Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

A common haplotype lowers PU.1 expression in myeloid cells and delays onset of Alzheimer's disease

Abstract

A genome-wide survival analysis of 14,406 Alzheimer's disease (AD) cases and 25,849 controls identified eight previously reported AD risk loci and 14 novel loci associated with age at onset. Linkage disequilibrium score regression of 220 cell types implicated the regulation of myeloid gene expression in AD risk. The minor allele of rs1057233 (G), within the previously reported CELF1 AD risk locus, showed association with delayed AD onset and lower expression of SPI1 in monocytes and macrophages. SPI1 encodes PU.1, a transcription factor critical for myeloid cell development and function. AD heritability was enriched within the PU.1 cistrome, implicating a myeloid PU.1 target gene network in AD. Finally, experimentally altered PU.1 levels affected the expression of mouse orthologs of many AD risk genes and the phagocytic activity of mouse microglial cells. Our results suggest that lower SPI1 expression reduces AD risk by regulating myeloid gene expression and cell function.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: AD-survival and myeloid eQTL associations.
Figure 2: SPI1 (PU.1) expression and ChIP-seq analysis.
Figure 3: PU.1 modulates the phagocytic activity of BV2 microglial cells.
Figure 4: Genes regulated with differential expression of Spi1 in BV2 microglial cells.

Similar content being viewed by others

References

  1. Lambert, J.-C. et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat. Genet. 45, 1452–1458 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Naj, A.C. et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease. Nat. Genet. 43, 436–441 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Harold, D. et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease. Nat. Genet. 41, 1088–1093 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Seshadri, S. et al. Genome-wide analysis of genetic loci associated with Alzheimer disease. J. Am. Med. Assoc. 303, 1832–1840 (2010).

    CAS  Google Scholar 

  5. Hollingworth, P. et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nat. Genet. 43, 429–435 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Naj, A.C. et al. Effects of multiple genetic loci on age at onset in late-onset Alzheimer disease: a genome-wide association study. JAMA Neurol. 71, 1394–1404 (2014).

    PubMed  PubMed Central  Google Scholar 

  7. Kamboh, M.I. et al. Genome-wide association analysis of age-at-onset in Alzheimer's disease. Mol. Psychiatry 17, 1340–1346 (2012).

    CAS  PubMed  Google Scholar 

  8. Bennett, C. et al. Evidence that the APOE locus influences rate of disease progression in late onset familial Alzheimer's Disease but is not causative. Am. J. Med. Genet. 60, 1–6 (1995).

    CAS  PubMed  Google Scholar 

  9. Slooter, A.J. et al. Risk estimates of dementia by apolipoprotein E genotypes from a population-based incidence study: the Rotterdam Study. Arch. Neurol. 55, 964–968 (1998).

    CAS  PubMed  Google Scholar 

  10. Thambisetty, M., An, Y. & Tanaka, T. Alzheimer's disease risk genes and the age-at-onset phenotype. Neurobiol. Aging 34, 2696 e1–2696.e5 (2013).

    PubMed  PubMed Central  Google Scholar 

  11. Jones, E.L. et al. Evidence that PICALM affects age at onset of Alzheimer's dementia in Down syndrome. Neurobiol. Aging 34, 2441 e1–2441.e5 (2013).

    PubMed  PubMed Central  Google Scholar 

  12. Cruchaga, C. et al. GWAS of cerebrospinal fluid tau levels identifies risk variants for Alzheimer's disease. Neuron 78, 256–268 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Kauwe, J.S.K. et al. Alzheimer's disease risk variants show association with cerebrospinal fluid amyloid beta. Neurogenetics 10, 13–17 (2009).

    CAS  PubMed  Google Scholar 

  14. Gusev, A. et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 48, 245–252 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Jonsson, T. et al. Variant of TREM2 associated with the risk of Alzheimer's disease. N. Engl. J. Med. 368, 107–116 (2013).

    CAS  PubMed  Google Scholar 

  16. Guerreiro, R. et al. TREM2 variants in Alzheimer's disease. N. Engl. J. Med. 368, 117–127 (2013).

    CAS  PubMed  Google Scholar 

  17. Bradshaw, E.M. et al. CD33 Alzheimer's disease locus: altered monocyte function and amyloid biology. Nat. Neurosci. 16, 848–850 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Zhang, B. et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer's disease. Cell 153, 707–720 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Gjoneska, E. et al. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer's disease. Nature 518, 365–369 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Chan, G. et al. CD33 modulates TREM2: convergence of Alzheimer loci. Nat. Neurosci. 18, 1556–1558 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Raj, T. et al. Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science 344, 519–523 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhu, Z. et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat. Genet. 48, 481–487 (2016).

    CAS  PubMed  Google Scholar 

  23. GTEx Consortium. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

  24. Finucane, H.K. et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet. 47, 1228–1235 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Cross-Disorder Group of the Psychiatric Genomics Consortium. Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet 381, 1371–1379 (2013).

  26. Garnier, S. et al. Genome-wide haplotype analysis of cis expression quantitative trait loci in monocytes. PLoS Genet. 9, e1003240 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Fairfax, B.P. et al. Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression. Science 343, 1246949 (2014).

    PubMed  PubMed Central  Google Scholar 

  28. Zhang, Y. et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89, 37–53 (2016).

    CAS  PubMed  Google Scholar 

  29. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Bennett, M.L. et al. New tools for studying microglia in the mouse and human CNS. Proc. Natl. Acad. Sci. USA 113, E1738–E1746 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Giambartolomei, C. et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 10, e1004383 (2014).

    PubMed  PubMed Central  Google Scholar 

  32. Hikami, K. et al. Association of a functional polymorphism in the 3′-untranslated region of SPI1 with systemic lupus erythematosus. Arthritis Rheum. 63, 755–763 (2011).

    CAS  PubMed  Google Scholar 

  33. Ward, L.D. & Kellis, M. HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res. 40, D930–D934 (2012).

    CAS  PubMed  Google Scholar 

  34. Pham, T.H. et al. Dynamic epigenetic enhancer signatures reveal key transcription factors associated with monocytic differentiation states. Blood 119, e161–e171 (2012).

    CAS  PubMed  Google Scholar 

  35. Steinberg, S. et al. Loss-of-function variants in ABCA7 confer risk of Alzheimer's disease. Nat. Genet. 47, 445–447 (2015).

    CAS  PubMed  Google Scholar 

  36. Bernstein, B.E. et al.; ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Google Scholar 

  37. Sakai, K. et al. A neuronal VLDLR variant lacking the third complement-type repeat exhibits high capacity binding of apoE containing lipoproteins. Brain Res. 1276, 11–21 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Bajari, T.M., Strasser, V., Nimpf, J. & Schneider, W.J. A model for modulation of leptin activity by association with clusterin. FASEB J. 17, 1505–1507 (2003).

    CAS  PubMed  Google Scholar 

  39. Speliotes, E.K. et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat. Genet. 42, 937–948 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Strawbridge, R.J. et al. Genome-wide association identifies nine common variants associated with fasting proinsulin levels and provides new insights into the pathophysiology of type 2 diabetes. Diabetes 60, 2624–2634 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Satoh, J., Asahina, N., Kitano, S. & Kino, Y. A comprehensive profile of ChIP-seq-based PU.1/Spi1 target genes in microglia. Gene Regul. Syst. Bio. 8, 127–139 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Daniel, B. et al. The active enhancer network operated by liganded RXR supports angiogenic activity in macrophages. Genes Dev. 28, 1562–1577 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. McKercher, S.R. et al. Targeted disruption of the PU.1 gene results in multiple hematopoietic abnormalities. EMBO J. 15, 5647–5658 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Beers, D.R. et al. Wild-type microglia extend survival in PU.1 knockout mice with familial amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA 103, 16021–16026 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Mak, K.S., Funnell, A.P.W., Pearson, R.C.M. & Crossley, M. PU.1 and haematopoietic cell fate: dosage matters. Int. J. Cell Biol. 2011, 808524 (2011).

    PubMed  PubMed Central  Google Scholar 

  46. Smith, A.M. et al. The transcription factor PU.1 is critical for viability and function of human brain microglia. Glia 61, 929–942 (2013).

    PubMed  Google Scholar 

  47. Butovsky, O. et al. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nat. Neurosci. 17, 131–143 (2014).

    CAS  PubMed  Google Scholar 

  48. Wang, Y. et al. IL-34 is a tissue-restricted ligand of CSF1R required for the development of Langerhans cells and microglia. Nat. Immunol. 13, 753–760 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Dagher, N.N. et al. Colony-stimulating factor 1 receptor inhibition prevents microglial plaque association and improves cognition in 3xTg-AD mice. J. Neuroinflammation 12, 139 (2015).

    PubMed  PubMed Central  Google Scholar 

  50. Alpérovitch, A. et al.; 3C Study Group. Vascular factors and risk of dementia: design of the Three-City Study and baseline characteristics of the study population. Neuroepidemiology 22, 316–325 (2003).

    Google Scholar 

  51. Dreses-Werringloer, U. et al. A polymorphism in CALHM1 influences Ca2+ homeostasis, Abeta levels, and Alzheimer's disease risk. Cell 133, 1149–1161 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Lopez, O.L. et al. Evaluation of dementia in the cardiovascular health cognition study. Neuroepidemiology 22, 1–12 (2003).

    PubMed  Google Scholar 

  53. Dawber, T.R. & Kannel, W.B. The Framingham study. An epidemiological approach to coronary heart disease. Circulation 34, 553–555 (1966).

    CAS  PubMed  Google Scholar 

  54. Feinleib, M., Kannel, W.B., Garrison, R.J., McNamara, P.M. & Castelli, W.P. The Framingham offspring study. Design and preliminary data. Prev. Med. 4, 518–525 (1975).

    CAS  PubMed  Google Scholar 

  55. Splansky, G.L. et al. The third generation cohort of the National Heart, Lung, and Blood Institute's Framingham heart study: design, recruitment, and initial examination. Am. J. Epidemiol. 165, 1328–1335 (2007).

    PubMed  Google Scholar 

  56. Beiser, A., D'Agostino, R.B. Sr., Seshadri, S., Sullivan, L.M. & Wolf, P.A. Computing estimates of incidence, including lifetime risk: Alzheimer's disease in the Framingham study. the Practical Incidence Estimators (PIE) macro. Stat. Med. 19, 1495–1522 (2000).

    CAS  PubMed  Google Scholar 

  57. Bachman, D.L. et al. Incidence of dementia and probable Alzheimer's disease in a general population: the Framingham Study. Neurology 43, 515–519 (1993).

    CAS  PubMed  Google Scholar 

  58. Farmer, M.E. et al. Neuropsychological test performance in Framingham: a descriptive study. Psychol. Rep. 60, 1023–1040 (1987).

    CAS  PubMed  Google Scholar 

  59. DeCarli, C. et al. Measures of brain morphology and infarction in the Framingham heart study: establishing what is normal. Neurobiol. Aging 26, 491–510 (2005).

    PubMed  Google Scholar 

  60. Au, R. et al. New norms for a new generation: cognitive performance in the framingham offspring cohort. Exp. Aging Res. 30, 333–358 (2004).

    PubMed  Google Scholar 

  61. Seshadri, S. & Wolf, P.A. Lifetime risk of stroke and dementia: current concepts, and estimates from the Framingham Study. Lancet Neurol. 6, 1106–1114 (2007).

    PubMed  Google Scholar 

  62. Hofman, A. et al. The Rotterdam Study: 2014 objectives and design update. Eur. J. Epidemiol. 28, 889–926 (2013).

    CAS  PubMed  Google Scholar 

  63. Fagan, A.M. et al. Inverse relation between in vivo amyloid imaging load and cerebrospinal fluid Abeta42 in humans. Ann. Neurol. 59, 512–519 (2006).

    CAS  PubMed  Google Scholar 

  64. Peskind, E., Nordberg, A., Darreh-Shori, T. & Soininen, H. Safety of lumbar puncture procedures in patients with Alzheimer's disease. Curr. Alzheimer Res. 6, 290–292 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Grimmer, T. et al. Beta amyloid in Alzheimer's disease: increased deposition in brain is reflected in reduced concentration in cerebrospinal fluid. Biol. Psychiatry 65, 927–934 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Blennow, K., Hampel, H., Weiner, M. & Zetterberg, H. Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nat. Rev. Neurol. 6, 131–144 (2010).

    CAS  PubMed  Google Scholar 

  67. Willer, C.J., Li, Y. & Abecasis, G.R. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–2191 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Peskind, E.R. et al. Age and apolipoprotein E*4 allele effects on cerebrospinal fluid beta-amyloid 42 in adults with normal cognition. Arch. Neurol. 63, 936–939 (2006).

    PubMed  Google Scholar 

  69. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Howie, B.N., Donnelly, P. & Marchini, J. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet. 5, e1000529 (2009).

    PubMed  PubMed Central  Google Scholar 

  71. Cox, D.R. Regression Models and Life-Tables. J. R. Stat. Soc. B. 34, 187–220 (1972).

    Google Scholar 

  72. Prentice, R.L. & Breslow, N.E. Retrospective studies and failure time models. Biometrika 65, 153–158 (1978).

    Google Scholar 

  73. van der Net, J.B. et al. Cox proportional hazards models have more statistical power than logistic regression models in cross-sectional genetic association studies. Eur. J. Hum. Genet. 16, 1111–1116 (2008).

    CAS  PubMed  Google Scholar 

  74. Pruim, R.J. et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26, 2336–2337 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Zhou, X. et al. Exploring long-range genome interactions using the WashU Epigenome Browser. Nat. Methods 10, 375–376 (2013).

    CAS  PubMed  Google Scholar 

  76. Griffon, A. et al. Integrative analysis of public ChIP-seq experiments reveals a complex multi-cell regulatory landscape. Nucleic Acids Res. 43, e27 (2015).

    PubMed  Google Scholar 

  77. Sloan, C.A. et al. ENCODE data at the ENCODE portal. Nucleic Acids Res. 44, D726–D732 (2016).

    CAS  PubMed  Google Scholar 

  78. Roe, J.S., Mercan, F., Rivera, K., Pappin, D.J. & Vakoc, C.R. BET bromodomain inhibition suppresses the function of hematopoietic transcription factors in acute myeloid leukemia. Mol. Cell 58, 1028–1039 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Wang, X. & Seed, B. A PCR primer bank for quantitative gene expression analysis. Nucleic Acids Res. 31, e154 (2003).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the patients, control subjects and their family members for participating in and supporting the research projects included in this study. We thank M. Diamond (UT Southwestern Medical Center) for the BV2 cell line and the Flow Cytometry CORE at the Icahn School of Medicine at Mount Sinai Hospital.

IGAP: This study incorporated imputed summary results from the GERAD1 genome-wide association study. GERAD was supported by the Wellcome Trust, the MRC, Alzheimer's Research UK (ARUK) and the Welsh government. ADGC and CHARGE were supported by the US National Institutes of Health, National Institute on Aging (NIH-NIA), including grants U01 AG032984 and R01 AG033193. CHARGE was also supported by Erasmus Medical Center and Erasmus University.

Cardiff University was supported by the Wellcome Trust, Medical Research Council (MRC), Alzheimer's Research UK (ARUK) and the Welsh Assembly Government. Cambridge University and Kings College London acknowledge support from the MRC. ARUK supported sample collections at the South West Dementia Bank and the Universities of Nottingham, Manchester and Belfast. The Belfast group acknowledges support from the Alzheimer′s Society, Ulster Garden Villages, N. Ireland R&D Office and the Royal College of Physicians/Dunhill Medical Trust. The MRC and Mercer's Institute for Research on Aging supported the Trinity College group. The South West Dementia Brain Bank acknowledges support from Bristol Research into Alzheimer's and Care of the Elderly. The Charles Wolfson Charitable Trust supported the OPTIMA group. Washington University was funded by NIH grants, Barnes Jewish Foundation and by the Charles and Joanne Knight Alzheimer's Research Initiative. Patient recruitment for the MRC Prion Unit/UCL Department of Neurodegenerative Disease collection was supported by the UCLH/UCL Biomedical Centre and NIHR Queen Square Dementia Biomedical Research Unit. LASER-AD was funded by Lundbeck SA. The Bonn group was supported by the German Federal Ministry of Education and Research (BMBF), Competence Network Dementia and Competence Network Degenerative Dementia and by the Alfried Krupp von Bohlen und Halbach-Stiftung. The GERAD Consortium also used samples ascertained by the NIMH AD Genetics Initiative.

The KORA F4 studies were financed by Helmholtz Zentrum München; German Research Center for Environmental Health; BMBF; German National Genome Research Network and the Munich Center of Health Sciences. The Heinz Nixdorf Recall cohort was funded by the Heinz Nixdorf Foundation (Dr. jur. G.Schmidt, Chairman) and BMBF. Coriell Cell Repositories is supported by NINDS and the Intramural Research Program of the National Institute on Aging. We acknowledge use of genotype data from the 1958 Birth Cohort collection, funded by the MRC and the Wellcome Trust, which was genotyped by the Wellcome Trust Case Control Consortium and the Type-1 Diabetes Genetics Consortium, sponsored by the National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Allergy and Infectious Diseases, National Human Genome Research Institute, National Institute of Child Health and Human Development and Juvenile Diabetes Research Foundation International.

ADNI: Data collection and sharing for this project was funded by the Alzheimer′s Disease Neuroimaging Initiative (National Institutes of Health Grant U01 AG024904) and DOD ADNI (Department of Defense award number W81XWH-12-2-0012). ADNI is funded by the National Institute on Aging and the National Institute of Biomedical Imaging and Bioengineering and through generous contributions from the following: AbbVie, Alzheimer's Association; Alzheimer's Drug Discovery Foundation; Araclon Biotech; BioClinica, Inc.; Biogen; Bristol-Myers Squibb Company; CereSpir, Inc.; Eisai Inc.; Elan Pharmaceuticals, Inc.; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche Ltd and its affiliated company, Genentech, Inc.; Fujirebio; GE Healthcare; IXICO Ltd.; Janssen Alzheimer Immunotherapy Research & Development, LLC.; Johnson & Johnson Pharmaceutical Research & Development LLC.; Lumosity; Lundbeck; Merck & Co., Inc.; Meso Scale Diagnostics, LLC.; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer Inc.; Piramal Imaging; Servier; Takeda Pharmaceutical Company; and Transition Therapeutics. The Canadian Institutes of Health Research is providing funds to support ADNI clinical sites in Canada. Private sector contributions are facilitated by the Foundation for the National Institutes of Health (https://fnih.org/). The grantee organization is the Northern California Institute for Research and Education, and the study is coordinated by the Alzheimer's Disease Cooperative Study at the University of California, San Diego. ADNI data are disseminated by the Laboratory for Neuro Imaging at the University of Southern California.

We thank the Cardiogenics (European Project reference LSHM-CT-2006-037593) project for providing summary statistics for the cis-eQTL-based analyses. We also thank the ENCODE Consortium and R. Myers' lab (HAIB) for providing ChIP-seq datasets.

This work was supported by grants from the National Institutes of Health (U01 AG049508, R01-AG035083 and RF-AG054011 (to A.M.G.) and R01-AG044546 and RF1AG053303 (to C.C.)), the JPB Foundation (to A.M.G.) and F Prime (to A.M.G.). The recruitment and clinical characterization of research participants at Washington University were supported by NIH P50 AG05681, P01 AG03991 and P01 AG026276. Kuan-lin Huang received fellowship funding in part from the Ministry of Education in Taiwan and the Lucille P. Markey Special Emphasis Pathway in Human Pathobiology. Ke Hao is partially supported by the National Natural Science Foundation of China (Grant Nos. 21477087 and 91643201) and by the Ministry of Science and Technology of China (Grant No. 2016YFC0206507). This work was supported by access to equipment made possible by the Hope Center for Neurological Disorders and the Departments of Neurology and Psychiatry at Washington University School of Medicine.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

A.M.G., E.M. and K.-L.H. conceived and designed the experiments. K.-L.H., S.C.J., O.H., A.D., M.K., J.C., J.C.L., V.C., C.B., B.G.-B., Y.D., A.M., T.R., A.E.R., J.L.D.-A., M.V.F, L.I., B.Z., I.B., C.C. and E.M. performed data analysis. A.A.P. performed phagocytosis assays, western blotting and qPCR validation. S.B., B.P.F., J.B., R.S., V.E.-P., R.M., J.L.H., L.A.F., M.A.P.-V., S.S., J.W., P.A., G.D.S., J.S.K.K., K.H. and C.C. provided and processed the data. A.M.G. supervised data analysis and functional experiments. K.-L.H., A.A.P., E.M. and A.M.G. wrote and edited the manuscript. All authors read and approved the manuscript.

Corresponding author

Correspondence to Alison M Goate.

Ethics declarations

Competing interests

A.M.G. is on the scientific advisory board for Denali Therapeutics and has served as a consultant for AbbVie and Cognition Therapeutics.

Additional information

A full list of members and affiliations appears in the Supplementary Note.

Integrated supplementary information

Supplementary Figure 1 Result and quality control analysis of the IGAP AD-survival meta-analysis.

(a) Manhattan plot and QQ-plot of the GWAS. The final meta-analysis showed little evidence of genomic inflation (λ = 1.026). (b) The average standard error versus the number of cohorts with consistent directionalities of effect sizes.

Supplementary Figure 2 Kaplan-Meier plots of AAOS associations.

Kaplan-Meier plots of survival analysis associations in the ADGC cohort of (a) rs1057233, (b) rs10919252, (c) rs567075, (d) rs7867518, (e) rs7930318, (f) rs4803758.

Supplementary Figure 3 Forest plots of AAOS associations.

Forest plots of survival analysis associations across IGAP cohorts of (a) rs1057233, (b) rs10919252, (c) rs567075, (d) rs7867518, (e) rs7930318, (f) rs4803758.

Supplementary Figure 4 Cell-type-specific expression of eQTL-associated genes in brain.

Cell-type specific expression of MS4A4A (no mouse homolog available), SPI1, MYBPC3, MS4A6A and SELL in human and mouse brains based on the brain RNA-Seq database.

Supplementary Figure 5 Linkage disequilibrium (LD) plot of SNPs of interest in the SPI1 (CELF1) locus.

Supplementary Figure 6 SMR plots showing the associations at the SPI1 locus.

SMR plots showing the associations at the SPI1/CELF1 locus from AAOS GWAS and eQTLs in (a) monocytes and (b) macrophages.

Supplementary Figure 7 SPI1 (PU.1) ChIP-seq binding sites and other epigenetic signatures at AD-associated loci in human CD14+ monocytes.

PU.1 binding sites, DNase I hypersensitive sites, histone modifications, and chromatin states at the locus of (a) ABCA7, (b) APOE, (c) BIN1, (d) SPI1, (e) PICALM, and (f) TYROBP.

Supplementary Figure 8 Analysis of phagocytosis in BV2 microglial cells.

(a) Flow cytometry histograms of BV2 cells transfected with pcDNA3 (pcDNA) or pcDNA3-FLAG-PU.1 (FLAG-PU.1) with pCMV-GFP for overexpression and scrambled shRNA (shSCR) or shRNA targeting PU.1 (shA, shB and shD) in pGFP-V-RS vector for knock-down of PU.1 after 3 hours of incubation with pHrodo-labeled zymosan. Cells were gated on GFP+ population. (b) Flow cytometry analysis of the number of gated cells in a presented as mean ± s.d., pcDNA 67.03 ± 6.883, pcDNA + 1 μM Cyt 15.64 ± 16.24, FLAG-PU.1 82.71 ± 4.74, shSCR 77.17 ± 3.115, shA 48.63 ± 2.285, shB 28.92 ± 2.495, shD 22.76 ± 1.595. pcDNA vs pcDNA + 1 μM Cyt P < 0.0001, pcDNA vs FLAG-PU.1 P = 0.0306, shSCR vs shA P = 0.0002, shSCR vs shB P < 0.0001, shSCR vs shD P < 0.0001. F(6,13) = 58.68, n = 3. (c) Flow cytometry analysis of the geometric mean fluorescent pHrodo intensity in a presented as mean ± s.d., pcDNA 10952 ± 2206, pcDNA + 1 μM Cyt 1533 ± 47, FLAG-PU.1 15226 ± 2701, shSCR 13129 ± 4617, shA 9937 ± 2168, shB 8872 ± 2019, shD 8754 ± 1856. pcDNA vs pcDNA + 1 μM Cyt P = 0.0092. F(6,13) = 6.228, n = 3. (d) Flow cytometry histograms of BV2 cells transfected as in (a) and gated on GFP- populations. (e) Flow cytometry analysis of number of gated cells in d presented as mean ± s.d., pcDNA 63.92 ± 6.575, pcDNA + 1 μM Cyt 14.21 ± 13.66, FLAG-PU.1 67.54 ± 4.826, shSCR 68.31 ± 5.784, shA 67.27 ± 4.144, shB 65.19 ± 4.268, shD 60.3 ± 2.181. pcDNA vs pcDNA + 1 μM Cyt P < 0.0001. F(6,13) = 22.53, n = 3. (f) Flow cytometry analysis of geometric mean fluorescent pHrodo intensity in d presented as mean ± s.d., pcDNA 9186 ± 2863, pcDNA + 1 μM Cyt 1545 ± 147, FLAG-PU.1 9931 ± 2458, shSCR 9849 ± 3012, shA 10903 ± 2949, shB 10912 ± 2494, shD 10934 ± 2685. pcDNA vs pcDNA + 1 μM Cyt P = 0.0367. F(6,13) = 3.473, n = 3. (g) Phagocytic index of BV2 GFP- cells analyzed in (e) and (f) presented as mean ± s.d., pcDNA 0.5954 ± 0.2223, pcDNA + 1 μM Cyt 0.0209 ± 0.0189, FLAG-PU.1 0.6745 ± 0.188, shSCR 0.6765 ± 0.2274, shA 0.7382 ± 0.2255, shB 0.7131 ± 0.1742, shD 0.6612 ± 0.1748. pcDNA vs pcDNA + 1 μM Cyt P = 0.0331. F(6,13) = 3.53, n = 3. Cytochalasin D (Cyt) treatment was used as a negative control. * P < 0.05, ** P < 0.01, *** P < 0.001, one-way ANOVA with Sidak’s post hoc multiple comparisons test between selected groups.

Supplementary Figure 9 Expression levels of genes related to phagocytosis that were not affected by altered Spi1 expression.

BV2 cells were transiently transfected with pcDNA3-FLAG-PU.1 and pCMV-GFP or pGFP-V-RS-shB against PU.1. pcDNA3 and pGFP-V-RS-shSCR were used as controls. RNA was extracted from sorted GFP+ cells and used for qPCR validation of expression levels for genes of interest. Values are presented as mean ± s.d., n = 4 samples collected independently.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9. (PDF 1328 kb)

Supplementary Methods Checklist (PDF 196 kb)

Supplementary Note (PDF 68 kb)

Supplementary Table 1

Quality control of the age-at-onset survival GWAS. The number of filtered SNPs and samples are shown for each of the 7 cohort included for the IGAP meta-analysis. Additionally, the Schoenfeld P values were recorded for each of the 22 top AAOS GWAS SNPs across the 7 IGAP cohorts, and the covariates in Cox models for the ADGC and GERAD cohorts. (XLSX 16 kb)

Supplementary Table 2

Association results from the previous IGAP logistic regression and the matching cohort logistic regression of the 22 top AD-associated SNPs in AAOS GWAS. (XLSX 12 kb)

Supplementary Table 3

The 22 top SNPs significantly associated with age-at-onset defined survival in AD and their tag SNPs (R2 ≥ 0.8). (XLSX 476 kb)

Supplementary Table 4

Significant cis-eQTL associations between the 22 survival-associated SNPs across 10 brain regions in the BRAINEAC dataset. Bonferroni-corrected threshold: P=0.05/292,000 probes = 1.7x10-7. (XLSX 8 kb)

Supplementary Table 5

Significant cis-eQTL associations between the 22 survival-associated SNPs across tissues in the GTEx dataset. (XLSX 72 kb)

Supplementary Table 6

LD score regression analysis results for IGAP AD and PGC SCZ GWAS SNP heritability partitioned using 220 human cell types (CT, and 10 cell type groups, CTG)-specific functional annotations as described by Finucane et al. (XLSX 116 kb)

Supplementary Table 7

Replication of monocyte/macrophage cis-eQTL associations in CD14+ human monocytes from 432 individuals of European ancestry. Bonferroni-corrected threshold: P=0.05/15421 probes = 3.24x10-6. (XLSX 8 kb)

Supplementary Table 8

Coloc analysis results for the SPI1/CELF1, MS4A and SELL loci, using AAOS GWAS SNPs and Cardiogenics monocyte (MC) or macrophage (MP) cis-eQTL datasets. Estimated posterior probabilities for the following mutually exclusive hypotheses are shown in bold when surpassing the threshold PP ≥ 0.8: H0, neither trait has a genetic association in the region; H1: only trait 1 (cis-eQTL) has a genetic association in the region; H2: only trait 2 (GWAS) has a genetic association in the region; H3: both traits are associated, but with different causal variants; H4: both traits are associated and share a single causal variant. (XLSX 88 kb)

Supplementary Table 9

Conditional analysis of monocyte/macrophage SPI1 eQTL associations of 6 SNPs of interest: rs1057233, rs10838698, rs10838699, rs7928163, rs1377416, and rs10838725. Bonferroni-corrected threshold: P=0.05/6 SNPs = 0.00833. (XLSX 11 kb)

Supplementary Table 10

SMR/HEIDI analysis results for 6 SNPs of interest in the SPI1/CELF1 locus: rs1057233, rs10838698, rs10838699, rs7928163, rs1377416 and rs10838725, using AAOS GWAS SNPs and Cardiogenics monocyte (MC) or macrophage (MP) cis-eQTL datasets. Bonferroni-corrected threshold: SMR P=0.05/(6 SNPs x 17 probes) = 0.00049 [green cells]. A nominal significance threshold P ≥ 0.05 [red cells] is used for the HEIDI test as motivated by Zhu et al. Only probes with nominally significant SMR P are shown. (XLSX 84 kb)

Supplementary Table 11

AD-associated genes expressed in human brain myeloid cells with nearby SPI1 (PU.1) binding sites in human blood myeloid cells. (XLSX 45 kb)

Supplementary Table 12

LD score regression analysis results for IGAP AD and PGC SCZ GWAS SNP heritability partitioned using SPI1 (PU.1) ChIP-Seq binding sites in human monocytes and macrophages, as well as SPI1 (PU.1) and POLR2AphosphoS5 ChIP-Seq binding sites in human HL60 cells. (XLSX 13 kb)

Supplementary Table 13

Genes regulated in BV2 microglial cells with differential expression of Spi1. (XLSX 12 kb)

Supplementary Table 14

Primers used for qPCR validation of gene expression in BV2 microglial cells. (XLSX 14 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, Kl., Marcora, E., Pimenova, A. et al. A common haplotype lowers PU.1 expression in myeloid cells and delays onset of Alzheimer's disease. Nat Neurosci 20, 1052–1061 (2017). https://doi.org/10.1038/nn.4587

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4587

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing