scholarly journals Involvement of the NFX1-repressor complex in PKC-δ-induced repression of hTERT transcription

2016 ◽  
Vol 160 (5) ◽  
pp. 309-313 ◽  
Author(s):  
Shuntaro Yamashita ◽  
Kaoru Fujii ◽  
Chong Zhao ◽  
Hiroshi Takagi ◽  
Yoshinori Katakura
2009 ◽  
Vol 83 (9) ◽  
pp. 4376-4385 ◽  
Author(s):  
Haidong Gu ◽  
Bernard Roizman

ABSTRACT Among the early events in herpes simplex virus 1 replication are localization of ICP0 in ND10 bodies and accumulation of viral DNA-protein complexes in structures abutting ND10. ICP0 degrades components of ND10 and blocks silencing of viral DNA, achieving the latter by dislodging HDAC1 or -2 from the lysine-specific demethylase 1 (LSD1)/CoREST/REST repressor complex. The role of this process is apparent from the observation that a dominant-negative CoREST protein compensates for the absence of ICP0 in a cell-dependent fashion. HDAC1 or -2 and the CoREST/REST complex are independently translocated to the nucleus once viral DNA synthesis begins. The focus of this report is twofold. First, we report that in infected cells, LSD1, a key component of the repressor complex, is partially degraded or remains stably associated with CoREST and is ultimately also translocated, in part, to the cytoplasm. Second, we examined the distribution of the components of the repressor complex and ICP8 early in infection in wild-type-virus- and ICP0 mutant virus-infected cells. The repressor component and ultimately ICP8 localize in structures that abut the ND10 nuclear bodies. There is no evidence that the two compartments fuse. We propose that ICP0 must dynamically interact with both compartments in order to accomplish its functions of degrading PML and SP100 and suppressing silencing of viral DNA through its interactions with CoREST. In turn, the remodeling of the viral DNA-protein complex enables recruitment of ICP8 and initiation of formation of replication compartments.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Helena R. R. Wells ◽  
Fatin N. Zainul Abidin ◽  
Maxim B. Freidin ◽  
Frances M. K. Williams ◽  
Sally J. Dawson

AbstractTinnitus is a prevalent condition in which perception of sound occurs without an external stimulus. It is often associated with pre-existing hearing loss or noise-induced damage to the auditory system. In some individuals it occurs frequently or even continuously and leads to considerable distress and difficulty sleeping. There is little knowledge of the molecular mechanisms involved in tinnitus which has hindered the development of treatments. Evidence suggests that tinnitus has a heritable component although previous genetic studies have not established specific risk factors. From a total of 172,608 UK Biobank participants who answered questions on tinnitus we performed a case–control genome-wide association study for self-reported tinnitus. Final sample size used in association analysis was N = 91,424. Three variants in close proximity to the RCOR1 gene reached genome wide significance: rs4906228 (p = 1.7E−08), rs4900545 (p = 1.8E−08) and 14:103042287_CT_C (p = 3.50E−08). RCOR1 encodes REST Corepressor 1, a component of a co-repressor complex involved in repressing neuronal gene expression in non-neuronal cells. Eleven other independent genetic loci reached a suggestive significance threshold of p < 1E−06.


2020 ◽  
Vol 94 (22) ◽  
Author(s):  
Yonggang Pei ◽  
Josiah Hiu-yuen Wong ◽  
Hem Chandra Jha ◽  
Tian Tian ◽  
Zhi Wei ◽  
...  

ABSTRACT Epstein-Barr virus (EBV) was discovered as the first human tumor virus more than 50 years ago. EBV infects more than 90% of the human population worldwide and is associated with numerous hematologic malignancies and epithelial malignancies. EBV establishes latent infection in B cells, which is the typical program seen in lymphomagenesis. Understanding EBV-mediated transcription regulatory networks is one of the current challenges that will uncover new insights into the mechanism of viral-mediated lymphomagenesis. Here, we describe the regulatory profiles of several cellular factors (E2F6, E2F1, Rb, HDAC1, and HDAC2) together with EBV latent nuclear antigens using next-generation sequencing (NGS) analysis. Our results show that the E2F-Rb-HDAC complex exhibits similar distributions in genomic regions of EBV-positive cells and is associated with oncogenic super-enhancers involving long-range regulatory regions. Furthermore, EBV latent antigens cooperatively hijack this complex to bind at KLFs gene loci and facilitate KLF14 gene expression in lymphoblastoid cell lines (LCLs). These results demonstrate that EBV latent antigens can function as master regulators of this multisubunit repressor complex (E2F-Rb-HDAC) to reverse its suppressive activities and facilitate downstream gene expression that can contribute to viral-induced lymphomagenesis. These results provide novel insights into targets for the development of new therapeutic interventions for treating EBV-associated lymphomas. IMPORTANCE Epstein-Barr virus (EBV), as the first human tumor virus, infects more than 90% of the human population worldwide and is associated with numerous human cancers. Exploring EBV-mediated transcription regulatory networks is critical to understand viral-associated lymphomagenesis. However, the detailed mechanism is not fully explored. Now we describe the regulatory profiles of the E2F-Rb-HDAC complex together with EBV latent antigens, and we found that EBV latent antigens cooperatively facilitate KLF14 expression by antagonizing this multisubunit repressor complex in EBV-positive cells. This provides potential therapeutic targets for the treatment of EBV-associated cancers.


Genetics ◽  
1991 ◽  
Vol 128 (1) ◽  
pp. 29-35
Author(s):  
D N Arvidson ◽  
M Shapiro ◽  
P Youderian

Abstract The Escherichia coli trpR gene encodes tryptophan aporepressor, which binds the corepressor ligand, L-tryptophan, to form an active repressor complex. The side chain of residue valine 58 of Trp aporepressor sits at the bottom of the corepressor (L-tryptophan) binding pocket. Mutant trpR genes encoding changes of Val58 to the other 19 naturally occurring amino acids were made. Each of the mutant proteins requires a higher intracellular concentration of tryptophan for activation of DNA binding than wild-type aporepressor. Whereas wild-type aporepressor is activated better by 5-methyltryptophan (5-MT) than by tryptophan, Ile58 and other mutant aporepressors prefer tryptophan to 5-MT as corepressor, and Ala58 and Gly58 prefer 5-MT much more strongly than wild-type aporepressor in vivo. These mutant aporepressors are the first examples of DNA-binding proteins with altered specificities of cofactor recognition.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3759-3759
Author(s):  
Jaesung Seo ◽  
Li Li ◽  
Donald Small

Mutations of DNMT3A are one the most frequently observed alterations in AML patients. The DNMT3A R882 mutation appears to confer a dominant-negative loss-of function effect and changes the DNA binding preference according to recent studies. DNMT3A R882 mutations are also found at increasing frequency with age in healthy elderly populations and are one of the earliest pre-malignant alterations in the clonal evolution progression to leukemia. Studies have shown that DNMT3A mutations decrease overall DNA methylation and through less clear mechanisms, also impact the epigenetic landscape by effecting changes in histone acetylation. Here we investigated potential mechanisms by which loss of DNMT3A activity changes histone acetylation. DNMT3A binds with many proteins that regulate chromatin biology and gene transcription. Among those interacting proteins, we focused on the DNMT3A-HDAC interaction and their regulation of target gene suppression. To investigate whether there are differences in binding of wild-type vs. mutant DNMT3A to HDACs, we performed immunoprecipitation and Western blotting assays using Myc- tagged wild-type and R882 mutated DNMT3A. We found that DNMT3A R882 mutants showed reduced interaction with HDAC1 and 2. In addition, upon treatment with HDAC inhibitors (HDACi), DNMT3A mutant protein was more easily dissociated from HDAC1/2 than was wild-type DNMT3A. Intriguingly, covalent modification of DNMT3A R882 by SUMO1 protein was significantly enhanced relative to wild type DNMT3A. Together, we suggest that the weak complex formation between mutant DNMT3A and HDACs results from augmented SUMOylation of the R882 mutant. Because the DNMT3A R882 mutation reduces its methyl transferase activity, we investigated which genes would be upregulated from the DNMT3A repressor complex. To do this, we established isogenic TF-1 cell lines that harbor haploid DNMT3A knockout (DNMT3A+/-) using the Cripsr-Cas9 system. We also treated cells with HDACi and 5-azacytidine (5-aza) which inhibit HDAC and DNMT, respectively. Interestingly, we discovered that PD-L1 expression is induced by HDACi and 5-aza treatment. Chemical inhibition by 5-aza or genetic inhibition by knockout reduces DNMT3A activity and synergized with HDACi to increase PD-L1 expression. Flow cytometry analysis also demonstrated increased membrane PD-L1 expression in response to HDACi. We also found out that DNMT3A+/- resulted in higher Histone H3K27 acetylation, which is known as a gene activation mark. Higher H3K27 acetylation in DNMT3A+/- cell confirms the findings by other groups but the mechanisms by which this occurs are unknown. We suggest that haploinsufficiency of DNMT3A results in a reduced DNMT3A-HDAC interaction leading to higher H3K27 acetylation and increased PD-L1 expression. Our results also revealed that HDACi treatment induced cell cycle arrest, DNA damage and apoptosis at increasing levels in DNMT3A+/- cell. Even though the DNMT3A+/- TF-1 showed increased sensitivity to HDACi treatment, we observed a correlation of higher phosphor- ERK1/2 and PD-L1 levels in the surviving cells. The enhanced expression of PD-L1 and activation of ERK1/2 may explain in part how mutated DNMT3A contributes to drug resistance and immune checkpoint avoidance. Many oncology clinical trials are underway utilizing HDACi. However, the questions of which mutational backgrounds might be most sensitive to these agents and how to best combine them with other agents remain to be answered. To test whether reduced DNMT3A activity increases PD-L1 expression in vivo, we crossed floxed DNMT3A mice with Mx1-Cre mice. After 4 weeks of induction of Cre recombinase by injecting pIpC in the progeny carrying both genetically engineered changes, lineage depleted mouse BM cells were analyzed for PD-L1 expression using quantitative RT-PCR. BM cells derived from DNMT3A knockout mice showed increased expression of PD-L1 compared to wild-type mice. Treatment of these BM cells with an HDACi and/or 5-aza resulted in a synergistic induction of PD-L1 expression for the combination. Taken together, we suggest that mutant DNMT3A induces higher H3K27 acetylation along with PD-L1 expression due to a looser complex between HDAC1 and mutant DNMT3A. Therefore, we suggest that combined treatment with an HDACi and an immune checkpoint inhibitor targeting the PD-L1/PD-1 axis might be a promising strategy for treating DNMT3A mutant AML patients. Disclosures Small: InSilico Medicine: Membership on an entity's Board of Directors or advisory committees; Pharos I, B & T: Consultancy, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document