breast cancer cohorts (n = 104, n = 253 and n = 277). The expression-methylation quantitative trait loci associations form two main clusters; one relates to tumor infiltrating immune cell signatures and the other to estrogen receptor signaling. In the estrogen related cluster, using ChromHMM segmentation and transcription factor chromatin immunoprecipitation sequencing data, we identify transcriptional networks regulated in a cell lineage-specific manner by DNA methylation at enhancers. These networks are strongly dominated by ERα, FOXA1 or GATA3 and their targets were functionally validated using knockdown by small interfering RNA or GRO-seq analysis after transcriptional stimulation with estrogen.
Collapse
|
31
|
Sharp GC, Salas LA, Monnereau C, Allard C, Yousefi P, Everson TM, Bohlin J, Xu Z, Huang RC, Reese SE, Xu CJ, Baïz N, Hoyo C, Agha G, Roy R, Holloway JW, Ghantous A, Merid SK, Bakulski KM, Küpers LK, Zhang H, Richmond RC, Page CM, Duijts L, Lie RT, Melton PE, Vonk JM, Nohr EA, Williams-DeVane C, Huen K, Rifas-Shiman SL, Ruiz-Arenas C, Gonseth S, Rezwan FI, Herceg Z, Ekström S, Croen L, Falahi F, Perron P, Karagas MR, Quraishi BM, Suderman M, Magnus MC, Jaddoe VWV, Taylor JA, Anderson D, Zhao S, Smit HA, Josey MJ, Bradman A, Baccarelli AA, Bustamante M, Håberg SE, Pershagen G, Hertz-Picciotto I, Newschaffer C, Corpeleijn E, Bouchard L, Lawlor DA, Maguire RL, Barcellos LF, Davey Smith G, Eskenazi B, Karmaus W, Marsit CJ, Hivert MF, Snieder H, Fallin MD, Melén E, Munthe-Kaas MC, Arshad H, Wiemels JL, Annesi-Maesano I, Vrijheid M, Oken E, Holland N, Murphy SK, Sørensen TIA, Koppelman GH, Newnham JP, Wilcox AJ, Nystad W, London SJ, Felix JF, Relton CL. Maternal BMI at the start of pregnancy and offspring epigenome-wide DNA methylation: findings from the pregnancy and childhood epigenetics (PACE) consortium. Hum Mol Genet 2017; 26:4067-4085. [PMID: 29016858 PMCID: PMC5656174 DOI: 10.1093/hmg/ddx290] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/23/2017] [Accepted: 07/17/2017] [Indexed: 12/16/2022] Open
Abstract
Pre-pregnancy maternal obesity is associated with adverse offspring outcomes at birth and later in life. Individual studies have shown that epigenetic modifications such as DNA methylation could contribute. Within the Pregnancy and Childhood Epigenetics (PACE) Consortium, we meta-analysed the association between pre-pregnancy maternal BMI and methylation at over 450,000 sites in newborn blood DNA, across 19 cohorts (9,340 mother-newborn pairs). We attempted to infer causality by comparing the effects of maternal versus paternal BMI and incorporating genetic variation. In four additional cohorts (1,817 mother-child pairs), we meta-analysed the association between maternal BMI at the start of pregnancy and blood methylation in adolescents. In newborns, maternal BMI was associated with small (<0.2% per BMI unit (1 kg/m2), P < 1.06 × 10-7) methylation variation at 9,044 sites throughout the genome. Adjustment for estimated cell proportions greatly attenuated the number of significant CpGs to 104, including 86 sites common to the unadjusted model. At 72/86 sites, the direction of the association was the same in newborns and adolescents, suggesting persistence of signals. However, we found evidence for acausal intrauterine effect of maternal BMI on newborn methylation at just 8/86 sites. In conclusion, this well-powered analysis identified robust associations between maternal adiposity and variations in newborn blood DNA methylation, but these small effects may be better explained by genetic or lifestyle factors than a causal intrauterine mechanism. This highlights the need for large-scale collaborative approaches and the application of causal inference techniques in epigenetic epidemiology.
Collapse
Affiliation(s)
- Gemma C Sharp
- MRC Integrative Epidemiology Unit
- School of Social and Community Medicine
- School of Oral and Dental Sciences, University of Bristol, Bristol, UK
| | - Lucas A Salas
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Claire Monnereau
- The Generation R Study Group
- Department of Epidemiology
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Catherine Allard
- Centre de Recherche du Centre Hospitalier, Université de Sherbrooke, QC, Canada
| | - Paul Yousefi
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California Berkeley
| | - Todd M Everson
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jon Bohlin
- Department of Infection Epidemiology and Modeling, Norwegian Institute of Public Health, Oslo, Norway
| | - Zongli Xu
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Rae-Chi Huang
- Telethon Kids Institute, University of Western Australia, Crawley, WA 6009, Australia
| | - Sarah E Reese
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Cheng-Jian Xu
- Department of Pulmonology, GRIAC Research Institute
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nour Baïz
- Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Sorbonne Université, UPMC Univ Paris 06, INSERM, Pierre Louis Institute of Epidemiology and Public Health, Saint-Antoine Medical School, Paris, France
| | - Cathrine Hoyo
- Department of Biological Sciences
- Center for Human Health and the Environment, North Carolina State University, NC, USA
| | - Golareh Agha
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Ritu Roy
- University of California San Francisco, CA, USA
- HDF Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Computational Biology Core
| | - John W Holloway
- Human Development & Health, Faculty of Medicine, University of Southampton, UK
| | - Akram Ghantous
- Epigenetics Group, International Agency for Research on Cancer, Lyon, France
| | - Simon K Merid
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kelly M Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, MI, USA
| | - Leanne K Küpers
- MRC Integrative Epidemiology Unit
- School of Social and Community Medicine
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics, and Environmental Health Sciences, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit
- School of Social and Community Medicine
| | - Christian M Page
- Department of Non-Communicable Disease, Norwegian Institute of Public Health, Oslo, Norway
| | - Liesbeth Duijts
- The Generation R Study Group
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rolv T Lie
- Department of Global Public Health and Primary Care, University of Bergen, Norway
- Medical Birth Registry of Norway, Norwegian Institute of Public Health, Bergen, Norway
| | - Phillip E Melton
- The Curtin UWA Centre for Genetic Origins of Health and Disease, Faculty of Health Sciences, Curtin University Health Sciences, Curtin University and Faculty of Medicine Dentistry & Health Sciences, The University of Western Australia, Perth, Australia
- Faculty of Medicine Dentistry & Health Sciences, The University of Western Australia, Perth, Australia
| | - Judith M Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, GRIAC Research Institute Groningen, The Netherlands
| | - Ellen A Nohr
- Research Unit for Gynaecology and Obstetrics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | - Karen Huen
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California Berkeley
| | - Sheryl L Rifas-Shiman
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, USA
| | - Carlos Ruiz-Arenas
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Semira Gonseth
- Department of Epidemiology and Biostatistics, University of California San Francisco, CA, USA
- School of Public Health, University of California Berkeley, CA, USA
| | - Faisal I Rezwan
- Human Development & Health, Faculty of Medicine, University of Southampton, UK
| | - Zdenko Herceg
- Epigenetics Group, International Agency for Research on Cancer, Lyon, France
| | - Sandra Ekström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lisa Croen
- Division of Research, Kaiser Permanente Northern California, CA, UDA
| | - Fahimeh Falahi
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Patrice Perron
- Centre de Recherche du Centre Hospitalier, Université de Sherbrooke, QC, Canada
- Department of Medicine, Université de Sherbrooke, QC, Canada
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Children's Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, USA
| | - Bilal M Quraishi
- Division of Epidemiology, Biostatistics, and Environmental Health Sciences, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Matthew Suderman
- MRC Integrative Epidemiology Unit
- School of Social and Community Medicine
| | - Maria C Magnus
- MRC Integrative Epidemiology Unit
- School of Social and Community Medicine
- Department of Non-Communicable Disease, Norwegian Institute of Public Health, Oslo, Norway
| | - Vincent W V Jaddoe
- The Generation R Study Group
- Department of Epidemiology
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jack A Taylor
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Denise Anderson
- Telethon Kids Institute, University of Western Australia, Crawley, WA 6009, Australia
| | - Shanshan Zhao
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Henriette A Smit
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Michele J Josey
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA
- Epidemiology and Biostatistics Department, University of South Carolina (Columbia), SC, USA
| | - Asa Bradman
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California Berkeley
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Mariona Bustamante
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Siri E Håberg
- Domain of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
| | - Irva Hertz-Picciotto
- Department of Public Health, School of Medicine, University of California, Davis, CA, USA
| | - Craig Newschaffer
- AJ Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | - Eva Corpeleijn
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Luigi Bouchard
- Department of Biochemistry, Université de Sherbrooke, QC, Canada
- ECOGENE-21 and Lipid Clinic, Chicoutimi Hospital, Saguenay, QC, Canada
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit
- School of Social and Community Medicine
| | - Rachel L Maguire
- Department of Biological Sciences
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - Lisa F Barcellos
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California Berkeley
| | - George Davey Smith
- MRC Integrative Epidemiology Unit
- School of Social and Community Medicine
| | - Brenda Eskenazi
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California Berkeley
| | - Wilfried Karmaus
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Carmen J Marsit
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Marie-France Hivert
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, USA
- Department of Medicine, Université de Sherbrooke, QC, Canada
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M Daniele Fallin
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
- Sachs’ Children’s Hospital, South General Hospital, Stockholm, Sweden
| | - Monica C Munthe-Kaas
- Department of Pediatric and Adolescent Medicine, Oslo University Hospital, Norway
- Norwegian Institute of Public Health, Oslo Norway
| | - Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
- The David Hide Asthma and Allergy Research Centre, Isle of Wight, UK
| | - Joseph L Wiemels
- Department of Epidemiology and Biostatistics, University of California San Francisco, CA, USA
| | - Isabella Annesi-Maesano
- Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Sorbonne Université, UPMC Univ Paris 06, INSERM, Pierre Louis Institute of Epidemiology and Public Health, Saint-Antoine Medical School, Paris, France
| | - Martine Vrijheid
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Emily Oken
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, USA
| | - Nina Holland
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California Berkeley
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Thorkild I A Sørensen
- MRC Integrative Epidemiology Unit
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section on Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Epidemiology, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
| | - Gerard H Koppelman
- Department of Paediatric Pulmonology and Paediatric Allergy, University of Groningen, University Medical Center Groningen, Beatrix Children’s Hospital, GRIAC Research Institute, Groningen, the Netherlands
| | - John P Newnham
- School of Women's and Infants' Health, The University of Western Australia, Crawley, WA 6009, Australia
| | - Allen J Wilcox
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Wenche Nystad
- Department of Non-Communicable Disease, Norwegian Institute of Public Health, Oslo, Norway
| | - Stephanie J London
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Janine F Felix
- The Generation R Study Group
- Department of Epidemiology
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit
- School of Social and Community Medicine
| |
Collapse
|
32
|
Abstract
In mammals, caloric restriction consistently results in extended lifespan. Epigenetic information encoded by DNA methylation is tightly regulated, but shows a striking drift associated with age that includes both gains and losses of DNA methylation at various sites. Here, we report that epigenetic drift is conserved across species and the rate of drift correlates with lifespan when comparing mice, rhesus monkeys, and humans. Twenty-two to 30-year-old rhesus monkeys exposed to 30% caloric restriction since 7–14 years of age showed attenuation of age-related methylation drift compared to ad libitum-fed controls such that their blood methylation age appeared 7 years younger than their chronologic age. Even more pronounced effects were seen in 2.7–3.2-year-old mice exposed to 40% caloric restriction starting at 0.3 years of age. The effects of caloric restriction on DNA methylation were detectable across different tissues and correlated with gene expression. We propose that epigenetic drift is a determinant of lifespan in mammals. Caloric restriction has been shown to increase lifespan in mammals. Here, the authors provide evidence that age-related methylation drift correlates with lifespan and that caloric restriction in mice and rhesus monkeys results in attenuation of age-related methylation drift.
Collapse
|
33
|
Stephanou C, Papasavva P, Zachariou M, Patsali P, Epitropou M, Ladas P, Al-Abdulla R, Christou S, Antoniou MN, Lederer CW, Kleanthous M. Suitability of small diagnostic peripheral-blood samples for cell-therapy studies. Cytotherapy 2017; 19:311-326. [PMID: 28088294 DOI: 10.1016/j.jcyt.2016.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/07/2016] [Accepted: 11/07/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND AIMS Primary hematopoietic stem and progenitor cells (HSPCs) are key components of cell-based therapies for blood disorders and are thus the authentic substrate for related research. We propose that ubiquitous small-volume diagnostic samples represent a readily available and as yet untapped resource of primary patient-derived cells for cell- and gene-therapy studies. METHODS In the present study we compare isolation and storage methods for HSPCs from normal and thalassemic small-volume blood samples, considering genotype, density-gradient versus lysis-based cell isolation and cryostorage media with different serum contents. Downstream analyses include viability, recovery, differentiation in semi-solid media and performance in liquid cultures and viral transductions. RESULTS We demonstrate that HSPCs isolated either by ammonium-chloride potassium (ACK)-based lysis or by gradient isolation are suitable for functional analyses in clonogenic assays, high-level HSPC expansion and efficient lentiviral transduction. For cryostorage of cells, gradient isolation is superior to ACK lysis, and cryostorage in freezing media containing 50% fetal bovine serum demonstrated good results across all tested criteria. For assays on freshly isolated cells, ACK lysis performed similar to, and for thalassemic samples better than, gradient isolation, at a fraction of the cost and hands-on time. All isolation and storage methods show considerable variation within sample groups, but this is particularly acute for density gradient isolation of thalassemic samples. DISCUSSION This study demonstrates the suitability of small-volume blood samples for storage and preclinical studies, opening up the research field of HSPC and gene therapy to any blood diagnostic laboratory with corresponding bioethics approval for experimental use of surplus material.
Collapse
Affiliation(s)
- Coralea Stephanou
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus; Gene Expression and Therapy Group, King's College London, United Kingdom
| | - Panayiota Papasavva
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus; Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Myria Zachariou
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Petros Patsali
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus; Gene Expression and Therapy Group, King's College London, United Kingdom
| | - Marilena Epitropou
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Petros Ladas
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Ruba Al-Abdulla
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Soteroulla Christou
- Thalassaemia Centre, Ministry of Health, Archbishop Makarios III Hospital, Nicosia, Cyprus
| | - Michael N Antoniou
- Gene Expression and Therapy Group, King's College London, United Kingdom
| | - Carsten W Lederer
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus; Cyprus School of Molecular Medicine, Nicosia, Cyprus.
| | - Marina Kleanthous
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus; Cyprus School of Molecular Medicine, Nicosia, Cyprus
| |
Collapse
|
34
|
Morin AM, Gatev E, McEwen LM, MacIsaac JL, Lin DTS, Koen N, Czamara D, Räikkönen K, Zar HJ, Koenen K, Stein DJ, Kobor MS, Jones MJ. Maternal blood contamination of collected cord blood can be identified using DNA methylation at three CpGs. Clin Epigenetics 2017; 9:75. [PMID: 28770015 PMCID: PMC5526324 DOI: 10.1186/s13148-017-0370-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/11/2017] [Indexed: 01/10/2023] Open
Abstract
Background Cord blood is a commonly used tissue in environmental, genetic, and epigenetic population studies due to its ready availability and potential to inform on a sensitive period of human development. However, the introduction of maternal blood during labor or cross-contamination during sample collection may complicate downstream analyses. After discovering maternal contamination of cord blood in a cohort study of 150 neonates using Illumina 450K DNA methylation (DNAm) data, we used a combination of linear regression and random forest machine learning to create a DNAm-based screening method. We identified a panel of DNAm sites that could discriminate between contaminated and non-contaminated samples, then designed pyrosequencing assays to pre-screen DNA prior to being assayed on an array. Results Maternal contamination of cord blood was initially identified by unusual X chromosome DNA methylation patterns in 17 males. We utilized our DNAm panel to detect contaminated male samples and a proportional amount of female samples in the same cohort. We validated our DNAm screening method on an additional 189 sample cohort using both pyrosequencing and DNAm arrays, as well as 9 publically available cord blood 450K data sets. The rate of contamination varied from 0 to 10% within these studies, likely related to collection specific methods. Conclusions Maternal blood can contaminate cord blood during sample collection at appreciable levels across multiple studies. We have identified a panel of markers that can be used to identify this contamination, either post hoc after DNAm arrays have been completed, or in advance using a targeted technique like pyrosequencing. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0370-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexander M Morin
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital, Department of Medical Genetics, University of British Columbia, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
| | - Evan Gatev
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital, Department of Medical Genetics, University of British Columbia, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
| | - Lisa M McEwen
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital, Department of Medical Genetics, University of British Columbia, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
| | - Julia L MacIsaac
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital, Department of Medical Genetics, University of British Columbia, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
| | - David T S Lin
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital, Department of Medical Genetics, University of British Columbia, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
| | - Nastassja Koen
- Department of Psychiatry and Mental Health, South African Medical Research Council (SAMRC) Unit on Anxiety and Stress Disorders, University of Cape Town, Groote Schuur Hospital, J2, Anzio Road, Observatory, Cape Town, South Africa
| | - Darina Czamara
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Kraepelinstraße 2-10, 80804 Munich, Germany
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, P.O.Box 63, 00014 Helsinki, Finland
| | - Heather J Zar
- Department of Paediatrics, MRC Unit on Child and Adolescent Health, University of Cape Town, Room 513 ICH Building Red Cross Children's Hospital Klipfontein Road, Cape Town, South Africa
| | - Karestan Koenen
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Kresge Building, 505, Boston, MA 02115 USA
| | - Dan J Stein
- Department of Psychiatry and Mental Health, South African Medical Research Council (SAMRC) Unit on Anxiety and Stress Disorders, University of Cape Town, Groote Schuur Hospital, J2, Anzio Road, Observatory, Cape Town, South Africa
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital, Department of Medical Genetics, University of British Columbia, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada.,Human Early Learning Partnership, University of British Columbia, 2208 East Mall, Vancouver, BC 02115 Canada
| | - Meaghan J Jones
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital, Department of Medical Genetics, University of British Columbia, 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
| |
Collapse
|
35
|
Gervin K, Page CM, Aass HCD, Jansen MA, Fjeldstad HE, Andreassen BK, Duijts L, van Meurs JB, van Zelm MC, Jaddoe VW, Nordeng H, Knudsen GP, Magnus P, Nystad W, Staff AC, Felix JF, Lyle R. Cell type specific DNA methylation in cord blood: A 450K-reference data set and cell count-based validation of estimated cell type composition. Epigenetics 2017; 11:690-698. [PMID: 27494297 DOI: 10.1080/15592294.2016.1214782] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epigenome-wide association studies of prenatal exposure to different environmental factors are becoming increasingly common. These studies are usually performed in umbilical cord blood. Since blood comprises multiple cell types with specific DNA methylation patterns, confounding caused by cellular heterogeneity is a major concern. This can be adjusted for using reference data consisting of DNA methylation signatures in cell types isolated from blood. However, the most commonly used reference data set is based on blood samples from adult males and is not representative of the cell type composition in neonatal cord blood. The aim of this study was to generate a reference data set from cord blood to enable correct adjustment of the cell type composition in samples collected at birth. The purity of the isolated cell types was very high for all samples (>97.1%), and clustering analyses showed distinct grouping of the cell types according to hematopoietic lineage. We explored whether this cord blood and the adult peripheral blood reference data sets impact the estimation of cell type composition in cord blood samples from an independent birth cohort (MoBa, n = 1092). This revealed significant differences for all cell types. Importantly, comparison of the cell type estimates against matched cell counts both in the cord blood reference samples (n = 11) and in another independent birth cohort (Generation R, n = 195), demonstrated moderate to high correlation of the data. This is the first cord blood reference data set with a comprehensive examination of the downstream application of the data through validation of estimated cell types against matched cell counts.
Collapse
Affiliation(s)
- Kristina Gervin
- a Department of Medical Genetics , Oslo University Hospital , Oslo , Norway
| | - Christian Magnus Page
- b Division of Mental and Physical Health , Norwegian Institute of Public Health , Oslo , Norway
| | | | - Michelle A Jansen
- d The Generation R Study Group, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,e Department of Pediatrics, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,f Department of Immunology, Erasmus MC , University Medical Center Rotterdam , the Netherlands
| | | | | | - Liesbeth Duijts
- d The Generation R Study Group, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,i Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,j Department of Pediatrics, Division of Neonatology, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,k Department of Epidemiology, Erasmus MC , University Medical Center Rotterdam , the Netherlands
| | - Joyce B van Meurs
- l Department of Internal Medicine, Erasmus MC , University Medical Center Rotterdam , the Netherlands
| | - Menno C van Zelm
- m Department of Immunology, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,n Department of Immunology and Pathology, Central Clinical School , Monash University , Melbourne , Victoria , Australia
| | - Vincent W Jaddoe
- d The Generation R Study Group, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,e Department of Pediatrics, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,k Department of Epidemiology, Erasmus MC , University Medical Center Rotterdam , the Netherlands
| | - Hedvig Nordeng
- o Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, School of Pharmacy , University of Oslo , Norway.,p PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences , University of Oslo , Oslo , Norway
| | - Gunn Peggy Knudsen
- q Health Data and Digitalisation , Norwegian Institute of Public Health , Oslo , Norway
| | - Per Magnus
- q Health Data and Digitalisation , Norwegian Institute of Public Health , Oslo , Norway
| | - Wenche Nystad
- b Division of Mental and Physical Health , Norwegian Institute of Public Health , Oslo , Norway
| | - Anne Cathrine Staff
- g Departments of Obstetrics and Gynecology , Oslo University Hospital , Oslo , Norway.,r Faculty of Medicine , University of Oslo , Oslo , Norway
| | - Janine F Felix
- d The Generation R Study Group, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,e Department of Pediatrics, Erasmus MC , University Medical Center Rotterdam , the Netherlands.,k Department of Epidemiology, Erasmus MC , University Medical Center Rotterdam , the Netherlands
| | - Robert Lyle
- a Department of Medical Genetics , Oslo University Hospital , Oslo , Norway.,o Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, School of Pharmacy , University of Oslo , Norway.,p PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences , University of Oslo , Oslo , Norway
| |
Collapse
|
36
|
de Goede OM, Lavoie PM, Robinson WP. Cord blood hematopoietic cells from preterm infants display altered DNA methylation patterns. Clin Epigenetics 2017; 9:39. [PMID: 28428831 PMCID: PMC5397745 DOI: 10.1186/s13148-017-0339-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/08/2017] [Indexed: 12/18/2022] Open
Abstract
Background Premature infants are highly vulnerable to infection. This is partly attributable to the preterm immune system, which differs from that of the term neonate in cell composition and function. Multiple studies have found differential DNA methylation (DNAm) between preterm and term infants’ cord blood; however, interpretation of these studies is limited by the confounding factor of blood cell composition. This study evaluates the epigenetic impact of preterm birth in isolated hematopoietic cell populations, reducing the concern of cell composition differences. Methods Genome-wide DNAm was measured using the Illumina 450K array in T cells, monocytes, granulocytes, and nucleated red blood cells (nRBCs) isolated from cord blood of 5 term and 5 preterm (<31 weeks gestational age) newborns. DNAm of hematopoietic cells was compared globally across the 450K array and through site-specific linear modeling. Results Nucleated red blood cells (nRBCs) showed the most extensive changes in DNAm, with 9258 differentially methylated (DM) sites (FDR < 5%, |Δβ| > 0.10) discovered between preterm and term infants compared to the <1000 prematurity-DM sites identified in white blood cell populations. The direction of DNAm change with gestational age at these prematurity-DM sites followed known patterns of hematopoietic differentiation, suggesting that term hematopoietic cell populations are more epigenetically mature than their preterm counterparts. Consistent shifts in DNAm between preterm and term cells were observed at 25 CpG sites, with many of these sites located in genes involved in growth and proliferation, hematopoietic lineage commitment, and the cytoskeleton. DNAm in preterm and term hematopoietic cells conformed to previously identified DNAm signatures of fetal liver and bone marrow, respectively. Conclusions This study presents the first genome-wide mapping of epigenetic differences in hematopoietic cells across the late gestational period. DNAm differences in hematopoietic cells between term and <31 weeks were consistent with the hematopoietic origin of these cells during ontogeny, reflecting an important role of DNAm in their regulation. Due to the limited sample size and the high coincidence of prematurity and multiple births, the relationship between cause of preterm birth and DNAm could not be evaluated. These findings highlight gene regulatory mechanisms at both cell-specific and systemic levels that may be involved in fetal immune system maturation. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0339-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Olivia M de Goede
- BC Children's Hospital Research Institute, Room 2082, 950W 28th Avenue, Vancouver, BC V5Z 4H4 Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Pascal M Lavoie
- BC Children's Hospital Research Institute, Room 2082, 950W 28th Avenue, Vancouver, BC V5Z 4H4 Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Wendy P Robinson
- BC Children's Hospital Research Institute, Room 2082, 950W 28th Avenue, Vancouver, BC V5Z 4H4 Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| |
Collapse
|
37
|
Volberg V, Yousefi P, Huen K, Harley K, Eskenazi B, Holland N. CpG Methylation across the adipogenic PPARγ gene and its relationship with birthweight and child BMI at 9 years. BMC MEDICAL GENETICS 2017; 18:7. [PMID: 28122515 PMCID: PMC5267417 DOI: 10.1186/s12881-016-0365-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 12/26/2016] [Indexed: 01/05/2023]
Abstract
Background To examine methylation of the peroxisome proliferator-activated receptor γ (PPARγ) gene and its relationship with child weight status, at birth and 9 years. Methods We measured PPARγ methylation across 23 CpG sites using the Infinium Illumina 450 k array for children from the Center for the Health Assessment of Mothers and Children of Salinas (CHAMACOS) cohort at birth (N = 373) and 9 years (N = 245). Results Methylation level correlation patterns across the 23 PPARγ CpG sites were conserved between birth and 9-year ages. We found high inter-CpG correlations between sites 1–3 (methylation block 1) and also between sites 18–23 (methylation block 2) for both time points, although these patterns were less pronounced at 9 years. Additionally, sites 1–3 (north shore) had the highest intra-CpG correlations over time (r = 0.24, 0.42, and 0.3; P = 0.002, P < 0.001, P < 0.001, respectively). PPARγ methylation levels tended to increase with age, and the largest differences were observed for north shore sites (7.4%). Adjusting for sex, both site 1 and site 20 (gene body) methylation at birth was significantly and inversely associated with birth weight (β = −0.13, P = 0.033; β = −0.09, P = 0.025, respectively). Similarly, we found that site 1 and site 20 methylation at 9 years was significantly and inversely associated with 9-year BMI z-score (β = −0.41, P = 0.015; β = −0.23, P = 0.045, respectively). Conclusion Our results indicate that PPARγ methylation is highly organized and conserved over time, and highlight the potential functional importance of north shore sites, adding to a better understanding of regional human methylome patterns. Overall, our results suggest that PPARγ methylation may be associated with child body size. Electronic supplementary material The online version of this article (doi:10.1186/s12881-016-0365-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vitaly Volberg
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California, 733 University Hall, Berkeley, CA, 94720-7360, USA
| | - Paul Yousefi
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California, 733 University Hall, Berkeley, CA, 94720-7360, USA
| | - Karen Huen
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California, 733 University Hall, Berkeley, CA, 94720-7360, USA
| | - Kim Harley
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California, 733 University Hall, Berkeley, CA, 94720-7360, USA
| | - Brenda Eskenazi
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California, 733 University Hall, Berkeley, CA, 94720-7360, USA
| | - Nina Holland
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California, 733 University Hall, Berkeley, CA, 94720-7360, USA.
| |
Collapse
|
38
|
Schroeder DI, Schmidt RJ, Crary-Dooley FK, Walker CK, Ozonoff S, Tancredi DJ, Hertz-Picciotto I, LaSalle JM. Placental methylome analysis from a prospective autism study. Mol Autism 2016; 7:51. [PMID: 28018572 PMCID: PMC5159983 DOI: 10.1186/s13229-016-0114-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/28/2016] [Indexed: 11/10/2022] Open
Abstract
Background Autism spectrum disorders (ASD) are increasingly prevalent neurodevelopmental disorders that are behaviorally diagnosed in early childhood. Most ASD cases likely arise from a complex mixture of genetic and environmental factors, an interface where the epigenetic marks of DNA methylation may be useful as risk biomarkers. The placenta is a potentially useful surrogate tissue characterized by a methylation pattern of partially methylated domains (PMDs) and highly methylated domains (HMDs) reflective of methylation patterns observed in the early embryo. Methods In this study, we investigated human term placentas from the MARBLES (Markers of Autism Risk in Babies: Learning Early Signs) prospective study by whole genome bisulfite sequencing. We also examined the utility of PMD/HMDs in detecting methylation differences consistent with ASD diagnosis at age three. Results We found that while human placental methylomes have highly reproducible PMD and HMD locations, there is a greater variation between individuals in methylation levels over PMDs than HMDs due to both sampling and individual variability. In a comparison of methylation differences in placental samples from 24 ASD and 23 typically developing (TD) children, a HMD containing a putative fetal brain enhancer near DLL1 was found to reach genome-wide significance and was validated for significantly higher methylation in ASD by pyrosequencing. Conclusions These results suggest that the placenta could be an informative surrogate tissue for predictive ASD biomarkers in high-risk families. Electronic supplementary material The online version of this article (doi:10.1186/s13229-016-0114-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Diane I Schroeder
- Department of Medical Microbiology and Immunology, Genome Center, Davis, CA 95616 USA
| | - Rebecca J Schmidt
- Department of Public Health Sciences, University of California, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| | | | - Cheryl K Walker
- Department of Obstetrics and Gynecology, University of California, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| | - Sally Ozonoff
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| | - Daniel J Tancredi
- Department of Pediatrics, University of California, Davis, CA 95616 USA
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, University of California, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, Genome Center, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| |
Collapse
|
39
|
Agha G, Hajj H, Rifas-Shiman SL, Just AC, Hivert MF, Burris HH, Lin X, Litonjua AA, Oken E, DeMeo DL, Gillman MW, Baccarelli AA. Birth weight-for-gestational age is associated with DNA methylation at birth and in childhood. Clin Epigenetics 2016; 8:118. [PMID: 27891191 PMCID: PMC5112715 DOI: 10.1186/s13148-016-0285-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/02/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Both higher and lower fetal growth are associated with cardio-metabolic health later in life, suggesting that prenatal developmental programming determines long-term cardiovascular disease risk. Epigenetic mechanisms, which orchestrate fetal growth and development, may offer insight on the early programming of health and disease. We investigated whether birth weight-for-gestational is associated with DNA methylation at birth and mid-childhood, measured via the Infinium 450K array. METHODS/RESULTS Participants were from Project Viva, a pre-birth cohort of pregnant women and their children in Eastern Massachusetts. After exclusion of participants with maternal type 1 or 2 diabetes and gestational age <34 weeks, we used DNA methylation assays from 476 venous umbilical cord blood samples and a subset of 235 who additionally had peripheral blood samples available in mid-childhood (age 7-10 years). Among 392,918 CpG sites analyzed, birth weight-for-gestational age z-score was associated with cord blood DNA methylation at 34 CpGs (false discovery rate P < 0.05), after adjusting for maternal age, race/ethnicity, education, smoking, parity, delivery mode, pre-pregnancy BMI, gestational diabetes status, child sex, and estimated cord blood cell proportions based on a cord blood reference panel. Two of these CpGs were previously reported in epigenome-wide analyses of birth weight, and several other CpGs map to genes relevant to fetal growth and development. Namely, higher birth weight-for-gestational age was associated with higher methylation at four CpGs at the PBX1 locus (e.g., β (95% CI) for lead signal at cg06750897 = 1.9 (1.2, 2.6)), which encodes a transcription factor that regulates embryonic development. Birth weight-for-gestational age was also associated with mid-childhood blood DNA methylation at four of the 34 CpGs identified in cord blood analyses, including sites at the PBX1 locus described. CONCLUSIONS We identified CpG sites where birth weight-for-gestational age was associated with DNA methylation at birth, and for a subset of these sites, birth weight-for-gestational age was also associated with DNA methylation at mid-childhood.
Collapse
Affiliation(s)
- Golareh Agha
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, New York, NY 10032 USA
| | - Hanine Hajj
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA USA
| | - Sheryl L. Rifas-Shiman
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA USA
| | - Allan C. Just
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Marie-France Hivert
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA USA
| | - Heather H. Burris
- Department of Neonatology, Beth Israel Deaconess Medical Center, Department of Pediatrics, Harvard Medical School, Boston, MA USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | | | - Emily Oken
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA USA
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Matthew W. Gillman
- Obesity Prevention Program, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722 West 168th Street, New York, NY 10032 USA
| |
Collapse
|
40
|
Knight AK, Craig JM, Theda C, Bækvad-Hansen M, Bybjerg-Grauholm J, Hansen CS, Hollegaard MV, Hougaard DM, Mortensen PB, Weinsheimer SM, Werge TM, Brennan PA, Cubells JF, Newport DJ, Stowe ZN, Cheong JLY, Dalach P, Doyle LW, Loke YJ, Baccarelli AA, Just AC, Wright RO, Téllez-Rojo MM, Svensson K, Trevisi L, Kennedy EM, Binder EB, Iurato S, Czamara D, Räikkönen K, Lahti JMT, Pesonen AK, Kajantie E, Villa PM, Laivuori H, Hämäläinen E, Park HJ, Bailey LB, Parets SE, Kilaru V, Menon R, Horvath S, Bush NR, LeWinn KZ, Tylavsky FA, Conneely KN, Smith AK. An epigenetic clock for gestational age at birth based on blood methylation data. Genome Biol 2016; 17:206. [PMID: 27717399 PMCID: PMC5054584 DOI: 10.1186/s13059-016-1068-z] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 09/20/2016] [Indexed: 12/18/2022] Open
Abstract
Background Gestational age is often used as a proxy for developmental maturity by clinicians and researchers alike. DNA methylation has previously been shown to be associated with age and has been used to accurately estimate chronological age in children and adults. In the current study, we examine whether DNA methylation in cord blood can be used to estimate gestational age at birth. Results We find that gestational age can be accurately estimated from DNA methylation of neonatal cord blood and blood spot samples. We calculate a DNA methylation gestational age using 148 CpG sites selected through elastic net regression in six training datasets. We evaluate predictive accuracy in nine testing datasets and find that the accuracy of the DNA methylation gestational age is consistent with that of gestational age estimates based on established methods, such as ultrasound. We also find that an increased DNA methylation gestational age relative to clinical gestational age is associated with birthweight independent of gestational age, sex, and ancestry. Conclusions DNA methylation can be used to accurately estimate gestational age at or near birth and may provide additional information relevant to developmental stage. Further studies of this predictor are warranted to determine its utility in clinical settings and for research purposes. When clinical estimates are available this measure may increase accuracy in the testing of hypotheses related to developmental age and other early life circumstances. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-1068-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna K Knight
- Genetics and Molecular Biology Program, Emory University, Atlanta, GA, USA
| | - Jeffrey M Craig
- Murdoch Childrens Research Institute and Department of Paediatrics, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Christiane Theda
- The Royal Women's Hospital, Murdoch Childrens Research Institute and University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Marie Bækvad-Hansen
- Section of Neonatal Genetics, Danish Centre for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark
| | - Jonas Bybjerg-Grauholm
- Section of Neonatal Genetics, Danish Centre for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark
| | - Christine S Hansen
- Section of Neonatal Genetics, Danish Centre for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark
| | - Mads V Hollegaard
- Section of Neonatal Genetics, Danish Centre for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark.,The Danish Neonatal Screening Biobank, Department for Congenital Disorders, Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark
| | - David M Hougaard
- Section of Neonatal Genetics, Danish Centre for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark.,The Danish Neonatal Screening Biobank, Department for Congenital Disorders, Statens Serum Institut, Artillerivej 5, DK-2300, Copenhagen S, Denmark
| | - Preben B Mortensen
- National Centre for Register-based Research, School of Business and Social Sciences, Aarhus University, Fuglesangs Allé 4, 8210, Aarhus V, Denmark
| | - Shantel M Weinsheimer
- Institute of Biological Psychiatry, Sct. Hans Mental Health Center, Copenhagen Mental Health Services, iPSYCH - The Lundbeck Foundation's Initiative for Integrative Psychiatric Research, Boserupvej, DK-4000, Roskilde, Denmark
| | - Thomas M Werge
- Institute of Biological Psychiatry, Sct. Hans Mental Health Center, Copenhagen Mental Health Services, iPSYCH - The Lundbeck Foundation's Initiative for Integrative Psychiatric Research, Boserupvej, DK-4000, Roskilde, Denmark
| | | | - Joseph F Cubells
- Genetics and Molecular Biology Program, Emory University, Atlanta, GA, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.,Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - D Jeffrey Newport
- Departments of Psychiatry & Behavioral Sciences and Obstetrics & Gynecology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Zachary N Stowe
- Departments of Psychiatry & Behavioral Sciences, Pediatrics, and Obstetrics & Gynecology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jeanie L Y Cheong
- Murdoch Childrens Research Institute and Department of Paediatrics, University of Melbourne, Parkville, Victoria, 3052, Australia.,The Royal Women's Hospital, Murdoch Childrens Research Institute and University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Philippa Dalach
- Murdoch Childrens Research Institute and Department of Paediatrics, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Lex W Doyle
- Murdoch Childrens Research Institute and Department of Paediatrics, University of Melbourne, Parkville, Victoria, 3052, Australia.,The Royal Women's Hospital, Murdoch Childrens Research Institute and University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Yuk J Loke
- Murdoch Childrens Research Institute and Department of Paediatrics, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Andrea A Baccarelli
- Laboratory of Environmental Precision Biosciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Allan C Just
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert O Wright
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mara M Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Katherine Svensson
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Letizia Trevisi
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Elisabeth B Binder
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA.,Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
| | - Stella Iurato
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max-Planck Institute of Psychiatry, Munich, Germany
| | - Katri Räikkönen
- Institute of Behavioral Sciences, University of Helsinki, 00014, Helsinki, Finland
| | - Jari M T Lahti
- Institute of Behavioral Sciences, University of Helsinki, 00014, Helsinki, Finland.,Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, Finland.,Folkhälsan Research Centre, Helsinki, Finland
| | - Anu-Katriina Pesonen
- Institute of Behavioral Sciences, University of Helsinki, 00014, Helsinki, Finland
| | - Eero Kajantie
- National Institute for Health and Welfare, Children's Hospital, Helsinki University Hospital, 00271, Helsinki, Finland.,University of Helsinki, 00029, Helsinki, Finland.,Department of Obstetrics and Gynecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Pia M Villa
- Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, 00014, Helsinki, Finland
| | - Hannele Laivuori
- Medical and Clinical Genetics, and Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, 00014, Helsinki, Finland.,Institute for Molecular Medicine Finland, University of Helsinki, 00014, Helsinki, Finland
| | - Esa Hämäläinen
- HUSLAB and Department of Clinical Chemistry, Helsinki University Central Hospital, 00014, Helsinki, Finland
| | - Hea Jin Park
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, US
| | - Lynn B Bailey
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, US
| | - Sasha E Parets
- Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Varun Kilaru
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, US
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, US
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine University of California Los Angeles, Los Angeles, CA, 90095, US.,Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, 90095, US
| | - Nicole R Bush
- Department of Psychiatry, University of California, San Francisco, CA, US.,Department of Pediatrics, University of California, San Francisco, CA, US
| | - Kaja Z LeWinn
- Department of Psychiatry, University of California, San Francisco, CA, US
| | - Frances A Tylavsky
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, US
| | - Karen N Conneely
- Genetics and Molecular Biology Program, Emory University, Atlanta, GA, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Alicia K Smith
- Genetics and Molecular Biology Program, Emory University, Atlanta, GA, USA. .,Department of Psychiatry & Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA. .,Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, US.
| |
Collapse
|
41
|
de Goede OM, Lavoie PM, Robinson WP. Characterizing the hypomethylated DNA methylation profile of nucleated red blood cells from cord blood. Epigenomics 2016; 8:1481-1494. [PMID: 27687885 DOI: 10.2217/epi-2016-0069] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM To provide insight into fetal nucleated red blood cell (nRBC) development using genome-wide DNA methylation (DNAm) profiling. MATERIALS & METHODS The DNAm profile (Illumina 450K array) of cord blood (n = 7) derived nRBCs was compared with B cells, CD4 and CD8 T cells, natural killer cells, granulocytes, monocytes and placenta (n = 5). RESULTS nRBCs and placenta had similarly low array-wide DNAm compared with white blood cells, but their patterns of hypomethylation differed at biologically relevant subsets of the array. High interindividual variability in nRBC DNAm was driven by a negative association between DNAm and nRBC count. CONCLUSION nRBC hypomethylation is likely an epigenetic signature of erythropoiesis rather than of early development. Variability in nRBC DNAm may stem from differences in the cell population's maturity or hematopoietic source.
Collapse
Affiliation(s)
- Olivia M de Goede
- Child & Family Research Institute, Vancouver, British Columbia, V5Z 4H4, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Pascal M Lavoie
- Child & Family Research Institute, Vancouver, British Columbia, V5Z 4H4, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Wendy P Robinson
- Child & Family Research Institute, Vancouver, British Columbia, V5Z 4H4, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| |
Collapse
|
42
|
Cardenas A, Allard C, Doyon M, Houseman EA, Bakulski KM, Perron P, Bouchard L, Hivert MF. Validation of a DNA methylation reference panel for the estimation of nucleated cells types in cord blood. Epigenetics 2016; 11:773-779. [PMID: 27668573 PMCID: PMC5221601 DOI: 10.1080/15592294.2016.1233091] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Cord blood is widely used as surrogate tissue in epigenome-wide association studies of prenatal conditions. Cell type composition variation across samples can be an important confounder of epigenome-wide association studies in blood that constitute a mixture of cells. We evaluated a newly developed cord blood reference panel to impute cell type composition from DNA methylation levels, including nucleated red blood cells (nRBCs). We estimated cell type composition from 154 unique cord blood samples with available DNA methylation data as well as direct measurements of nucleated cell types. We observed high correlations between the estimated and measured composition for nRBCs (r = 0.92, R2 = 0.85), lymphocytes (r = 0.77, R2 = 0.58), and granulocytes (r = 0.72, R2 = 0.52), and a moderate correlation for monocytes (r = 0.51, R2 = 0.25) as well as relatively low root mean square errors from the residuals ranging from 1.4 to 5.4%. These results validate the use of the cord blood reference panel and highlight its utility and limitations for epidemiological studies.
Collapse
Affiliation(s)
- Andres Cardenas
- a Department of Population Medicine , Harvard Pilgrim Health Care Institute, Harvard Medical School , Boston , MA , USA
| | - Catherine Allard
- b Centre de Recherche du Center Hospitalier Universitaire de Sherbrooke , Sherbrooke, Quebec , Canada
| | - Myriam Doyon
- b Centre de Recherche du Center Hospitalier Universitaire de Sherbrooke , Sherbrooke, Quebec , Canada
| | - E Andres Houseman
- c College of Public Health and Human Sciences, Oregon State University , Corvallis , OR , USA
| | - Kelly M Bakulski
- d Department of Epidemiology , University of Michigan School of Public Health , Ann Arbor , MI , USA
| | - Patrice Perron
- b Centre de Recherche du Center Hospitalier Universitaire de Sherbrooke , Sherbrooke, Quebec , Canada.,e Department of Medicine , Faculty of Medicine and Life Sciences, Université de Sherbrooke , Sherbrooke, Quebec , Canada
| | - Luigi Bouchard
- b Centre de Recherche du Center Hospitalier Universitaire de Sherbrooke , Sherbrooke, Quebec , Canada.,f Department of Biochemistry , Faculty of medicine and life sciences, Université de Sherbrooke , Sherbrooke, Quebec , Canada.,g ECOGENE-21 and Lipid Clinic, Chicoutimi Hospital , Saguenay , Canada
| | - Marie-France Hivert
- a Department of Population Medicine , Harvard Pilgrim Health Care Institute, Harvard Medical School , Boston , MA , USA.,b Centre de Recherche du Center Hospitalier Universitaire de Sherbrooke , Sherbrooke, Quebec , Canada.,e Department of Medicine , Faculty of Medicine and Life Sciences, Université de Sherbrooke , Sherbrooke, Quebec , Canada.,h Massachusetts General Hospital , Boston , MA , USA
| |
Collapse
|
43
|
Kan B, Razzaghian HR, Lavoie PM. An Immunological Perspective on Neonatal Sepsis. Trends Mol Med 2016; 22:290-302. [PMID: 26993220 PMCID: PMC5104533 DOI: 10.1016/j.molmed.2016.02.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/08/2016] [Indexed: 02/07/2023]
Abstract
Despite concerted international efforts, mortality from neonatal infections remains unacceptably high in some areas of the world, particularly for premature infants. Recent developments in flow cytometry and next-generation sequencing technologies have led to major discoveries over the past few years, providing a more integrated understanding of the developing human immune system in the context of its microbial environment. We review these recent findings, focusing on how in human newborns incomplete maturation of the immune system before a full term of gestation impacts on their vulnerability to infection. We also discuss some of the clinical implications of this research in guiding the design of more-accurate age-adapted diagnostic and preventive strategies for neonatal sepsis.
Collapse
Affiliation(s)
- Bernard Kan
- Child and Family Research Institute, Vancouver, British Columbia, Canada; Experimental Medicine Program, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Hamid Reza Razzaghian
- Child and Family Research Institute, Vancouver, British Columbia, Canada; Experimental Medicine Program, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Pascal M Lavoie
- Child and Family Research Institute, Vancouver, British Columbia, Canada; Experimental Medicine Program, Department of Medicine, University of British Columbia, Vancouver, Canada; Department of Pediatrics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
44
|
Bakulski KM, Feinberg JI, Andrews SV, Yang J, Brown S, L McKenney S, Witter F, Walston J, Feinberg AP, Fallin MD. DNA methylation of cord blood cell types: Applications for mixed cell birth studies. Epigenetics 2016; 11:354-62. [PMID: 27019159 DOI: 10.1080/15592294.2016.1161875] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Epigenome-wide association studies of disease widely use DNA methylation measured in blood as a surrogate tissue. Cell proportions can vary between people and confound associations of exposure or outcome. An adequate reference panel for estimating cell proportions from adult whole blood for DNA methylation studies is available, but an analogous cord blood cell reference panel is not yet available. Cord blood has unique cell types and the epigenetic signatures of standard cell types may not be consistent throughout the life course. Using magnetic bead sorting, we isolated cord blood cell types (nucleated red blood cells, granulocytes, monocytes, natural killer cells, B cells, CD4(+)T cells, and CD8(+)T cells) from 17 live births at Johns Hopkins Hospital. We confirmed enrichment of the cell types using fluorescence assisted cell sorting and ran DNA from the separated cell types on the Illumina Infinium HumanMethylation450 BeadChip array. After filtering, the final analysis was on 104 samples at 429,794 probes. We compared cell type specific signatures in cord to each other and methylation at 49.2% of CpG sites on the array differed by cell type (F-test P < 10(-8)). Differences between nucleated red blood cells and the remainder of the cell types were most pronounced (36.9% of CpG sites at P < 10(-8)) and 99.5% of these sites were hypomethylated relative to the other cell types. We also compared the mean-centered sorted cord profiles to the available adult reference panel and observed high correlation between the overlapping cell types for granulocytes and monocytes (both r=0.74), and poor correlation for CD8(+)T cells and NK cells (both r=0.08). We further provide an algorithm for estimating cell proportions in cord blood using the newly developed cord reference panel, which estimates biologically plausible cell proportions in whole cord blood samples.
Collapse
Affiliation(s)
- Kelly M Bakulski
- a Department of Epidemiology , Johns Hopkins University Bloomberg School of Public Health , Baltimore , Maryland , USA.,b Center for Epigenetics, Johns Hopkins University School of Medicine , Baltimore , Maryland , USA.,c Department of Epidemiology , University of Michigan School of Public Health , Ann Arbor , Michigan , USA
| | - Jason I Feinberg
- b Center for Epigenetics, Johns Hopkins University School of Medicine , Baltimore , Maryland , USA.,d Department of Mental Health , Johns Hopkins University Bloomberg School of Public Health , Baltimore , Maryland , USA
| | - Shan V Andrews
- a Department of Epidemiology , Johns Hopkins University Bloomberg School of Public Health , Baltimore , Maryland , USA
| | - Jack Yang
- e Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Shannon Brown
- a Department of Epidemiology , Johns Hopkins University Bloomberg School of Public Health , Baltimore , Maryland , USA.,f Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health , Baltimore , Maryland , USA
| | - Stephanie L McKenney
- g Division of Neonatology, Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Frank Witter
- h Division of Gynecology and Obstetrics, Johns Hopkins University School of Medicine , Baltimore , Maryland , USA.,i Department of International Health , Johns Hopkins University Bloomberg School of Public Health , Baltimore , Maryland , USA
| | - Jeremy Walston
- e Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Andrew P Feinberg
- b Center for Epigenetics, Johns Hopkins University School of Medicine , Baltimore , Maryland , USA.,d Department of Mental Health , Johns Hopkins University Bloomberg School of Public Health , Baltimore , Maryland , USA
| | - M Daniele Fallin
- b Center for Epigenetics, Johns Hopkins University School of Medicine , Baltimore , Maryland , USA.,d Department of Mental Health , Johns Hopkins University Bloomberg School of Public Health , Baltimore , Maryland , USA.,f Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health , Baltimore , Maryland , USA
| |
Collapse
|