Informacja

Drogi użytkowniku, aplikacja do prawidłowego działania wymaga obsługi JavaScript. Proszę włącz obsługę JavaScript w Twojej przeglądarce.

Tytuł pozycji:

A cell-type deconvolution meta-analysis of whole blood EWAS reveals lineage-specific smoking-associated DNA methylation changes.

Tytuł:
A cell-type deconvolution meta-analysis of whole blood EWAS reveals lineage-specific smoking-associated DNA methylation changes.
Autorzy:
You C; CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
Wu S; CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.; Human Phenome Institute, Fudan University, 825 Zhangheng Road, Shanghai, China.; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.
Zheng SC; CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
Zhu T; CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
Jing H; CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
Flagg K; Guangzhou Regenerative Medicine Guangdong Laboratory, Guangzhou, China.
Wang G; Department of Computer Science and Technology, Tsinghua University, Beijing, China.
Jin L; CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.; Human Phenome Institute, Fudan University, 825 Zhangheng Road, Shanghai, China.; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.
Wang S; CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China. .; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China. .
Teschendorff AE; CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China. .; UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London, WC1E 6BT, UK. .
Źródło:
Nature communications [Nat Commun] 2020 Sep 22; Vol. 11 (1), pp. 4779. Date of Electronic Publication: 2020 Sep 22.
Typ publikacji:
Journal Article; Meta-Analysis; Research Support, Non-U.S. Gov't
Język:
English
Imprint Name(s):
Original Publication: [London] : Nature Pub. Group
MeSH Terms:
Epigenome*
DNA Methylation/*drug effects
Smoking/*adverse effects
Algorithms ; Asian People ; Blood ; CpG Islands ; Epigenomics/methods ; Ethnicity ; Female ; Humans ; Lymphocytes ; Male ; Middle Aged ; Models, Statistical ; Myeloid Cells
References:
Petronis, A. Epigenetics as a unifying principle in the aetiology of complex traits and diseases. Nature 465, 721–727 (2010). (PMID: 20535201)
Herceg, Z. et al. Roadmap for investigating epigenome deregulation and environmental origins of cancer. Int. J. Cancer 142, 874–882 (2018). (PMID: 28836271)
Christensen, B. C. et al. Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet. 5, e1000602 (2009). (PMID: 196804442718614)
Teschendorff, A. E. et al. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 20, 440–446 (2010). (PMID: 202199442847747)
Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 14, R115 (2013). (PMID: 241389284015143)
Hannum, G. et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol. Cell 49, 359–367 (2013).
Zeilinger, S. et al. Tobacco smoking leads to extensive genome-wide changes in DNA methylation. PloS ONE 8, e63812 (2013). (PMID: 236911013656907)
Gao, X., Jia, M., Zhang, Y., Breitling, L. P. & Brenner, H. DNA methylation changes of whole blood cells in response to active smoking exposure in adults: a systematic review of DNA methylation studies. Clin. Epigenet. 7, 113 (2015).
Teschendorff, A. E. et al. Correlation of smoking-associated DNA methylation changes in buccal cells with DNA methylation changes in epithelial cancer. JAMA Oncol. 1, 476–485 (2015). (PMID: 26181258)
Fasanelli, F. et al. Hypomethylation of smoking-related genes is associated with future lung cancer in four prospective cohorts. Nat. Commun. 6, 10192 (2015). (PMID: 266670484682166)
Baglietto, L. et al. DNA methylation changes measured in pre-diagnostic peripheral blood samples are associated with smoking and lung cancer risk. Int. J. Cancer 140, 50–61 (2017). (PMID: 27632354)
Zhang, Y. et al. Smoking-associated DNA methylation markers predict lung cancer incidence. Clin. Epigenet. 8, 127 (2016).
Zhang, Y. et al. Comparison and combination of blood DNA methylation at smoking-associated genes and at lung cancer-related genes in prediction of lung cancer mortality. International journal of cancer. Int. J. Cancer 139, 2482–2492 (2016). (PMID: 27503000)
Zhang, Y. et al. Smoking-associated DNA methylation biomarkers and their predictive value for all-cause and cardiovascular mortality. Environ. Health Perspect. 124, 67–74 (2016). (PMID: 26017925)
Lu, A. T. et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 11, 303–327 (2019). (PMID: 306691196366976)
Gao, X. et al. Tobacco smoking and smoking-related DNA methylation are associated with the development of frailty among older adults. Epigenetics 12, 149–156 (2017). (PMID: 28001461)
Battram, T. et al. Appraising the causal relevance of DNA methylation for risk of lung cancer. Int. J. Epidemiol. 48, 1493–1504 (2019). (PMID: 315491736857764)
Chen, Y., Widschwendter, M. & Teschendorff, A. E. Systems-epigenomics inference of transcription factor activity implicates aryl-hydrocarbon-receptor inactivation as a key event in lung cancer development. Genome Biol. 18, 236 (2017). (PMID: 292628475738803)
Lappalainen, T. & Greally, J. M. Associating cellular epigenetic models with human phenotypes. Nat. Rev. Genet. 18, 441–451 (2017). (PMID: 28555657)
Teschendorff, A. E. & Relton, C. L. Statistical and integrative system-level analysis of DNA methylation data. Nat. Rev. Genet. 19, 129–147 (2018). (PMID: 29129922)
Zhu, T., Zheng, S. C., Paul, D. S., Horvath, S. & Teschendorff, A. E. Cell and tissue type independent age-associated DNA methylation changes are not rare but common. Aging 10, 3541–3557 (2018). (PMID: 304828856286821)
Breitling, L. P., Yang, R., Korn, B., Burwinkel, B. & Brenner, H. Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am. J. Hum. Genet. 88, 450–457 (2011). (PMID: 214579053071918)
Shenker, N. S. et al. Epigenome-wide association study in the European Prospective Investigation into Cancer and Nutrition (EPIC-Turin) identifies novel genetic loci associated with smoking. Hum. Mol. Genet. 22, 843–851 (2013). (PMID: 23175441)
Sun, Y. V. et al. Epigenomic association analysis identifies smoking-related DNA methylation sites in African Americans. Hum. Genet. 132, 1027–1037 (2013). (PMID: 236575043744600)
Tsaprouni, L. G. et al. Cigarette smoking reduces DNA methylation levels at multiple genomic loci but the effect is partially reversible upon cessation. Epigenetics 9, 1382–1396 (2014). (PMID: 254246924623553)
Harlid, S., Xu, Z., Panduri, V., Sandler, D. P. & Taylor, J. A. CpG sites associated with cigarette smoking: analysis of epigenome-wide data from the Sister Study. Environ. Health Perspect. 122, 673–678 (2014). (PMID: 247045854080519)
Ambatipudi, S. et al. Tobacco smoking-associated genome-wide DNA methylation changes in the EPIC study. Epigenomics 8, 599–618 (2016). (PMID: 26864933)
Joehanes, R. et al. Epigenetic signatures of cigarette smoking. Circul. Cardiovasc. Genet. 9, 436–447 (2016).
Joubert, B. R. et al. DNA methylation in newborns and maternal smoking in pregnancy: genome-wide consortium meta-analysis. Am. J. Hum. Genet. 98, 680–696 (2016). (PMID: 270406904833289)
Monick, M. M. et al. Coordinated changes in AHRR methylation in lymphoblasts and pulmonary macrophages from smokers. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 159B, 141–151 (2012).
Philibert, R. A. et al. Coordinated DNA methylation and gene expression changes in smoker alveolar macrophages: specific effects on VEGF receptor 1 expression. J. Leukoc. Biol. 92, 621–631 (2012). (PMID: 224276823427615)
Ringh, M. V. et al. Tobacco smoking induces changes in true DNA methylation, hydroxymethylation and gene expression in bronchoalveolar lavage cells. EBioMedicine 46, 290–304 (2019). (PMID: 313034976710853)
Barrow, T. M. et al. Smoking is associated with hypermethylation of the APC 1A promoter in colorectal cancer: the ColoCare Study. J. Pathol. 243, 366–375 (2017). (PMID: 287917285647242)
Stueve, T. R. et al. Epigenome-wide analysis of DNA methylation in lung tissue shows concordance with blood studies and identifies tobacco smoke-inducible enhancers. Hum. Mol. Genet. 26, 3014–3027 (2017). (PMID: 288545645886283)
Zheng, S. C. et al. A novel cell-type deconvolution algorithm reveals substantial contamination by immune cells in saliva, buccal and cervix. Epigenomics 10, 925–940 (2018). (PMID: 29693419)
Vieira Braga, F. A. et al. A cellular census of human lungs identifies novel cell states in health and in asthma. Nat. Med. 25, 1153–1163 (2019). (PMID: 31209336)
Theda, C. et al. Quantitation of the cellular content of saliva and buccal swab samples. Sci. Rep. 8, 6944 (2018). (PMID: 297206145932057)
Eipel, M. et al. Epigenetic age predictions based on buccal swabs are more precise in combination with cell type-specific DNA methylation signatures. Aging 8, 1034–1048 (2016). (PMID: 272491024931852)
van Dongen, J. et al. Genome-wide analysis of DNA methylation in buccal cells: a study of monozygotic twins and mQTLs. Epigenet. Chromatin 11, 54 (2018).
Zheng, S. C., Breeze, C. E., Beck, S. & Teschendorff, A. E. Identification of differentially methylated cell types in epigenome-wide association studies. Nat. Methods 15, 1059–1066 (2018). (PMID: 305048706277016)
Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).
Rosenthal, R. et al. Neoantigen-directed immune escape in lung cancer evolution. Nature 567, 479–485 (2019). (PMID: 69541006954100)
Duncan, M. S. et al. Association of smoking cessation with subsequent risk of cardiovascular disease. J. Am. Med. Assoc. 322, 642–650 (2019).
Pan, S. et al. DNA methylome analysis reveals distinct epigenetic patterns of ascending aortic dissection and bicuspid aortic valve. Cardiovasc. Res. 113, 692–704 (2017). (PMID: 28444195)
Reynolds, L. M. et al. Tobacco exposure-related alterations in DNA methylation and gene expression in human monocytes: the Multi-Ethnic Study of Atherosclerosis (MESA). Epigenetics 12, 1092–1100 (2017). (PMID: 29166816)
Su, D. et al. Distinct epigenetic effects of tobacco smoking in whole blood and among leukocyte subtypes. PloS ONE 11, e0166486 (2016). (PMID: 279359725147832)
Bauer, M. et al. Tobacco smoking differently influences cell types of the innate and adaptive immune system-indications from CpG site methylation. Clin. Epigenet. 7, 83 (2015).
Li, Z., Wu, Z., Jin, P. & Wu, H. Dissecting differential signals in high-throughput data from complex tissues. Bioinformatics https://doi.org/10.1093/bioinformatics/btz196 (2019).
Luo, X., Yang, C. & Wei, Y. Detection of cell-type-specific risk-CpG sites in epigenome-wide association studies. Nat. Commun. 10, 3113 (2019). (PMID: 313083666629651)
Rahmani, E. et al. Cell-type-specific resolution epigenetics without the need for cell sorting or single-cell biology. Nat. Commun. 10, 3417 (2019). (PMID: 313669096668473)
Moran, S., Arribas, C. & Esteller, M. Validation of a DNA methylation microarray for 850,000 CpG sites of the human genome enriched in enhancer sequences. Epigenomics 8, 389–399 (2016).
Teschendorff, A. E. et al. DNA methylation outliers in normal breast tissue identify field defects that are enriched in cancer. Nat. Commun. 7, 10478 (2016). (PMID: 268230934740178)
Teschendorff, A. E. et al. A beta-mixture quantile normalization method for correcting probe design bias in Illumina Infinium 450 k DNA methylation data. Bioinformatics 29, 189–196 (2013). (PMID: 23175756)
Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).
Teschendorff, A. E. et al. An epigenetic signature in peripheral blood predicts active ovarian cancer. PloS ONE 4, e8274 (2009). (PMID: 200198732793425)
Teschendorff, A. E., Breeze, C. E., Zheng, S. C. & Beck, S. A comparison of reference-based algorithms for correcting cell-type heterogeneity in Epigenome-Wide Association Studies. BMC Bioinform. 18, 105 (2017).
Liu, Y. et al. Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in rheumatoid arthritis. Nat. Biotechnol. 31, 142–147 (2013). (PMID: 35986323598632)
Zhang, X. et al. Machine learning selected smoking-associated DNA methylation signatures that predict HIV prognosis and mortality. Clin. Epigenet. 10, 155 (2018).
Sikdar, S., Datta, S. & Datta, S. EAMA: Empirically adjusted meta-analysis for large-scale simultaneous hypothesis testing in genomic experiments. PloS ONE 12, e0187287 (2017). (PMID: 290882755663489)
Efron, B. Large-scale simultaneous hypothesis testing: the choice of null hypothesis. JASA 99, 96–104 (2004).
Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995).
Breeze, C. E. et al. eFORGE v2.0: updated analysis of cell type-specific signal in epigenomic data. Bioinformatics 35, 4767–4769 (2019). (PMID: 311612106853678)
Breeze, C. E. et al. eFORGE: a Tool for Identifying Cell Type-Specific Signal in Epigenomic Data. Cell Rep. 17, 2137–2150 (2016). (PMID: 278519745120369)
Adams, D. et al. BLUEPRINT to decode the epigenetic signature written in blood. Nat. Biotechnol. 30, 224–226 (2012). (PMID: 22398613)
Scott, C. L. et al. The transcription factor ZEB2 is required to maintain the tissue-specific identities of macrophages. Immunity 49, 312–325 e315 (2018). (PMID: 300761026104815)
Scott, C. L. & Omilusik, K. D. ZEBs: novel players in immune cell development and function. Trends Immunol. 40, 431–446 (2019). (PMID: 30956067)
Oliveira, L. M., Teixeira, F. M. E. & Sato, M. N. Impact of retinoic acid on immune cells and inflammatory diseases. Mediators Inflamm. 2018, 3067126 (2018). (PMID: 301588326109577)
Fircanis, S., Merriam, P., Khan, N. & Castillo, J. J. The relation between cigarette smoking and risk of acute myeloid leukemia: an updated meta-analysis of epidemiological studies. Am. J. Hematol. 89, E125–E132 (2014). (PMID: 24753145)
Kane, E. V., Roman, E., Cartwright, R., Parker, J. & Morgan, G. Tobacco and the risk of acute leukaemia in adults. Br. J. Cancer 81, 1228–1233 (1999). (PMID: 105848862374333)
Takai, H., Wang, R. C., Takai, K. K., Yang, H. & de Lange, T. Tel2 regulates the stability of PI3K-related protein kinases. Cell 131, 1248–1259 (2007). (PMID: 18160036)
Horejsi, Z. et al. CK2 phospho-dependent binding of R2TP complex to TEL2 is essential for mTOR and SMG1 stability. Mol. Cell 39, 839–850 (2010). (PMID: 20864032)
Hoshii, T. et al. mTORC1 is essential for leukemia propagation but not stem cell self-renewal. J. Clin. Investig. 122, 2114–2129 (2012). (PMID: 22622041)
Huan, T. et al. A whole-blood transcriptome meta-analysis identifies gene expression signatures of cigarette smoking. Hum. Mol. Genet. 25, 4611–4623 (2016). (PMID: 281585905975607)
Guida, F. et al. Dynamics of smoking-induced genome-wide methylation changes with time since smoking cessation. Hum. Mol. Genet. 24, 2349–2359 (2015). (PMID: 255561844380075)
Lee, K. W. & Pausova, Z. Cigarette smoking and DNA methylation. Front. Genet. 4, 132 (2013). (PMID: 238822783713237)
Opitz, C. A. et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 478, 197–203 (2011). (PMID: 21976023)
Beamer, C. A. & Shepherd, D. M. Role of the aryl hydrocarbon receptor (AhR) in lung inflammation. Semin. Immunopathol. 35, 693–704 (2013). (PMID: 23963493)
Vaz, M. et al. Chronic cigarette smoke-induced epigenomic changes precede sensitization of bronchial epithelial cells to single-step transformation by KRAS mutations. Cancer Cell 32, 360–376. e366 (2017). (PMID: 288986975596892)
Cheng, X. et al. Smoking affects gene expression in blood of patients with ischemic stroke. Ann. Clin. Transl. Neurol. 6, 1748–1756 (2019). (PMID: 314369166764500)
Merhi, M. et al. Impact of inflammation, gene variants, and cigarette smoking on coronary artery disease risk. Inflamm. Res. 64, 415–422 (2015). (PMID: 25902778)
Yao, Q. et al. Synergistic effect of ALOX5AP polymorphisms and cigarette smoking on the risk of atherosclerotic cerebral infarction in a Northern Han Chinese population. J. Clin. Neurosci. 21, 975–979 (2014). (PMID: 24411318)
Aryee, M. J. et al. Minfi: a flexible and comprehensive Bioconductor package for the analysis of Infinium DNA methylation microarrays. Bioinformatics 30, 1363–1369 (2014). (PMID: 244783394016708)
Troyanskaya, O. et al. Missing value estimation methods for DNA microarrays. Bioinformatics 17, 520–525 (2001). (PMID: 11395428)
Leek, J. T., Johnson, W. E., Parker, H. S., Jaffe, A. E. & Storey, J. D. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics https://doi.org/10.1093/bioinformatics/bts034 (2012).
Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl Acad. Sci. USA 100, 9440–9445 (2003).
Chen, Y. A. et al. Discovery of cross-reactive probes and polymorphic CpGs in the Illumina Infinium HumanMethylation450 microarray. Epigenetics 8, 203–209 (2013). (PMID: 233146983592906)
Reynolds, L. M. et al. Age-related variations in the methylome associated with gene expression in human monocytes and T cells. Nat. Commun. 5, 5366 (2014). (PMID: 254041684280798)
Zheng, S. C. et al. Correcting for cell-type heterogeneity in epigenome-wide association studies: revisiting previous analyses. Nat. Methods 14, 216–217 (2017). (PMID: 282452196659733)
Entry Date(s):
Date Created: 20200923 Date Completed: 20201013 Latest Revision: 20221207
Update Code:
20240105
PubMed Central ID:
PMC7508850
DOI:
10.1038/s41467-020-18618-y
PMID:
32963246
Czasopismo naukowe
Highly reproducible smoking-associated DNA methylation changes in whole blood have been reported by many Epigenome-Wide-Association Studies (EWAS). These epigenetic alterations could have important implications for understanding and predicting the risk of smoking-related diseases. To this end, it is important to establish if these DNA methylation changes happen in all blood cell subtypes or if they are cell-type specific. Here, we apply a cell-type deconvolution algorithm to identify cell-type specific DNA methylation signals in seven large EWAS. We find that most of the highly reproducible smoking-associated hypomethylation signatures are more prominent in the myeloid lineage. A meta-analysis further identifies a myeloid-specific smoking-associated hypermethylation signature enriched for DNase Hypersensitive Sites in acute myeloid leukemia. These results may guide the design of future smoking EWAS and have important implications for our understanding of how smoking affects immune-cell subtypes and how this may influence the risk of smoking related diseases.

Ta witryna wykorzystuje pliki cookies do przechowywania informacji na Twoim komputerze. Pliki cookies stosujemy w celu świadczenia usług na najwyższym poziomie, w tym w sposób dostosowany do indywidualnych potrzeb. Korzystanie z witryny bez zmiany ustawień dotyczących cookies oznacza, że będą one zamieszczane w Twoim komputerze. W każdym momencie możesz dokonać zmiany ustawień dotyczących cookies