Your browser doesn't support javascript.
loading
A distant trophoblast-specific enhancer controls HLA-G expression at the maternal-fetal interface.
Ferreira, Leonardo M R; Meissner, Torsten B; Mikkelsen, Tarjei S; Mallard, William; O'Donnell, Charles W; Tilburgs, Tamara; Gomes, Hannah A B; Camahort, Raymond; Sherwood, Richard I; Gifford, David K; Rinn, John L; Cowan, Chad A; Strominger, Jack L.
Afiliación
  • Ferreira LM; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138; leonardoferreira@fas.harvard.edu jlstrom@fas.harvard.edu.
  • Meissner TB; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138;
  • Mikkelsen TS; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138; The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138;
  • Mallard W; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138; The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142;
  • O'Donnell CW; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138;
  • Tilburgs T; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138;
  • Gomes HA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138;
  • Camahort R; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138;
  • Sherwood RI; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115;
  • Gifford DK; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139;
  • Rinn JL; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138; The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142; Harvard Stem Cell Institute
  • Cowan CA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138; The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138;
  • Strominger JL; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138; leonardoferreira@fas.harvard.edu jlstrom@fas.harvard.edu.
Proc Natl Acad Sci U S A ; 113(19): 5364-9, 2016 May 10.
Article en En | MEDLINE | ID: mdl-27078102
ABSTRACT
HLA-G, a nonclassical HLA molecule uniquely expressed in the placenta, is a central component of fetus-induced immune tolerance during pregnancy. The tissue-specific expression of HLA-G, however, remains poorly understood. Here, systematic interrogation of the HLA-G locus using massively parallel reporter assay (MPRA) uncovered a previously unidentified cis-regulatory element 12 kb upstream of HLA-G with enhancer activity, Enhancer L Strikingly, clustered regularly-interspaced short palindromic repeats (CRISPR)/Cas9-mediated deletion of this enhancer resulted in ablation of HLA-G expression in JEG3 cells and in primary human trophoblasts isolated from placenta. RNA-seq analysis demonstrated that Enhancer L specifically controls HLA-G expression. Moreover, DNase-seq and chromatin conformation capture (3C) defined Enhancer L as a cell type-specific enhancer that loops into the HLA-G promoter. Interestingly, MPRA-based saturation mutagenesis of Enhancer L identified motifs for transcription factors of the CEBP and GATA families essential for placentation. These factors associate with Enhancer L and regulate HLA-G expression. Our findings identify long-range chromatin looping mediated by core trophoblast transcription factors as the mechanism controlling tissue-specific HLA-G expression at the maternal-fetal interface. More broadly, these results establish the combination of MPRA and CRISPR/Cas9 deletion as a powerful strategy to investigate human immune gene regulation.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Trofoblastos / Embarazo / Elementos de Facilitación Genéticos / Regulación del Desarrollo de la Expresión Génica / Histocompatibilidad Materno-Fetal / Antígenos HLA-G / Intercambio Materno-Fetal Tipo de estudio: Prognostic_studies Límite: Female / Humans Idioma: En Revista: Proc Natl Acad Sci U S A Año: 2016 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Trofoblastos / Embarazo / Elementos de Facilitación Genéticos / Regulación del Desarrollo de la Expresión Génica / Histocompatibilidad Materno-Fetal / Antígenos HLA-G / Intercambio Materno-Fetal Tipo de estudio: Prognostic_studies Límite: Female / Humans Idioma: En Revista: Proc Natl Acad Sci U S A Año: 2016 Tipo del documento: Article