U.S. flag

An official website of the United States government

Format

Send to:

Choose Destination
    • Showing Current items.

    Prdm14 PR domain containing 14 [ Mus musculus (house mouse) ]

    Gene ID: 383491, updated on 6-Oct-2024

    GeneRIFs: Gene References Into Functions

    GeneRIFPubMed TitleDate
    Interplay between CTCF boundaries and a super enhancer controls cohesin extrusion trajectories and gene expression.

    Interplay between CTCF boundaries and a super enhancer controls cohesin extrusion trajectories and gene expression.
    Vos ESM, Valdes-Quezada C, Huang Y, Allahyar A, Verstegen MJAM, Felder AK, van der Vegt F, Uijttewaal ECH, Krijger PHL, de Laat W.

    08/28/2021
    Blastocyst complementation using Prdm14-deficient rats enables efficient germline transmission and generation of functional mouse spermatids in rats.

    Blastocyst complementation using Prdm14-deficient rats enables efficient germline transmission and generation of functional mouse spermatids in rats.
    Kobayashi T, Goto T, Oikawa M, Sanbo M, Yoshida F, Terada R, Niizeki N, Kajitani N, Kazuki K, Kazuki Y, Hochi S, Nakauchi H, Surani MA, Hirabayashi M., Free PMC Article

    03/20/2021
    the crucial role of Prdm14 in primordial germ cell specification is conserved between rat and mice

    Germline development in rat revealed by visualization and deletion of Prdm14.
    Kobayashi T, Kobayashi H, Goto T, Takashima T, Oikawa M, Ikeda H, Terada R, Yoshida F, Sanbo M, Nakauchi H, Kurimoto K, Hirabayashi M.

    08/15/2020
    Authors find that global upregulation of the repressive histone H3 lysine 27 trimethylation (H3K27me3) mark is PRDM14 dosage dependent in PGCs of both sexes. When focusing on XCR, we observed that PRDM14 is required for removal of H3K27me3 from the inactive X-chromosome, which, in contrast to global upregulation, takes place progressively along the PGC migration path.

    PRDM14 controls X-chromosomal and global epigenetic reprogramming of H3K27me3 in migrating mouse primordial germ cells.
    Mallol A, Guirola M, Payer B., Free PMC Article

    06/20/2020
    The pluripotency regulator PRDM14 requires the master hematopoietic regulator CBFA2T3 to initiate leukemia in progenitor cells, demonstrating an oncogenic role for CBFA2T3 and providing an avenue for targeting cancer-initiating cells.

    The Pluripotency Regulator PRDM14 Requires Hematopoietic Regulator CBFA2T3 to Initiate Leukemia in Mice.
    Tracey LJ, Brooke-Bisschop T, Jansen PWTC, Campos EI, Vermeulen M, Justice MJ., Free PMC Article

    05/2/2020
    the co-option of the PRDM14-CBFA2T complex from motor neurons into pluripotent cells may have maintained the transcriptional network for pluripotency during vertebrate evolution.

    Co-option of the PRDM14-CBFA2T complex from motor neurons to pluripotent cells during vertebrate evolution.
    Kawaguchi M, Sugiyama K, Matsubara K, Lin CY, Kuraku S, Hashimoto S, Suwa Y, Yong LW, Takino K, Higashida S, Kawamura D, Yu JK, Seki Y.

    09/14/2019
    we provide evidence that HOXA1 displays an unexpectedly long half-life and demonstrate that PRDM14 can reduce the stability and affect the transcriptional activity of HOXA1.

    PRDM14, a putative histone methyl-transferase, interacts with and decreases the stability and activity of the HOXA1 transcription factor.
    Draime A, Bridoux L, Belpaire M, Pringels T, Tys J, Rezsohazy R.

    07/14/2018
    Our results suggest that PRDM14 serves as an initial trigger for epigenetic changes and hyperdynamic plasticity in tumors via bivalent chromatin domains.

    Silencing PRDM14 expression by an innovative RNAi therapy inhibits stemness, tumorigenicity, and metastasis of breast cancer.
    Taniguchi H, Hoshino D, Moriya C, Zembutsu H, Nishiyama N, Yamamoto H, Kataoka K, Imai K., Free PMC Article

    04/28/2018
    PRDM14 recruited OCT3/4 to the enhancer regions of naive pluripotency genes via TET-base excision repair-mediated demethylation.

    PRDM14 Drives OCT3/4 Recruitment via Active Demethylation in the Transition from Primed to Naive Pluripotency.
    Okashita N, Suwa Y, Nishimura O, Sakashita N, Kadota M, Nagamatsu G, Kawaguchi M, Kashida H, Nakajima A, Tachibana M, Seki Y., Free PMC Article

    11/18/2017
    JARID2 physically interacts with ESRRB, SALL4A, and PRDM14

    Combined Overexpression of JARID2, PRDM14, ESRRB, and SALL4A Dramatically Improves Efficiency and Kinetics of Reprogramming to Induced Pluripotent Stem Cells.
    Iseki H, Nakachi Y, Hishida T, Yamashita-Sugahara Y, Hirasaki M, Ueda A, Tanimoto Y, Iijima S, Sugiyama F, Yagami K, Takahashi S, Okuda A, Okazaki Y.

    11/5/2016
    This repressive function is mediated through an ETO-family co-repressor Mtgr1, which tightly binds to the pre-SET/SET domains of Prdm14 and co-occupies its genomic targets in mouse embryonic stem cells.

    ETO family protein Mtgr1 mediates Prdm14 functions in stem cell maintenance and primordial germ cell formation.
    Nady N, Gupta A, Ma Z, Swigut T, Koide A, Koide S, Wysocka J., Free PMC Article

    10/8/2016
    the H3K27-specific demethylase Utx regulates the expression of the master regulators for XCI and XCR: Prdm14, Tsix, and Xist.

    Histone demethylation maintains Prdm14 and Tsix expression and represses xIst in embryonic stem cells.
    Kamikawa YF, Donohoe ME., Free PMC Article

    02/13/2016
    Both TET1 and TET2 are required for the repression of embryonic stem cells differentiation by PRDM14.

    PRDM14 maintains pluripotency of embryonic stem cells through TET-mediated active DNA demethylation.
    Okashita N, Sakashita N, Ito K, Mitsuya A, Suwa Y, Seki Y.

    12/19/2015
    Upregulation of Prdm14 results in a highly homogeneous population of authentic pluripotent colonies and prevents the abnormal silencing of the Dlk1-Dio3 locus.

    Ground-state conditions promote robust Prdm14 reactivation and maintain an active Dlk1-Dio3 region during reprogramming.
    Habib O, Habib G, Moon SH, Hong KS, Do JT, Choi Y, Chang SW, Chung HM., Free PMC Article

    12/6/2014
    PRDM14 is heterogeneously expressed in 4-cell-stage embryos. Forced expression of PRDM14 at the 2-cell stage leads to increased H3R26me2 and can induce a pluripotent ICM fate.

    Single-cell profiling of epigenetic modifiers identifies PRDM14 as an inducer of cell fate in the mammalian embryo.
    Burton A, Muller J, Tu S, Padilla-Longoria P, Guccione E, Torres-Padilla ME.

    08/30/2014
    PRDM14-induced leukemic cells contain high levels of activated NOTCH1 and downstream NOTCH1 targets, including MYC and HES1, and are sensitive to pharmacological inhibition of NOTCH1 with the gamma-secretase inhibitor DAPT

    A mouse model for inducible overexpression of Prdm14 results in rapid-onset and highly penetrant T-cell acute lymphoblastic leukemia (T-ALL).
    Carofino BL, Ayanga B, Justice MJ., Free PMC Article

    06/21/2014
    PRDM14 physically interacts with TET1 and TET2 and enhances the recruitment of TET1 and TET2 at target loci. Knockdown of TET1 and TET2 impaired transcriptional regulation and DNA demethylation by PRDM14.

    PRDM14 promotes active DNA demethylation through the ten-eleven translocation (TET)-mediated base excision repair pathway in embryonic stem cells.
    Okashita N, Kumaki Y, Ebi K, Nishi M, Okamoto Y, Nakayama M, Hashimoto S, Nakamura T, Sugasawa K, Kojima N, Takada T, Okano M, Seki Y.

    03/1/2014
    Tsix and PRDM14 directly link X-chromosome reactivation to stem cell pluripotency.

    Tsix RNA and the germline factor, PRDM14, link X reactivation and stem cell reprogramming.
    Payer B, Rosenberg M, Yamaji M, Yabuta Y, Koyanagi-Aoi M, Hayashi K, Yamanaka S, Saitou M, Lee JT., Free PMC Article

    02/22/2014
    Prdm14 promotes germline fate and naive pluripotency by repressing FGF signalling and DNA methylation.

    Prdm14 promotes germline fate and naive pluripotency by repressing FGF signalling and DNA methylation.
    Grabole N, Tischler J, Hackett JA, Kim S, Tang F, Leitch HG, Magnúsdóttir E, Surani MA., Free PMC Article

    11/2/2013
    PRDM14, ensures naive pluripotency through antagonizing activation of the FGFR signaling by the core pluripotency transcriptional circuitry, and repressing expression of DNA methyltransferases that modify the epigenome to a primed epiblast-like state.

    PRDM14 ensures naive pluripotency through dual regulation of signaling and epigenetic pathways in mouse embryonic stem cells.
    Yamaji M, Ueda J, Hayashi K, Ohta H, Yabuta Y, Kurimoto K, Nakato R, Yamada Y, Shirahige K, Saitou M.

    09/7/2013
    data provide the first direct evidence for the association of Prdm14 with cancer initiation in an in vivo mouse model and in human lymphoid malignancies, while suggesting mechanisms for Prdm14's mode of action

    Prdm14 initiates lymphoblastic leukemia after expanding a population of cells resembling common lymphoid progenitors.
    Dettman EJ, Simko SJ, Ayanga B, Carofino BL, Margolin JF, Morse HC 3rd, Justice MJ., Free PMC Article

    09/10/2011
    Prdm14 safeguards mouse embryonic stem cells maintenance by preventing induction of extraembryonic endoderm fates.

    Sequence-specific regulator Prdm14 safeguards mouse ESCs from entering extraembryonic endoderm fates.
    Ma Z, Swigut T, Valouev A, Rada-Iglesias A, Wysocka J.

    04/2/2011
    study implicates Prdm14 as a proto-oncogene involved in lymphoblastic lymphoma formation

    The zinc finger SET domain gene Prdm14 is overexpressed in lymphoblastic lymphomas with retroviral insertions at Evi32.
    Dettman EJ, Justice MJ., Free PMC Article

    01/21/2010
    launch of the germ cell lineage in mice is orchestrated by Blimp1 and Prdm14

    Specification of the germ cell lineage in mice: a process orchestrated by the PR-domain proteins, Blimp1 and Prdm14.
    Kurimoto K, Yamaji M, Seki Y, Saitou M.

    01/21/2010
    Study provides genetic evidence that Prdm14, with exclusive expression in the germ cell lineage and pluripotent cell lines, is critical for the reacquisition of potential pluripotency and successful epigenetic reprogramming.

    Critical function of Prdm14 for the establishment of the germ cell lineage in mice.
    Yamaji M, Seki Y, Kurimoto K, Yabuta Y, Yuasa M, Shigeta M, Yamanaka K, Ohinata Y, Saitou M.

    01/21/2010
    firstprevious page of 1 nextlast