Hostname: page-component-848d4c4894-x24gv Total loading time: 0 Render date: 2024-05-31T21:08:56.735Z Has data issue: false hasContentIssue false

Genome-wide DNA methylation profiling in nonagenarians suggests an effect of PM20D1 in late onset Alzheimer’s disease

Published online by Cambridge University Press:  16 December 2021

Carolina Coto-Vílchez
Affiliation:
Centro de Investigación en Biología Celular y Molecular, Universidad de Costa Rica, San José, Costa Rica
José J. Martínez-Magaña
Affiliation:
Instituto Nacional de Medicina Genómica, Mexico City, México
Lara Mora-Villalobos
Affiliation:
Centro de Investigación en Biología Celular y Molecular, Universidad de Costa Rica, San José, Costa Rica
Daniel Valerio
Affiliation:
Hospital Nacional de Geriatría y Gerontología de Costa Rica, San José, Costa Rica
Alma D. Genis-Mendoza
Affiliation:
Instituto Nacional de Medicina Genómica, Mexico City, México
Jeremy M. Silverman
Affiliation:
Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Humberto Nicolini
Affiliation:
Instituto Nacional de Medicina Genómica, Mexico City, México
Henriette Raventós
Affiliation:
Centro de Investigación en Biología Celular y Molecular, Universidad de Costa Rica, San José, Costa Rica Escuela de Biología, Universidad de Costa Rica, San José, Costa Rica
Gabriela Chavarria-Soley*
Affiliation:
Centro de Investigación en Biología Celular y Molecular, Universidad de Costa Rica, San José, Costa Rica Escuela de Biología, Universidad de Costa Rica, San José, Costa Rica
*
* Author for correspondence: Gabriela Chavarria-Soley, Email: gabriela.chavarriasoley@ucr.ac.cr

Abstract

Background

The aim of this study is to identify differentially methylated regions (DMRs) in the genomes of a sample of cognitively healthy individuals and a sample of individuals with LOAD, all of them nonagenarians from Costa Rica.

Methods

In this study, we compared whole blood DNA methylation profiles of 32 individuals: 21 cognitively healthy and 11 with LOAD, using the Infinium MethylationEPIC BeadChip. First, we calculated the epigenetic age of the participants based on Horvath’s epigenetic clock. DMRcate and Bumphunter were used to identify DMRs. After in silico and knowledge-based filtering of the DMRs, we performed a methylation quantitative loci (mQTL) analysis (rs708727 and rs960603).

Results

On average, the epigenetic age was 73 years in both groups, which represents a difference of over 20 years between epigenetic and chronological age in both affected and unaffected individuals. Methylation analysis revealed 11 DMRs between groups, which contain six genes and two pseudogenes. These genes are involved in cell cycle regulation, embryogenesis, synthesis of ceramides, and migration of interneurons to the cerebral cortex. One of the six genes is PM20D1, for which altered expression has been reported in LOAD. After genotyping previously reported mQTL SNPs for the gene, we found that average methylation in the PM20D1 DMR differs between genotypes for rs708727, but not for rs960603.

Conclusions

This work supports the possible role of PM20D1 in protection against AD, by showing differential methylation in blood of affected and unaffected nonagenarians. Our results also support the influence of genetic factors on PM20D1 methylation levels.

Type
Original Research
Copyright
© The Author(s), 2021. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Scheltens, P, Blennow, K, Breteler, MMB, et al. Alzheimer’s disease. Lancet Lond Engl. 2016;388(10043):505517. doi:10.1016/S0140-6736(15)01124-1.CrossRefGoogle ScholarPubMed
Farrer, LA, Cupples, LA, Haines, JL, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA. 1997;278(16):13491356.CrossRefGoogle ScholarPubMed
Reitz, C, Brayne, C, Mayeux, R. Epidemiology of Alzheimer disease. Nat Rev Neurol. 2011;7(3):137152. doi:10.1038/nrneurol.2011.2.CrossRefGoogle ScholarPubMed
Silva, MVF, Loures C de, MG, Alves, LCV, et al. Alzheimer’s disease: risk factors and potentially protective measures. J Biomed Sci. 2019;26(1):33. doi:10.1186/s12929-019-0524-y.CrossRefGoogle ScholarPubMed
Robinson, M, Lee, BY, Hane, FT. Recent progress in Alzheimer’s disease research, part 2: genetics and epidemiology. J Alzheimers Dis JAD. 2017;57(2):317330. doi:10.3233/JAD-161149.CrossRefGoogle ScholarPubMed
Cuyvers, E, Sleegers, K. Genetic variations underlying Alzheimer’s disease: evidence from genome-wide association studies and beyond. Lancet Neurol. 2016;15(8):857868. doi:10.1016/S1474-4422(16)00127-7.CrossRefGoogle ScholarPubMed
Andrews, SJ, Fulton-Howard, B, Goate, A. Interpretation of risk loci from genome-wide association studies of Alzheimer’s disease. Lancet Neurol. 2020;19(4):326335. doi:10.1016/S1474-4422(19)30435-1.CrossRefGoogle ScholarPubMed
Suri, S, Heise, V, Trachtenberg, AJ, et al. The forgotten APOE allele: a review of the evidence and suggested mechanisms for the protective effect of APOE ɛ2. Neurosci Biobehav Rev. 2013;37(10 Pt 2):28782886. doi:10.1016/j.neubiorev.2013.10.010.CrossRefGoogle ScholarPubMed
Vemuri, P, Knopman, DS, Lesnick, TG, et al. Evaluation of amyloid protective factors and Alzheimer disease neurodegeneration protective factors in elderly individuals. JAMA Neurol. 2017;74(6):718726. doi:10.1001/jamaneurol.2017.0244.CrossRefGoogle ScholarPubMed
Scacchi, R, De Bernardini, L, Mantuano, E, et al. Apolipoprotein E (APOE) allele frequencies in late-onset sporadic Alzheimer’s disease (AD), mixed dementia and vascular dementia: lack of association of epsilon 4 allele with AD in Italian octogenarian patients. Neurosci Lett. 1995;201(3):231234. doi:10.1016/0304-3940(95)12190-0.CrossRefGoogle ScholarPubMed
Corder, EH, Saunders, AM, Strittmatter, WJ, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261(5123):921923. doi:10.1126/science.8346443.CrossRefGoogle ScholarPubMed
Corder, EH, Saunders, AM, Risch, NJ, et al. Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease. Nat Genet. 1994;7(2):180184. doi:10.1038/ng0694-180.CrossRefGoogle ScholarPubMed
Valerio, D, Raventos, H, Schmeidler, J, et al. Association of apolipoprotein E-e4 and dementia declines with age. Am J Geriatr Psychiatry Off J Am Assoc Geriatr Psychiatry. 2014;22(10):957960. doi:10.1016/j.jagp.2014.03.008.CrossRefGoogle ScholarPubMed
Silverman, JM, Schmeidler, J. The protected survivor model: using resistant successful cognitive aging to identify protection in the very old. Med Hypotheses. 2018;110:914. doi:10.1016/j.mehy.2017.10.022.CrossRefGoogle ScholarPubMed
Sanchez-Mut, JV, Gräff, J. Epigenetic alterations in Alzheimer’s disease. Front Behav Neurosci. 2015;9:347. doi:10.3389/fnbeh.2015.00347.CrossRefGoogle ScholarPubMed
Zhang, L, Silva, TC, Young, JI, et al. Epigenome-wide meta-analysis of DNA methylation differences in prefrontal cortex implicates the immune processes in Alzheimer’s disease. Nat Commun. 2020;11(1):6114. doi:10.1038/s41467-020-19791-w.CrossRefGoogle ScholarPubMed
Liu, L, Lauro, BM, Ding, L, et al. Multiple BACE1 inhibitors abnormally increase the BACE1 protein level in neurons by prolonging its half-life. Alzheimers Dement J Alzheimers Assoc. 2019;15(9):11831194. doi:10.1016/j.jalz.2019.06.3918.CrossRefGoogle ScholarPubMed
Walton, E, Hass, J, Liu, J, et al. Correspondence of DNA methylation between blood and brain tissue and its application to schizophrenia research. Schizophr Bull. 2016;42(2):406414. doi:10.1093/schbul/sbv074.CrossRefGoogle ScholarPubMed
Edgar, RD, Jones, MJ, Meaney, MJ, et al. BECon: a tool for interpreting DNA methylation findings from blood in the context of brain. Transl Psychiatry. 2017;7(8):e1187. doi:10.1038/tp.2017.171.CrossRefGoogle ScholarPubMed
Braun, PR, Han, S, Hing, B, et al. Genome-wide DNA methylation comparison between live human brain and peripheral tissues within individuals. Transl Psychiatry. 2019;9(1):47. doi:10.1038/s41398-019-0376-y.CrossRefGoogle ScholarPubMed
Chen, J, Zang, Z, Braun, U, et al. Association of a reproducible epigenetic risk profile for schizophrenia with brain methylation and function. JAMA Psychiatry. 2020;77(6):628636. doi:10.1001/jamapsychiatry.2019.4792.CrossRefGoogle ScholarPubMed
Depp, CA, Jeste, DV. Definitions and predictors of successful aging: a comprehensive review of larger quantitative studies. Am J Geriatr Psychiatry Off J Am Assoc Geriatr Psychiatry. 2006;14(1):620. doi:10.1097/01.JGP.0000192501.03069.bc.CrossRefGoogle ScholarPubMed
Silverman, JM, Schnaider-Beeri, M, Grossman, HT, et al. A phenotype for genetic studies of successful cognitive aging. Am J Med Genet Part B Neuropsychiatr Genet Off Publ Int Soc Psychiatr Genet. 2008;147B(2):167173. doi:10.1002/ajmg.b.30483.CrossRefGoogle ScholarPubMed
Greenwood, TA, Beeri, MS, Schmeidler, J, et al. Heritability of cognitive functions in families of successful cognitive aging probands from the Central Valley of Costa Rica. J Alzheimers Dis JAD. 2011;27(4):897907. doi:10.3233/JAD-2011-110782.CrossRefGoogle ScholarPubMed
Rosero-Bixby, L, Dow, WH. Surprising SES gradients in mortality, health, and biomarkers in a Latin American population of adults. J Gerontol B Psychol Sci Soc Sci. 2009;64(1):105117. doi:10.1093/geronb/gbn004.CrossRefGoogle Scholar
Rosero-Bixby, L. The exceptionally high life expectancy of Costa Rican nonagenarians. Demography. 2008;45(3):673691. doi:10.1353/dem.0.0011.CrossRefGoogle ScholarPubMed
Rosero-Bixby, L, Dow, WH. Exploring why Costa Rica outperforms the United States in life expectancy: a tale of two inequality gradients. Proc Natl Acad Sci U S A. 2016;113(5):11301137. doi:10.1073/pnas.1521917112.CrossRefGoogle ScholarPubMed
Hughes, CP, Berg, L, Danziger, WL, et al. A new clinical scale for the staging of dementia. Br J Psychiatry J Ment Sci. 1982;140:566572. doi:10.1192/bjp.140.6.566.CrossRefGoogle ScholarPubMed
Folstein, MF, Folstein, SE, McHugh, PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189198. doi:10.1016/0022-3956(75)90026-6.CrossRefGoogle ScholarPubMed
Cheung, WY, Hubert, N, Landry, BS. A simple and rapid DNA microextraction method for plant, animal, and insect suitable for RAPD and other PCR analyses. PCR Methods Appl. 1993;3(1):6970. doi:10.1101/gr.3.1.69.CrossRefGoogle ScholarPubMed
Aryee, MJ, Jaffe, AE, Corrada-Bravo, H, et al. Minfi: a flexible and comprehensive Bioconductor package for the analysis of Infinium DNA methylation microarrays. Bioinforma Oxf Engl. 2014;30(10):13631369. doi:10.1093/bioinformatics/btu049.CrossRefGoogle ScholarPubMed
Fortin, J-P, Labbe, A, Lemire, M, et al. Functional normalization of 450k methylation array data improves replication in large cancer studies. Genome Biol. 2014;15(12):503. doi:10.1186/s13059-014-0503-2.CrossRefGoogle ScholarPubMed
Bibikova, M, Barnes, B, Tsan, C, et al. High density DNA methylation array with single CpG site resolution. Genomics. 2011;98(4):288295. doi:10.1016/j.ygeno.2011.07.007.CrossRefGoogle ScholarPubMed
Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14(10):R115. doi:10.1186/gb-2013-14-10-r115.CrossRefGoogle ScholarPubMed
Houseman, EA, Accomando, WP, Koestler, DC, et al. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics. 2012;13:86. doi:10.1186/1471-2105-13-86.CrossRefGoogle ScholarPubMed
Morris, TJ, Butcher, LM, Feber, A, et al. ChAMP: 450k chip analysis methylation pipeline. Bioinforma Oxf Engl. 2014;30(3):428430. doi:10.1093/bioinformatics/btt684.CrossRefGoogle ScholarPubMed
Sanchez-Mut, JV, Heyn, H, Silva, BA, et al. PM20D1 is a quantitative trait locus associated with Alzheimer’s disease. Nat Med. 2018;24(5):598603. doi:10.1038/s41591-018-0013-y.CrossRefGoogle ScholarPubMed
Sanchez-Mut, JV, Glauser, L, Monk, D, et al. Comprehensive analysis of PM20D1 QTL in Alzheimer’s disease. Clin Epigenetics. 2020;12(1):20. doi:10.1186/s13148-020-0814-y CrossRefGoogle ScholarPubMed
Horvath, S, Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat Rev Genet. 2018;19(6):371384. doi:10.1038/s41576-018-0004-3.CrossRefGoogle ScholarPubMed
Marioni, RE, Shah, S, McRae, AF, et al. DNA methylation age of blood predicts all-cause mortality in later life. Genome Biol. 2015;16:25. doi:10.1186/s13059-015-0584-6.CrossRefGoogle ScholarPubMed
Marioni, RE, Shah, S, McRae, AF, et al. The epigenetic clock is correlated with physical and cognitive fitness in the Lothian Birth Cohort 1936. Int J Epidemiol. 2015;44(4):13881396. doi:10.1093/ije/dyu277.CrossRefGoogle ScholarPubMed
Levine, ME, Lu, AT, Bennett, DA, et al. Epigenetic age of the pre-frontal cortex is associated with neuritic plaques, amyloid load, and Alzheimer’s disease related cognitive functioning. Aging. 2015;7(12):11981211. doi:10.18632/aging.100864.CrossRefGoogle ScholarPubMed
McEwen, LM, Morin, AM, Edgar, RD, et al. Differential DNA methylation and lymphocyte proportions in a Costa Rican high longevity region. Epigenetics Chromatin. 2017;10:21. doi:10.1186/s13072-017-0128-2.CrossRefGoogle Scholar
Horvath, S, Gurven, M, Levine, ME, et al. An epigenetic clock analysis of race/ethnicity, sex, and coronary heart disease. Genome Biol. 2016;17(1):171. doi:10.1186/s13059-016-1030-0.CrossRefGoogle Scholar
Armstrong, NJ, Mather, KA, Thalamuthu, A, et al. Aging, exceptional longevity and comparisons of the Hannum and Horvath epigenetic clocks. Epigenomics. 2017;9(5):689700. doi:10.2217/epi-2016-0179.CrossRefGoogle ScholarPubMed
Horvath, S, Pirazzini, C, Bacalini, MG, et al. Decreased epigenetic age of PBMCs from Italian semi-supercentenarians and their offspring. Aging. 2015;7(12):11591170. doi:10.18632/aging.100861.CrossRefGoogle ScholarPubMed
Heyn, H, Moran, S, Hernando-Herraez, I, et al. DNA methylation contributes to natural human variation. Genome Res. 2013;23(9):13631372. doi:10.1101/gr.154187.112.CrossRefGoogle ScholarPubMed
GTEx Consortium. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348(6235):648660. doi:10.1126/science.1262110.CrossRefGoogle Scholar
Li, QS, Vasanthakumar, A, Davis, JW, et al. Association of peripheral blood DNA methylation level with Alzheimer’s disease progression. Clin Epigenetics. 2021;13(1):191. doi:10.1186/s13148-021-01179-2.CrossRefGoogle ScholarPubMed
Wang, Q, Chen, Y, Readhead, B, et al. Longitudinal data in peripheral blood confirm that PM20D1 is a quantitative trait locus (QTL) for Alzheimer’s disease and implicate its dynamic role in disease progression. Clin Epigenetics. 2020;12(1):189. doi:10.1186/s13148-020-00984-5.CrossRefGoogle ScholarPubMed
Long, JZ, Svensson, KJ, Bateman, LA, et al. The secreted enzyme PM20D1 regulates lipidated amino acid uncouplers of mitochondria. Cell. 2016;166(2):424435. doi:10.1016/j.cell.2016.05.071.CrossRefGoogle ScholarPubMed
Larrick, JW, Larrick, JW, Mendelsohn, AR. Uncoupling mitochondrial respiration for diabesity. Rejuvenation Res. 2016;19(4):337340. doi:10.1089/rej.2016.1859.CrossRefGoogle ScholarPubMed
Profenno, LA, Porsteinsson, AP, Faraone, SV. Meta-analysis of Alzheimer’s disease risk with obesity, diabetes, and related disorders. Biol Psychiatry. 2010;67(6):505512. doi:10.1016/j.biopsych.2009.02.013.CrossRefGoogle ScholarPubMed
Simon-Sanchez, J, Schulte, C, Bras, JM, et al. Genome-Wide Association Study reveals genetic risk underlying Parkinson’s disease. Nat Genet. 2009;41(12):13081312. doi:10.1038/ng.487.CrossRefGoogle ScholarPubMed
Feinberg, AP, Irizarry, RA, Fradin, D, et al. Personalized epigenomic signatures that are stable over time and covary with body mass index. Sci Transl Med. 2010;2(49):49ra67. doi:10.1126/scitranslmed.3001262.CrossRefGoogle ScholarPubMed
Maltby, VE, Lea, RA, Sanders, KA, et al. Differential methylation at MHC in CD4+ T cells is associated with multiple sclerosis independently of HLA-DRB1. Clin Epigenetics. 2017;9:71. doi:10.1186/s13148-017-0371-1.CrossRefGoogle ScholarPubMed
Lavdas, AA, Grigoriou, M, Pachnis, V, et al. The medial ganglionic eminence gives rise to a population of early neurons in the developing cerebral cortex. J Neurosci Off J Soc Neurosci. 1999;19(18):78817888.CrossRefGoogle ScholarPubMed
Zhang, Z, Gutierrez, D, Li, X, et al. The LIM homeodomain transcription factor LHX6: a transcriptional repressor that interacts with pituitary homeobox 2 (PITX2) to regulate odontogenesis. J Biol Chem. 2013;288(4):24852500. doi:10.1074/jbc.M112.402933.CrossRefGoogle ScholarPubMed
Grigoriou, M, Tucker, AS, Sharpe, PT, et al. Expression and regulation of Lhx6 and Lhx7, a novel subfamily of LIM homeodomain encoding genes, suggests a role in mammalian head development. Dev Camb Engl. 1998;125(11):20632074.Google ScholarPubMed
Ben-David, O, Futerman, AH. The role of the ceramide acyl chain length in neurodegeneration: involvement of ceramide synthases. Neuromolecular Med. 2010;12(4):341350. doi:10.1007/s12017-010-8114-x.CrossRefGoogle ScholarPubMed
Becker, I, Wang-Eckhardt, L, Yaghootfam, A, et al. Differential expression of (dihydro)ceramide synthases in mouse brain: oligodendrocyte-specific expression of CerS2/Lass2. Histochem Cell Biol. 2008;129(2):233241. doi:10.1007/s00418-007-0344-0.CrossRefGoogle ScholarPubMed
Riebeling, C, Allegood, JC, Wang, E, et al. Two mammalian longevity assurance gene (LAG1) family members, trh1 and trh4, regulate dihydroceramide synthesis using different fatty acyl-CoA donors. J Biol Chem. 2003;278(44):4345243459. doi:10.1074/jbc.M307104200.CrossRefGoogle ScholarPubMed
Mizutani, Y, Kihara, A, Igarashi, Y. Mammalian Lass6 and its related family members regulate synthesis of specific ceramides. Biochem J. 2005;390(Pt 1):263271. doi:10.1042/BJ20050291.CrossRefGoogle ScholarPubMed
Radner, FPW, Marrakchi, S, Kirchmeier, P, et al. Mutations in CERS3 cause autosomal recessive congenital ichthyosis in humans. PLoS Genet. 2013;9(6):e1003536. doi:10.1371/journal.pgen.1003536.CrossRefGoogle ScholarPubMed
Jeon, SH, Lee, K, Lee, KS, et al. Characterization of the direct physical interaction of nc886, a cellular non-coding RNA, and PKR. FEBS Lett. 2012;586(19):34773484. doi:10.1016/j.febslet.2012.07.076.CrossRefGoogle ScholarPubMed
Jeon, SH, Johnson, BH, Lee, YS. A tumor surveillance model: a non-coding RNA senses neoplastic cells and its protein partner signals cell death. Int J Mol Sci. 2012;13(10):1313413139. doi:10.3390/ijms131013134.CrossRefGoogle Scholar
Fort, RS, Mathó, C, Geraldo, MV, et al. Nc886 is epigenetically repressed in prostate cancer and acts as a tumor suppressor through the inhibition of cell growth. BMC Cancer. 2018;18(1):127. doi:10.1186/s12885-018-4049-7.CrossRefGoogle ScholarPubMed
Lee, H-S, Lee, K, Jang, H-J, et al. Epigenetic silencing of the non-coding RNA nc886 provokes oncogenes during human esophageal tumorigenesis. Oncotarget. 2014;5(11):34723481. doi:10.18632/oncotarget.1927.CrossRefGoogle ScholarPubMed
Lee, K-S, Park, J-L, Lee, K, et al. nc886, a non-coding RNA of anti-proliferative role, is suppressed by CpG DNA methylation in human gastric cancer. Oncotarget. 2014;5(11):39443955. doi:10.18632/oncotarget.2047.CrossRefGoogle ScholarPubMed
Romanelli, V, Nakabayashi, K, Vizoso, M, et al. Variable maternal methylation overlapping the nc886/vtRNA2-1 locus is locked between hypermethylated repeats and is frequently altered in cancer. Epigenetics. 2014;9(5):783790. doi:10.4161/epi.28323.CrossRefGoogle ScholarPubMed
Korrodi-Gregório, L, Abrantes, J, Muller, T, et al. Not so pseudo: the evolutionary history of protein phosphatase 1 regulatory subunit 2 and related pseudogenes. BMC Evol Biol. 2013;13:242. doi:10.1186/1471-2148-13-242.CrossRefGoogle ScholarPubMed
Wong, GW, Yasuda, S, Madhusudhan, MS, et al. Human tryptase epsilon (PRSS22), a new member of the chromosome 16p13.3 family of human serine proteases expressed in airway epithelial cells. J Biol Chem. 2001;276(52):4916949182. doi:10.1074/jbc.M108677200.CrossRefGoogle ScholarPubMed
Fagerberg, L, Hallström, BM, Oksvold, P, et al. Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics MCP. 2014;13(2):397406. doi:10.1074/mcp.M113.035600.CrossRefGoogle ScholarPubMed
Hindorff, LA, Bonham, VL, Brody, LC, et al. Prioritizing diversity in human genomics research. Nat Rev Genet. 2018;19(3):175185. doi:10.1038/nrg.2017.89.CrossRefGoogle ScholarPubMed
Peterson, RE, Kuchenbaecker, K, Walters, RK, et al. Genome-wide association studies in ancestrally diverse populations: opportunities, methods, pitfalls, and recommendations. Cell. 2019;179(3):589603. doi:10.1016/j.cell.2019.08.051.CrossRefGoogle ScholarPubMed