Skip to main content
  • Submit
  • About
    • Editorial Board
    • PNAS Staff
    • FAQ
    • Accessibility Statement
    • Rights and Permissions
    • Site Map
  • Contact
  • Journal Club
  • Subscribe
    • Subscription Rates
    • Subscriptions FAQ
    • Open Access
    • Recommend PNAS to Your Librarian
  • Log in
  • My Cart

Main menu

  • Home
  • Articles
    • Current
    • Special Feature Articles - Most Recent
    • Special Features
    • Colloquia
    • Collected Articles
    • PNAS Classics
    • List of Issues
  • Front Matter
  • News
    • For the Press
    • This Week In PNAS
    • PNAS in the News
  • Podcasts
  • Authors
    • Information for Authors
    • Editorial and Journal Policies
    • Submission Procedures
    • Fees and Licenses
  • Submit
  • About
    • Editorial Board
    • PNAS Staff
    • FAQ
    • Accessibility Statement
    • Rights and Permissions
    • Site Map
  • Contact
  • Journal Club
  • Subscribe
    • Subscription Rates
    • Subscriptions FAQ
    • Open Access
    • Recommend PNAS to Your Librarian

User menu

  • Log in
  • My Cart

Search

  • Advanced search
Home
Home

Advanced Search

  • Home
  • Articles
    • Current
    • Special Feature Articles - Most Recent
    • Special Features
    • Colloquia
    • Collected Articles
    • PNAS Classics
    • List of Issues
  • Front Matter
  • News
    • For the Press
    • This Week In PNAS
    • PNAS in the News
  • Podcasts
  • Authors
    • Information for Authors
    • Editorial and Journal Policies
    • Submission Procedures
    • Fees and Licenses

New Research In

Physical Sciences

Featured Portals

  • Physics
  • Chemistry
  • Sustainability Science

Articles by Topic

  • Applied Mathematics
  • Applied Physical Sciences
  • Astronomy
  • Computer Sciences
  • Earth, Atmospheric, and Planetary Sciences
  • Engineering
  • Environmental Sciences
  • Mathematics
  • Statistics

Social Sciences

Featured Portals

  • Anthropology
  • Sustainability Science

Articles by Topic

  • Economic Sciences
  • Environmental Sciences
  • Political Sciences
  • Psychological and Cognitive Sciences
  • Social Sciences

Biological Sciences

Featured Portals

  • Sustainability Science

Articles by Topic

  • Agricultural Sciences
  • Anthropology
  • Applied Biological Sciences
  • Biochemistry
  • Biophysics and Computational Biology
  • Cell Biology
  • Developmental Biology
  • Ecology
  • Environmental Sciences
  • Evolution
  • Genetics
  • Immunology and Inflammation
  • Medical Sciences
  • Microbiology
  • Neuroscience
  • Pharmacology
  • Physiology
  • Plant Biology
  • Population Biology
  • Psychological and Cognitive Sciences
  • Sustainability Science
  • Systems Biology
Research Article

An in vivo screen of noncoding loci reveals that Daedalus is a gatekeeper of an Ikaros-dependent checkpoint during haematopoiesis

View ORCID ProfileChristian C. D. Harman, View ORCID ProfileWill Bailis, Jun Zhao, View ORCID ProfileLouisa Hill, Rihao Qu, Ruaidhrí P. Jackson, Justin A. Shyer, Holly R. Steach, Yuval Kluger, View ORCID ProfileLoyal A. Goff, View ORCID ProfileJohn L. Rinn, Adam Williams, Jorge Henao-Mejia, and Richard A. Flavell
PNAS January 19, 2021 118 (3) e1918062118; https://doi.org/10.1073/pnas.1918062118
Christian C. D. Harman
aDepartment of Genetics, Yale School of Medicine, New Haven, CT 06520;
bHoward Hughes Medical Institute, New Haven, CT 06520;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Christian C. D. Harman
Will Bailis
cDivision of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, PA 19104;
dDepartment of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Will Bailis
Jun Zhao
eDepartment of Immunobiology, Yale School of Medicine, New Haven, CT 06520;
fDepartment of Pathology, Yale School of Medicine, New Haven, CT 06510;
gProgram of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Louisa Hill
hResearch Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Louisa Hill
Rihao Qu
eDepartment of Immunobiology, Yale School of Medicine, New Haven, CT 06520;
fDepartment of Pathology, Yale School of Medicine, New Haven, CT 06510;
gProgram of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ruaidhrí P. Jackson
eDepartment of Immunobiology, Yale School of Medicine, New Haven, CT 06520;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Justin A. Shyer
eDepartment of Immunobiology, Yale School of Medicine, New Haven, CT 06520;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Holly R. Steach
eDepartment of Immunobiology, Yale School of Medicine, New Haven, CT 06520;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yuval Kluger
fDepartment of Pathology, Yale School of Medicine, New Haven, CT 06510;
gProgram of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520;
iApplied Mathematics Program, Yale University, New Haven, CT 06511;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Loyal A. Goff
jBroad Institute of MIT and Harvard, Cambridge, MA 02142;
kThe Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Loyal A. Goff
John L. Rinn
jBroad Institute of MIT and Harvard, Cambridge, MA 02142;
lDepartment of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138;
mDepartment of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115;
nDepartment of Biochemistry, University of Colorado, BioFrontiers Institute, Boulder, CO 80301;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for John L. Rinn
Adam Williams
oThe Jackson Laboratory for Genomic Medicine, Farmington, CT 06032;
pDepartment of Genetics and Genomic Sciences, University of Connecticut Health Center, Farmington, CT 06030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jorge Henao-Mejia
dDepartment of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Richard A. Flavell
bHoward Hughes Medical Institute, New Haven, CT 06520;
eDepartment of Immunobiology, Yale School of Medicine, New Haven, CT 06520;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: richard.flavell@yale.edu
  1. Contributed by Richard A. Flavell, December 7, 2020 (sent for review December 5, 2019; reviewed by Cornelis Murre and Ellen V. Rothenberg)

  • Article
  • Figures & SI
  • Info & Metrics
  • PDF
Loading

Significance

The development of lymphocytes is critical for host immunity and relies on a series of developmental checkpoints regulated by key transcription factors such as Ikaros. We hypothesized that nonprotein-coding loci might represent an additional layer of control in lymphocyte development. We identified a noncoding region (Daedalus) whose absence leads to a profound loss of Ikaros protein and a severe reduction in early lymphocyte progenitors. In contrast to Ikaros deletion, removal of Daedalus also led to an increase in red-blood-cell colony formation, suggesting that Daedalus functions as a lineage-specific stabilizer of Ikaros activity, thus acting as a “gatekeeper” of a newly identified lymphoid-erythroid checkpoint. This finding presents a paradigm potentially applicable to the control of all developmental programs.

Abstract

Haematopoiesis relies on tightly controlled gene expression patterns as development proceeds through a series of progenitors. While the regulation of hematopoietic development has been well studied, the role of noncoding elements in this critical process is a developing field. In particular, the discovery of new regulators of lymphopoiesis could have important implications for our understanding of the adaptive immune system and disease. Here we elucidate how a noncoding element is capable of regulating a broadly expressed transcription factor, Ikaros, in a lymphoid lineage-specific manner, such that it imbues Ikaros with the ability to specify the lymphoid lineage over alternate fates. Deletion of the Daedalus locus, which is proximal to Ikaros, led to a severe reduction in early lymphoid progenitors, exerting control over the earliest fate decisions during lymphoid lineage commitment. Daedalus locus deletion led to alterations in Ikaros isoform expression and a significant reduction in Ikaros protein. The Daedalus locus may function through direct DNA interaction as Hi-C analysis demonstrated an interaction between the two loci. Finally, we identify an Ikaros-regulated erythroid-lymphoid checkpoint that is governed by Daedalus in a lymphoid-lineage–specific manner. Daedalus appears to act as a gatekeeper of Ikaros’s broad lineage-specifying functions, selectively stabilizing Ikaros activity in the lymphoid lineage and permitting diversion to the erythroid fate in its absence. These findings represent a key illustration of how a transcription factor with broad lineage expression must work in concert with noncoding elements to orchestrate hematopoietic lineage commitment.

  • lymphocytes
  • Ikaros
  • noncoding
  • hematopoiesis

Footnotes

  • ↵1C.C.D.H. and W.B. contributed equally to this work.

  • ↵2To whom correspondence may be addressed. Email: richard.flavell{at}yale.edu.
  • Author contributions: C.C.D.H., W.B., R.P.J., L.A.G., J.L.R., A.W., J.H.-M., and R.A.F. designed research; R.A.F. supervised the research and edited the manuscript; C.C.D.H., W.B., L.H., R.P.J., J.A.S., H.R.S., L.A.G., J.L.R., A.W., and J.H.-M. performed research; J.Z., R.Q., and Y.K. contributed new reagents/analytic tools; C.C.D.H., W.B., J.Z., L.H., R.Q., R.P.J., J.A.S., H.R.S., Y.K., L.A.G., J.L.R., and R.A.F. analyzed data; and C.C.D.H. and W.B. wrote the paper.

  • Reviewers: C.M., University of California San Diego; and E.V.R., California Institute of Technology.

  • Competing interest statement: R.A.F. is a consultant for GSK and Zai Lab Ltd.

  • This article contains supporting information online at https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1918062118/-/DCSupplemental.

Data Availability.

All sequencing data for this study was uploaded to the Gene Expression Omnibus (GEO) repository under accession superseries GSE153879 unless otherwise specified. Hi-C sequencing data for this study is available at the GEO repository under the accession nos. GSM4350200 and GSM4350198 (in GSE140975) and was published in Hill et al. (52).

Published under the PNAS license.

View Full Text

References

  1. 1.↵
    1. T. Boehm
    , Design principles of adaptive immune systems. Nat. Rev. Immunol. 11, 307–317 (2011).
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. P. D. Greenberg,
    2. S. R. Riddell
    , Deficient cellular immunity: Finding and fixing the defects. Science 285, 546–551 (1999).
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    1. R. H. Buckley
    , Primary cellular immunodeficiencies. J. Allergy Clin. Immunol. 109, 747–757 (2002).
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. R. H. Buckley
    , Primary immunodeficiency diseases due to defects in lymphocytes. N. Engl. J. Med. 343, 1313–1324 (2000).
    OpenUrlCrossRefPubMed
  5. 5.↵
    1. C. Cobaleda,
    2. A. Schebesta,
    3. A. Delogu,
    4. M. Busslinger
    , Pax5: The guardian of B cell identity and function. Nat. Immunol. 8, 463–470 (2007).
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. J. Ramírez,
    2. K. Lukin,
    3. J. Hagman
    , From hematopoietic progenitors to B cells: Mechanisms of lineage restriction and commitment. Curr. Opin. Immunol. 22, 177–184 (2010).
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. M. Ye,
    2. T. Graf
    , Early decisions in lymphoid development. Curr. Opin. Immunol. 19, 123–128 (2007).
    OpenUrlCrossRefPubMed
  8. 8.↵
    1. D. Bryder,
    2. M. Sigvardsson
    , Shaping up a lineage: Lessons from B lymphopoesis. Curr. Opin. Immunol. 22, 148–153 (2010).
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. E. Deniz,
    2. B. Erman
    , Long noncoding RNA (lincRNA), a new paradigm in gene expression control. Funct. Integr. Genomics 17, 135–143 (2017).
    OpenUrl
  10. 10.↵
    1. J. D. Ransohoff,
    2. Y. Wei,
    3. P. A. Khavari
    , The functions and unique features of long intergenic non-coding RNA. Nat. Rev. Mol. Cell Biol. 19, 143–157 (2018).
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. J. R. Alvarez-Dominguez,
    2. H. F. Lodish
    , Emerging mechanisms of long noncoding RNA function during normal and malignant hematopoiesis. Blood 130, 1965–1975 (2017).
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    1. V. R. Paralkar,
    2. M. J. Weiss
    , Long noncoding RNAs in biology and hematopoiesis. Blood 121, 4842–4846 (2013).
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. A. T. Satpathy,
    2. H. Y. Chang
    , Long noncoding RNA in hematopoiesis and immunity. Immunity 42, 792–804 (2015).
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. E. V. Rothenberg
    , Transcriptional control of early T and B cell developmental choices. Annu. Rev. Immunol. 32, 283–321 (2014).
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. M. Guttman,
    2. J. L. Rinn
    , Modular regulatory principles of large non-coding RNAs. Nature 482, 339–346 (2012).
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. K. C. Wang,
    2. H. Y. Chang
    , Molecular mechanisms of long noncoding RNAs. Mol. Cell 43, 904–914 (2011).
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. P. E. Love,
    2. A. Bhandoola
    , Signal integration and crosstalk during thymocyte migration and emigration. Nat. Rev. Immunol. 11, 469–477 (2011).
    OpenUrlCrossRefPubMed
  18. 18.↵
    1. B. A. Schwarz et al
    ., Selective thymus settling regulated by cytokine and chemokine receptors. J. Immunol. 178, 2008–2017 (2007).
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    1. H. Schjerven et al
    ., Selective regulation of lymphopoiesis and leukemogenesis by individual zinc fingers of Ikaros. Nat. Immunol. 14, 1073–1083 (2013).
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. J. H. Wang et al
    ., Selective defects in the development of the fetal and adult lymphoid system in mice with an Ikaros null mutation. Immunity 5, 537–549 (1996).
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. K. Georgopoulos et al
    ., The Ikaros gene is required for the development of all lymphoid lineages. Cell 79, 143–156 (1994).
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. R. Marke,
    2. F. N. van Leeuwen,
    3. B. Scheijen
    , The many faces of IKZF1 in B-cell precursor acute lymphoblastic leukemia. Haematologica 103, 565–574 (2018).
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. S. Y. Ng,
    2. T. Yoshida,
    3. K. Georgopoulos
    , Ikaros and chromatin regulation in early hematopoiesis. Curr. Opin. Immunol. 19, 116–122 (2007).
    OpenUrlCrossRefPubMed
  24. 24.↵
    1. K. W. Tinsley et al
    ., Ikaros is required to survive positive selection and to maintain clonal diversity during T-cell development in the thymus. Blood 122, 2358–2368 (2013).
    OpenUrlAbstract/FREE Full Text
  25. 25.↵
    1. J. M. Alsiö,
    2. B. Tarchini,
    3. M. Cayouette,
    4. F. J. Livesey
    , Ikaros promotes early-born neuronal fates in the cerebral cortex. Proc. Natl. Acad. Sci. U.S.A. 110, E716–E725 (2013).
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    1. T. A. Schwickert et al
    ., Stage-specific control of early B cell development by the transcription factor Ikaros. Nat. Immunol. 15, 283–293 (2014).
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. M. Siatecka,
    2. J. J. Bieker
    , The multifunctional role of EKLF/KLF1 during erythropoiesis. Blood 118, 2044–2054 (2011).
    OpenUrlAbstract/FREE Full Text
  28. 28.↵
    1. L. Tumburu,
    2. S. L. Thein
    , Genetic control of erythropoiesis. Curr. Opin. Hematol. 24, 173–182 (2017).
    OpenUrl
  29. 29.↵
    1. M. Suzuki et al
    ., GATA factor switching from GATA2 to GATA1 contributes to erythroid differentiation. Genes Cells 18, 921–933 (2013).
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. S. Saleque,
    2. S. Cameron,
    3. S. H. Orkin
    , The zinc-finger proto-oncogene Gfi-1b is essential for development of the erythroid and megakaryocytic lineages. Genes Dev. 16, 301–306 (2002).
    OpenUrlAbstract/FREE Full Text
  31. 31.↵
    1. T. Yoshida,
    2. K. Georgopoulos
    , Ikaros fingers on lymphocyte differentiation. Int. J. Hematol. 100, 220–229 (2014).
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. J. Kim et al
    ., Ikaros DNA-binding proteins direct formation of chromatin remodeling complexes in lymphocytes. Immunity 10, 345–355 (1999).
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. J. D. Buenrostro,
    2. B. Wu,
    3. H. Y. Chang,
    4. W. J. Greenleaf
    , ATAC-seq: A method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. V. W. Yu,
    2. D. T. Scadden
    , Heterogeneity of the bone marrow niche. Curr. Opin. Hematol. 23, 331–338 (2016).
    OpenUrlCrossRefPubMed
  35. 35.↵
    1. A. Nichogiannopoulou,
    2. M. Trevisan,
    3. S. Neben,
    4. C. Friedrich,
    5. K. Georgopoulos
    , Defects in hemopoietic stem cell activity in Ikaros mutant mice. J. Exp. Med. 190, 1201–1214 (1999).
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    1. P. Papathanasiou et al
    ., Widespread failure of hematolymphoid differentiation caused by a recessive niche-filling allele of the Ikaros transcription factor. Immunity 19, 131–144 (2003).
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Y. Arinobu et al
    ., Reciprocal activation of GATA-1 and PU.1 marks initial specification of hematopoietic stem cells into myeloerythroid and myelolymphoid lineages. Cell Stem Cell 1, 416–427 (2007).
    OpenUrlCrossRefPubMed
  38. 38.↵
    1. C. Nerlov,
    2. E. Querfurth,
    3. H. Kulessa,
    4. T. Graf
    , GATA-1 interacts with the myeloid PU.1 transcription factor and represses PU.1-dependent transcription. Blood 95, 2543–2551 (2000).
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. N. Rekhtman,
    2. F. Radparvar,
    3. T. Evans,
    4. A. I. Skoultchi
    , Direct interaction of hematopoietic transcription factors PU.1 and GATA-1: Functional antagonism in erythroid cells. Genes Dev. 13, 1398–1411 (1999).
    OpenUrlAbstract/FREE Full Text
  40. 40.↵
    1. P. van Galen et al
    ., Reduced lymphoid lineage priming promotes human hematopoietic stem cell expansion. Cell Stem Cell 14, 94–106 (2014).
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. T. Yoshida,
    2. S. Y. Ng,
    3. K. Georgopoulos
    , Awakening lineage potential by Ikaros-mediated transcriptional priming. Curr. Opin. Immunol. 22, 154–160 (2010).
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. J. M. Engreitz et al
    ., Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature 539, 452–455 (2016).
    OpenUrlCrossRefPubMed
  43. 43.↵
    1. N. D. Tippens et al
    ., Transcription imparts architecture, function and logic to enhancer units. Nat. Genet. 52, 1067–1075 (2020).
    OpenUrl
  44. 44.↵
    1. H. Wang et al
    ., One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153, 910–918 (2013).
    OpenUrlCrossRefPubMed
  45. 45.↵
    1. B. Langmead,
    2. S. L. Salzberg
    , Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. H. Li et al.; 1000 Genome Project Data Processing Subgroup
    , The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).
    OpenUrlCrossRefPubMed
  47. 47.↵
    1. A. R. Quinlan,
    2. I. M. Hall
    , BEDTools: A flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).
    OpenUrlCrossRefPubMed
  48. 48.↵
    1. Y. Zhang et al
    ., Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).
    OpenUrlCrossRefPubMed
  49. 49.↵
    1. H. Thorvaldsdóttir,
    2. J. T. Robinson,
    3. J. P. Mesirov
    , Integrative genomics viewer (IGV): High-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).
    OpenUrlCrossRefPubMed
  50. 50.↵
    1. C. Trapnell et al
    ., Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).
    OpenUrlCrossRefPubMed
  51. 51.↵
    1. N. L. Bray,
    2. H. Pimentel,
    3. P. Melsted,
    4. L. Pachter
    , Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).
    OpenUrlCrossRefPubMed
  52. 52.↵
    1. L. Hill et al
    ., Wapl repression by Pax5 promotes V gene recombination by Igh loop extrusion. Nature 584, 142–147 (2020).
    OpenUrl
  53. 53.↵
    1. S. S. Rao et al
    ., A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).
    OpenUrlCrossRefPubMed
  54. 54.↵
    1. P. Bönelt et al
    ., Precocious expression of Blimp1 in B cells causes autoimmune disease with increased self-reactive plasma cells. EMBO J. 38, e100010 (2019).
    OpenUrlAbstract/FREE Full Text
  55. 55.↵
    1. A. Butler,
    2. P. Hoffman,
    3. P. Smibert,
    4. E. Papalexi,
    5. R. Satija
    , Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).
    OpenUrlCrossRefPubMed
  56. 56.↵
    1. G. C. Linderman,
    2. J. Zhao,
    3. Y. Kluger
    , Zero-preserving imputation of scRNA-seq data using low-rank approximation. bioRxiv [Preprint] (2020). http://doi.org/10.1101/397588 (Accessed 4 January 2021).

Log in using your username and password

Forgot your user name or password?

Log in through your institution

You may be able to gain access using your login credentials for your institution. Contact your library if you do not have a username and password.
If your organization uses OpenAthens, you can log in using your OpenAthens username and password. To check if your institution is supported, please see this list. Contact your library for more details.

Purchase access

You may purchase access to this article. This will require you to create an account if you don't already have one.

Subscribers, for more details, please visit our Subscriptions FAQ.

Please click here to log into the PNAS submission website.

PreviousNext
Back to top
Article Alerts
Email Article

Thank you for your interest in spreading the word on PNAS.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
An in vivo screen of noncoding loci reveals that Daedalus is a gatekeeper of an Ikaros-dependent checkpoint during haematopoiesis
(Your Name) has sent you a message from PNAS
(Your Name) thought you would like to see the PNAS web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
An in vivo screen of noncoding loci reveals that Daedalus is a gatekeeper of an Ikaros-dependent checkpoint during haematopoiesis
Christian C. D. Harman, Will Bailis, Jun Zhao, Louisa Hill, Rihao Qu, Ruaidhrí P. Jackson, Justin A. Shyer, Holly R. Steach, Yuval Kluger, Loyal A. Goff, John L. Rinn, Adam Williams, Jorge Henao-Mejia, Richard A. Flavell
Proceedings of the National Academy of Sciences Jan 2021, 118 (3) e1918062118; DOI: 10.1073/pnas.1918062118

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Request Permissions
Share
An in vivo screen of noncoding loci reveals that Daedalus is a gatekeeper of an Ikaros-dependent checkpoint during haematopoiesis
Christian C. D. Harman, Will Bailis, Jun Zhao, Louisa Hill, Rihao Qu, Ruaidhrí P. Jackson, Justin A. Shyer, Holly R. Steach, Yuval Kluger, Loyal A. Goff, John L. Rinn, Adam Williams, Jorge Henao-Mejia, Richard A. Flavell
Proceedings of the National Academy of Sciences Jan 2021, 118 (3) e1918062118; DOI: 10.1073/pnas.1918062118
Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Mendeley logo Mendeley
Proceedings of the National Academy of Sciences: 118 (3)
Table of Contents

Submit

Sign up for Article Alerts

Article Classifications

  • Biological Sciences
  • Immunology and Inflammation

Jump to section

  • Article
    • Abstract
    • Results
    • Discussion
    • Materials and Methods
    • Data Availability.
    • Acknowledgments
    • Footnotes
    • References
  • Figures & SI
  • Info & Metrics
  • PDF

You May Also be Interested in

Surgeons hands during surgery
Inner Workings: Advances in infectious disease treatment promise to expand the pool of donor organs
Despite myriad challenges, clinicians see room for progress.
Image credit: Shutterstock/David Tadevosian.
Setting sun over a sun-baked dirt landscape
Core Concept: Popular integrated assessment climate policy models have key caveats
Better explicating the strengths and shortcomings of these models will help refine projections and improve transparency in the years ahead.
Image credit: Witsawat.S.
Double helix
Journal Club: Noncoding DNA shown to underlie function, cause limb malformations
Using CRISPR, researchers showed that a region some used to label “junk DNA” has a major role in a rare genetic disorder.
Image credit: Nathan Devery.
Steamboat Geyser eruption.
Eruption of Steamboat Geyser
Mara Reed and Michael Manga explore why Yellowstone's Steamboat Geyser resumed erupting in 2018.
Listen
Past PodcastsSubscribe
Multi-color molecular model
Enzymatic breakdown of PET plastic
A study demonstrates how two enzymes—MHETase and PETase—work synergistically to depolymerize the plastic pollutant PET.
Image credit: Aaron McGeehan (artist).

Similar Articles

Site Logo
Powered by HighWire
  • Submit Manuscript
  • Twitter
  • Facebook
  • RSS Feeds
  • Email Alerts

Articles

  • Current Issue
  • Special Feature Articles – Most Recent
  • List of Issues

PNAS Portals

  • Anthropology
  • Chemistry
  • Classics
  • Front Matter
  • Physics
  • Sustainability Science
  • Teaching Resources

Information

  • Authors
  • Editorial Board
  • Reviewers
  • Librarians
  • Press
  • Site Map
  • PNAS Updates

Feedback    Privacy/Legal

Copyright © 2021 National Academy of Sciences. Online ISSN 1091-6490