Faculty Opinions recommendation of Crosstalk between C/EBPbeta phosphorylation, arginine methylation, and SWI/SNF/Mediator implies an indexing transcription factor code.

Author(s):  
Martha Bulyk
2010 ◽  
Vol 29 (6) ◽  
pp. 1105-1115 ◽  
Author(s):  
Elisabeth Kowenz-Leutz ◽  
Ole Pless ◽  
Gunnar Dittmar ◽  
Maria Knoblich ◽  
Achim Leutz

2021 ◽  
Vol 70 ◽  
pp. 89-96
Author(s):  
Weixian Deng ◽  
Elsie C Jacobson ◽  
Amanda J Collier ◽  
Kathrin Plath

2020 ◽  
Author(s):  
Evelyn Ramberger ◽  
Valeria Sapozhnikova ◽  
Elisabeth Kowenz-Leutz ◽  
Karin Zimmermann ◽  
Nathalie Nicot ◽  
...  

AbstractThe pioneering transcription factor C/EBPα coordinates cell fate and cell differentiation. C/EBPα represents an intrinsically disordered protein with multiple short linear motifs and extensive post-translational side chain modifications (PTM), reflecting its modularity and functional plasticity. Here, we combined arrayed peptide matrix screening (PRISMA) with biotin ligase proximity labeling proteomics (BioID) to generate a linear, isoform specific and PTM-dependent protein interaction map of C/EBPα in myeloid cells. The C/EBPα interactome comprises promiscuous and PTM-regulated interactions with protein machineries involved in gene expression, epigenetics, genome organization, DNA replication, RNA processing, and nuclear transport as the basis of functional C/EBPα plasticity. Protein interaction hotspots were identified that coincide with homologous conserved regions of the C/EBP family and revealed interaction motifs that score as molecular recognition features (MoRF). PTMs alter the interaction spectrum of multi-valent C/EBP-motifs to configure a multimodal transcription factor hub that allows interaction with multiple co-regulatory components, including BAF/SWI-SNF or Mediator complexes. Combining PRISMA and BioID acts as a powerful strategy to systematically explore the interactomes of intrinsically disordered proteins and their PTM-regulated, multimodal capacity.Key pointsIntegration of proximity labeling and arrayed peptide screen proteomics refines the interactome of C/EBPα isoformsHotspots of protein interactions in C/EBPα mostly occur in conserved short linear motifsInteractions of the BAF/SWI-SNF complex with C/EBPα are modulated by arginine methylation and isoform statusThe integrated experimental strategy suits systematic interactome studies of intrinsically disordered proteins


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3403-3403
Author(s):  
Xinyang Zhao ◽  
Ly P. Vu ◽  
Fabiana Perna ◽  
Fan Liu ◽  
Hao Xu ◽  
...  

Abstract Abstract 3403 RUNX1 is a transcription factor that is required for definitive hematopoietic development, and helps regulate long term hematopoietic stem cell self-renewal, platelet production, and lymphocyte development during adult hematopoiesis. RUNX1 is known to be modified via phosphorylation, acetylation, ubiquitination and methylation, for example on R208 and R210 by PRMT1, which activates its activating function. We continue to investigate how the methylation of RUNX1 by other protein arginine methyl transferases (PRMTs) regulates its function. Loop 9 of the DNA binding domain (the Runt domain) of RUNX1 contains an SGRGK sequence that is also present on the tails of histone H2A and H4. The histone tails of H4 and H2A can be methylated by a purified PRMT5 complex in vitro. An enzymatically active in vitro PRMT5 complex capable of methylating histones and SM proteins requires two subunits: both PRMT5 and MEP50, a WD 40 repeat domain protein. Nevertheless, this purified PRMT5/MEP50 complex cannot methylate the DNA binding domain of the RUNX1 protein in vitro. We show that RUNX1 also can be symmetrically methylated at R142 within the SGRGK motif in vitro by a nuclear PRMT5/MEP50 complex which also contains COPR5. We show after RUNX1 is methylated on R142 within the nucleus of HEL cells, RUNX1 is exported to the cytoplasm in a CRM1 dependent manner, as the export of methylated RUNX1 is blocked by lemptomycin B. CRM1 interacts with PRMT5, supporting that PRMT5 mediated arginine methylation tags protein for nuclear export. Therefore, PRMT5 not only involves in epigenetic regulation by methylation of histones but also it can directly controls the level of transcription factor proteins within the nucleus. Polycytocemia Vera patients who express the Jak2V617F mutation have low PRMT5 activity due to JAK2V617F mediated PRMT5 phosphorylation (Liu et al 2011). How Jak2 signaling affects RUNX1 methylation and RUNX1 localization within the nucleus is still under investigation. By controlling the amount of RUNX1 available within the cell nucleus, PRMT5 may regulate lineage differentiation potential and growth potential of hematopoietic stem and progenitor cells. The nuclear localization of RUNX1 can be changed through post translational modification such as arginine methylation in addition to point mutations and translocations involving RUNX1 found patients with leukemia and pre-leukemic diseases. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1309-1309
Author(s):  
Shinsuke Mizutani ◽  
Masafumi Taniwaki ◽  
Tsukasa Okuda

Abstract Abstract 1309 Acute Myeloid Leukemia 1 (AML1; also called as Runx1: Runt-related transcription factor 1) belongs to the Class II group of leukemia-associated mutation-target genes, and it encodes the DNA-binding subunit of the hetero-dimeric transcription factor complex, Core-Binding Factor (CBF). CBF plays pivotal roles in initial hematopoietic development during embryogenesis and in cellular differentiation of thrombotic and lymphatic lineages throughout adult life. Recent researches revealed that cellular AML1 polypeptide is processed with post-translational modifications, including phosphorylation, acethylation, ubiquitination, and methylation. Biological significance of these modifications on the AML1's function as the hematopoietic regulator, however, largely remains to be elucidated. In this study, we focused on the arginine-methylation as an initial step towards the comprehensive understanding for the AML1-regulating mechanism through these modifications. Arginine residues just downstream to the Runt-domain, which is located at N-terminal region of the molecule and functions as the binding site to DNA and CBF beta: the hetero-dimerization partner, are recently reported to be methylated, resulting in the inhibition of the corepressor-binding thus enhancing its trans- activating activity. In order to elucidate biological significance of these methylations, we performed a series of genetic experiments: First, we generated the non-methylatable double arginine-to-lysine (RRKK) mutant of AML1 at these residues, which should keep AML1-corepressor-binding. When this mutant was subjected to the luciferase reporter-assay using a target-gene construct, it showed lower trans- activating activity in comparison to that for wild-type molecule, as expected. However, this loss-of-function mutation appeared to be dispensable at least for in vitro function for hematopoietic regulation in that this RRKK mutant did rescue hematopoietic differentiation of the AML1-deficient murine ES cells in culture when expressed from a knock-in allele as was the case for the wild-type cDNA of mouse AML1. To further evaluate the biological activity of this mutant in the context of an entire animal, we introduced this mutant cDNA into AML1/Runx1 locus of mouse ES cells by means of a targeted-insertion (knock-in) strategy. Germline mutant mouse lines were successfully established, following blastocyst-injection of these ES cell clones. Heterozygous mice were healthy and fertile, and genotyping for the live pups generated from heterozygotes-crossing revealed that this arginine-mutant allele segregated according to the Mendelian ratio. Homozygous AML1RRKK/RRKK mice were born alive and grew up adult, circumventing the mid-embryonic death due to hematopoietic block that was originally described for the AML1-deficient mice, thus the in vitro notion that these arginine-methylations were not essential for the early hematopoietic development described above was further underscored. There were no significant differences so far observed in peripheral blood cell counts among mice of the AML1RRKK/RRKK or AML1WT/WT genotypes, in comparison to their wild-type littermates. Preliminary studies revealed that AML1RRKK/RRKK mice showed imbalance of the peripheral T cell populations, implying that these methylations may have roles in these cellular lineages. We are currently focusing on further examination of these mutant mice, paying special attention to the cellular lineages where genetic manifestations were observed for AML1 haploinsufficient mice and/or conditional AML1-deficient mice. We hope that these efforts will unveil the biological significance of the AML1 methylation in hematopoietic regulation. Disclosures: No relevant conflicts of interest to declare.


Cell ◽  
2003 ◽  
Vol 113 (1) ◽  
pp. 73-86 ◽  
Author(s):  
Douglas W. Allan ◽  
Susan E.St. Pierre ◽  
Irene Miguel-Aliaga ◽  
Stefan Thor

Cell Reports ◽  
2019 ◽  
Vol 29 (12) ◽  
pp. 4172-4185.e5 ◽  
Author(s):  
Xingting Guo ◽  
Chang Yin ◽  
Fu Yang ◽  
Yongchao Zhang ◽  
Huanwei Huang ◽  
...  

PLoS ONE ◽  
2013 ◽  
Vol 8 (11) ◽  
pp. e77928 ◽  
Author(s):  
Marta Garcia Del Barrio ◽  
Steeve Bourane ◽  
Katja Grossmann ◽  
Roland Schüle ◽  
Stefan Britsch ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document