Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 369
Filter
1.
Int J Med Sci ; 18(16): 3728-3737, 2021.
Article in English | MEDLINE | ID: mdl-34790046

ABSTRACT

Intrinsic or developing resistance to chemotherapy drugs including cisplatin (CDDP) remains the major limitation of cancer therapeutic efficacy in cancers. Recently, increasing evidence suggested that long noncoding RNAs (lncRNAs) play a critical role in various biological processes of tumors, and have been implicated in resistance to various drugs. However, the role of lncRNAs in cisplatin resistance is poorly understood. Here, we found that the expression of lncRNA CEBPA-DT/CEBPA/BCL2 was upregulated in cisplatin resistance OSCC cells (Cal27-CisR and HSC4-CisR) compared with their parental cells (Cal27 and HSC4). CEBPA-DT overexpression could upregulated both cytoplasmic and nuclear CEBPA expression. Down-regulation of CEBPA-DT enhances cisplatin sensitivity, facilitates cell apoptosis in cisplatin-resistant OSCC cells. In addition, we identified that CEBPA-DT regulates cisplatin chemosensitivity through CEBPA/BCL2-mediated cell apoptosis. Knockdown of CEBPA and BCL2 could alleviate the increasement of cisplatin resistance induced by CEBPA-DT overexpression. Our findings indicate that downregulation of lncRNA CEBPA-DT may be a potential therapy to overcome cisplatin resistance in OSCC.


Subject(s)
Carcinoma, Squamous Cell , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Mouth Neoplasms , RNA, Long Noncoding/physiology , Apoptosis/drug effects , Apoptosis/genetics , CCAAT-Enhancer-Binding Proteins/physiology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/physiology , Signal Transduction/genetics
2.
Nucleic Acids Res ; 49(11): 6053-6068, 2021 06 21.
Article in English | MEDLINE | ID: mdl-33939809

ABSTRACT

During S phase, the cooperation between the macromolecular complexes regulating DNA synthesis, epigenetic information maintenance and DNA repair is advantageous for cells, as they can rapidly detect DNA damage and initiate the DNA damage response (DDR). UHRF1 is a fundamental epigenetic regulator; its ability to coordinate DNA methylation and histone code is unique across proteomes of different species. Recently, UHRF1's role in DNA damage repair has been explored and recognized to be as important as its role in maintaining the epigenome. UHRF1 is a sensor for interstrand crosslinks and a determinant for the switch towards homologous recombination in the repair of double-strand breaks; its loss results in enhanced sensitivity to DNA damage. These functions are finely regulated by specific post-translational modifications and are mediated by the SRA domain, which binds to damaged DNA, and the RING domain. Here, we review recent studies on the role of UHRF1 in DDR focusing on how it recognizes DNA damage and cooperates with other proteins in its repair. We then discuss how UHRF1's epigenetic abilities in reading and writing histone modifications, or its interactions with ncRNAs, could interlace with its role in DDR.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , DNA Repair , Epigenome , Ubiquitin-Protein Ligases/physiology , Animals , CCAAT-Enhancer-Binding Proteins/chemistry , DNA Damage , Epigenesis, Genetic , Genomic Instability , Histone Code , Humans , Ubiquitin-Protein Ligases/chemistry
3.
JCI Insight ; 6(5)2021 03 08.
Article in English | MEDLINE | ID: mdl-33539326

ABSTRACT

Elevation of intraocular pressure (IOP) due to trabecular meshwork (TM) damage is associated with primary open-angle glaucoma (POAG). Myocilin mutations resulting in elevated IOP are the most common genetic causes of POAG. We have previously shown that mutant myocilin accumulates in the ER and induces chronic ER stress, leading to TM damage and IOP elevation. However, it is not understood how chronic ER stress leads to TM dysfunction and loss. Here, we report that mutant myocilin activated autophagy but was functionally impaired in cultured human TM cells and in a mouse model of myocilin-associated POAG (Tg-MYOCY437H). Genetic and pharmacological inhibition of autophagy worsened mutant myocilin accumulation and exacerbated IOP elevation in Tg-MYOCY437H mice. Remarkably, impaired autophagy was associated with chronic ER stress-induced transcriptional factor CHOP. Deletion of CHOP corrected impaired autophagy, enhanced recognition and degradation of mutant myocilin by autophagy, and reduced glaucoma in Tg-MYOCY437H mice. Stimulating autophagic flux via tat-beclin 1 peptide or torin 2 promoted autophagic degradation of mutant myocilin and reduced elevated IOP in Tg-MYOCY437H mice. Our study provides an alternate treatment strategy for myocilin-associated POAG by correcting impaired autophagy in the TM.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Glaucoma, Open-Angle/metabolism , Glycoproteins/metabolism , Ocular Hypertension/metabolism , Trabecular Meshwork/metabolism , Animals , Autophagy , Cells, Cultured , Humans , Mice, Inbred C57BL , Mice, Knockout
4.
Yi Chuan ; 43(1): 84-93, 2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33509777

ABSTRACT

As a best-characterized epigenetic modification, DNA methylation plays an important role in mammalian development. Uhrf1 is a critical epigenetic regulator that can bind to hemimethylated DNA and recruit DNA methyltransferase 1 to maintain DNA methylation. So far, the role of Uhrf1-mediated DNA methylation in intestinal development is still unknown. In order to investigate the impact of Uhrf1 deletion in intestinal development, we have successfully constructed the epithelial-specific Uhrf1 knockout mouse model. After Uhrf1 ablation, we found the mutant mice exhibited abnormal epithlial structure with less and shorter villi and shrinked crypts compared with wild type mice via hematoxylin-eosin staining. Further analysis showed that Uhrf1 deletion in the intestinal epithelium significantly decreased the cell proliferation and induced cell apoptosis. In addition, Uhrf1 deletion inhibited the normal epithelial differentiation and the expression of intestinal stem cell marker genes. Preliminary mechanism study revealed that loss of Uhrf1 caused global DNA hypomethylation which induced DNA damage in crypt cells. Taken together, our data suggested that DNA methylation mediated by Uhrf1 is vital for the normal intestinal development. Our results enriched the in vivo role of Uhrf1 and laid the foundation for further epigenetic regulatory mechanism exploration.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , DNA Methylation , Epigenesis, Genetic , Intestines/growth & development , Ubiquitin-Protein Ligases/physiology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Gene Deletion , Mice , Mice, Knockout , Ubiquitin-Protein Ligases/genetics
5.
PLoS Biol ; 18(12): e3000975, 2020 12.
Article in English | MEDLINE | ID: mdl-33306668

ABSTRACT

The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase and critical regulator of cell cycle progression. Despite its vital role, it has remained challenging to globally map APC/C substrates. By combining orthogonal features of known substrates, we predicted APC/C substrates in silico. This analysis identified many known substrates and suggested numerous candidates. Unexpectedly, chromatin regulatory proteins are enriched among putative substrates, and we show experimentally that several chromatin proteins bind APC/C, oscillate during the cell cycle, and are degraded following APC/C activation, consistent with being direct APC/C substrates. Additional analysis revealed detailed mechanisms of ubiquitylation for UHRF1, a key chromatin regulator involved in histone ubiquitylation and DNA methylation maintenance. Disrupting UHRF1 degradation at mitotic exit accelerates G1-phase cell cycle progression and perturbs global DNA methylation patterning in the genome. We conclude that APC/C coordinates crosstalk between cell cycle and chromatin regulatory proteins. This has potential consequences in normal cell physiology, where the chromatin environment changes depending on proliferative state, as well as in disease.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Chromatin/metabolism , Ubiquitin-Protein Ligases/metabolism , Anaphase-Promoting Complex-Cyclosome/physiology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/physiology , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cell Line , Chromatin/genetics , Computer Simulation , HEK293 Cells , HeLa Cells , Humans , Protein Processing, Post-Translational , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitination
6.
Med Oncol ; 37(12): 109, 2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33170359

ABSTRACT

CEBPA and c-MYC genes belong to TF and play an essential role in hematologic malignancies development. Furthermore, these genes also co-regulate with RUNX1 and lead to bone marrow differentiation and may contribute to the leukemic transformation. Understanding the function and full characteristics of selected genes in the group of patients with AML can be helpful in assessing prognosis, and their usefulness as prognostic factors can be revealed. The aim of the study was to evaluate CEBPA and c-MYC mRNA expression level and to seek their association with demographical and clinical features of AML patients such as: age, gender, FAB classification, mortality or leukemia cell karyotype. Obtained results were also correlated with the expression level of the RUNX gene family. To assess of relative gene expression level the qPCR method was used. The expression levels of CEBPA and c-MYC gene varied among patients. Neither CEBPA nor c-MYC expression levels differed significantly between women and men (p=0.8325 and p=0.1698, respectively). No statistically significant correlation between age at the time of diagnosis and expression of CEBPA (p=0.4314) or c-MYC (p=0.9524) was stated. There were no significant associations between relative CEBPA (p=0.4247) or c-MYC (p=0.4655) expression level and FAB subtype and mortality among the enrolled patients (p=0.5858 and p=0.8437, respectively). However, it was observed that c-MYC and RUNX1 expression levels were significantly positively correlated (rS=0.328, p=0.0411). Overall, AML pathogenesis involves a complex interaction among CEBPA, c-MYC and RUNX family genes.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Leukemia, Myeloid, Acute/etiology , Proto-Oncogene Proteins c-myc/genetics , Adolescent , Adult , Aged , Aged, 80 and over , CCAAT-Enhancer-Binding Proteins/physiology , Core Binding Factor Alpha 2 Subunit/genetics , Female , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Proto-Oncogene Proteins c-myc/physiology , RNA, Messenger/analysis , Young Adult
7.
Mol Biol Cell ; 31(23): 2511-2521, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32877276

ABSTRACT

Adipogenesis is regulated by a cascade of signals that drive transcriptional reprogramming in adipocytes. Here, we report that nuclear actin regulates the chromatin states that establish tissue- specific expression during adipogenesis. To study the role of ß-actin in adipocyte differentiation, we conducted RNA sequencing on wild-type and ß-actin knockout mouse embryonic fibroblasts (MEFs) after reprograming to adipocytes. We found that ß-actin depletion affects induction of several adipogenic genes during transcriptional reprograming. This impaired regulation of adipogenic genes is linked to reduced expression of the pioneer factor Cebpa and is rescued by reintroducing NLS-tagged ß-actin. ATAC-Seq in knockout MEFs revealed that actin-dependent reduction of Cebpa expression correlates with decreased chromatin accessibility and loss of chromatin association of the ATPase Brg1. This, in turn, impairs CEBPB's association with its Cebpa promoter-proximal binding site during adipogenesis. We propose a role for the nuclear ß-actin pool in maintaining open chromatin for transcriptional reprogramming during adipogenic differentiation.


Subject(s)
Actins/metabolism , Adipogenesis/genetics , Chromatin/metabolism , 3T3-L1 Cells , Actins/physiology , Adipocytes/metabolism , Adipogenesis/physiology , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Cell Differentiation/genetics , Chromatin/physiology , Fibroblasts/metabolism , Mice , Promoter Regions, Genetic/genetics , Transcriptional Activation/physiology
8.
Cell Rep ; 32(4): 107958, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32726623

ABSTRACT

UHRF1 is a key regulator in DNA methylation maintenance. It binds histone H3K9me2/3 and hemi-methylated DNA and recruits DNMT1 to DNA replication forks during S phase. However, the regulatory mechanism of hemi-methylated DNA binding activity of UHRF1 remains unknown. In this study, we reveal that acetylation of UHRF1 is regulated by PCAF and HDAC1. We show that UHRF1 acetylation at K490 attenuates its binding affinity to hemi-methylated DNA. We analyze genome-wide DNA methylation and gene-expression patterns using stable cell lines and discover that cells where the endogenous UHRF1 is replaced with an acetyl-mimetic (UHRF1 K490Q) mutant show deficiencies in inherited DNA methylation and show different gene-expression patterns in genes related to cell survival. These results reveal that precise regulation of UHRF1 acetylation is required to maintain DNA methylation during cell division and control cell survival.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA Methylation/physiology , Ubiquitin-Protein Ligases/metabolism , Acetylation , CCAAT-Enhancer-Binding Proteins/physiology , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HCT116 Cells , HEK293 Cells , Histone Deacetylase 1/metabolism , Histones/metabolism , Humans , K562 Cells , Nuclear Proteins/metabolism , Protein Binding , Protein Processing, Post-Translational , Ubiquitin-Protein Ligases/physiology , p300-CBP Transcription Factors/metabolism
9.
Cell Biol Int ; 44(10): 2075-2085, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32652867

ABSTRACT

DNA-binding protein A (dbpA) is reported to be upregulated in many cancers and associated with tumor progress. The present study aimed to investigate the role of dbpA in 5-fluorouracil (5-FU)-resistant and oxaliplatin (L-OHP)-resistant colorectal cancer (CRC) cells. We found that 5-FU and L-OPH treatment promoted the expression of dbpA. Enhanced dbpA promoted the drug resistance of SW620 cells to 5-FU and L-OHP. DbpA knockdown inhibited cell proliferation, induced cell apoptosis, and cell cycle arrested in SW620/5-FU and SW620/L-OHP cells. Besides, dbpA short hairpin RNA (shRNA) enhanced the cytotoxicity of 5-FU and L-OHP to SW620/5-FU and SW620/L-OHP cells. Meanwhile, dbpA shRNA inhibited the activation of the Wnt/ß-catenin pathway that induced by 5-FU stimulation in SW620/5-FU cells. Activation of the Wnt/ß-catenin pathway or overexpression of checkpoint kinase 1 (Chk1) abrogated the promoting effect of dbpA downregulation on 5-FU sensitivity of CRC cells. Importantly, downregulation of dbpA suppressed tumor growth and promoted CRC cells sensitivity to 5-FU in vivo. Our study indicated that the knockdown of dbpA enhanced the sensitivity of CRC cells to 5-FU via Wnt/ß-catenin/Chk1 pathway, and DbpA may be a potential therapeutic target to sensitize drug resistance CRC to 5-FU and L-OHP.


Subject(s)
Apoptosis/drug effects , CCAAT-Enhancer-Binding Proteins/physiology , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/physiology , Animals , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Wnt Signaling Pathway/drug effects
10.
Br J Haematol ; 190(4): 495-507, 2020 08.
Article in English | MEDLINE | ID: mdl-32086816

ABSTRACT

Recent advances in sequencing technologies have allowed for the identification of recurrent mutations in acute myeloid leukaemia (AML). The transcription factor CCAAT enhancer binding protein alpha (CEBPA) is frequently mutated in AML, and biallelic CEBPA-mutant AML was recognised as a separate disease entity in the recent World Health Organization classification. However, CEBPA mutations are co-occurring with other aberrations in AML, and together these lesions form the clonal hierarchy that comprises the leukaemia in the patient. Here, we aim to review the current understanding of co-occurring mutations in CEBPA-mutated AML and their implications for disease biology and clinical outcome. We will put emphasis on patterns of cooperation, how these lesions cooperate with CEBPA mutations and the underlying potential molecular mechanisms. Finally, we will relate this to patient outcome and future options for personalised medicine.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Neoplasm Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , CCAAT-Enhancer-Binding Proteins/physiology , Child , Child, Preschool , Clonal Evolution , DNA Methylation , Female , Histone Code , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Leukemia, Myeloid, Acute/classification , Male , Middle Aged , Neoplasm Proteins/physiology , Precision Medicine , RNA Splicing Factors/genetics , RNA Splicing Factors/physiology , Recurrence , Transcription Factors/genetics , Transcription Factors/physiology , Treatment Outcome , Young Adult
11.
Cell Tissue Res ; 378(3): 427-440, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31309319

ABSTRACT

The differentiation of sensory neurons involves gene expression changes induced by specific transcription factors. Vomeronasal sensory neurons (VSNs) in the mouse vomeronasal organ (VNO) consist of two major subpopulations of neurons expressing vomeronasal 1 receptor (V1r)/Gαi2 or vomeronasal 2 receptor (V2r)/Gαo, which differentiate from a common neural progenitor. We previously demonstrated that the differentiation and survival of VSNs were inhibited in ATF5 transcription factor-deficient mice (Nakano et al. Cell Tissue Res 363:621-633, 2016). These defects were more prominent in V2r/Gαo-type than in V1r/Gαi2-type VSNs; however, the molecular mechanisms responsible for the differentiation of V2r/Gαo-type VSNs by ATF5 remain unclear. To identify a cofactor involved in ATF5-regulated VSN differentiation, we investigated the expression and function of CCAAT/enhancer-binding protein gamma (C/EBPγ, Cebpg), which is a major C/EBP family member expressed in the mouse VNO and dimerizes with ATF5. The results obtained showed that C/EBPγ mRNAs and proteins were broadly expressed in the postmitotic VSNs of the neonatal VNO, and their expression decreased by the second postnatal week. The C/EBPγ protein was expressed in the nuclei of approximately 70% of VSNs in the neonatal VNO, and 20% of the total VSNs co-expressed C/EBPγ and ATF5 proteins. We examined the trans-acting effects of C/EBPγ and ATF5 on V2r transcription and found that the co-expression of C/EBPγ and ATF5, but not C/EBPγ or ATF5 alone, increased Vmn2r66 promoter reporter activity via the C/EBP:ATF response element (CARE) in Neuro2a cells. These results suggest the role of C/EBPγ on ATF5-regulated VSN differentiation in early postnatal development.


Subject(s)
Activating Transcription Factors/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Sensory Receptor Cells , Vomeronasal Organ , Animals , Cell Differentiation , Cell Line, Tumor , Mice , Mice, Inbred C57BL , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Vomeronasal Organ/growth & development , Vomeronasal Organ/metabolism
12.
Dev Cell ; 50(1): 43-56.e6, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31231040

ABSTRACT

Two major functions of the epigenome are to regulate gene expression and to suppress transposons. It is unclear how these functions are balanced during physiological challenges requiring tissue regeneration, where exquisite coordination of gene expression is essential. Transcriptomic analysis of seven time points following partial hepatectomy identified the epigenetic regulator UHRF1, which is essential for DNA methylation, as dynamically expressed during liver regeneration in mice. UHRF1 deletion in hepatocytes (Uhrf1HepKO) caused genome-wide DNA hypomethylation but, surprisingly, had no measurable effect on gene or transposon expression or liver homeostasis. Partial hepatectomy of Uhrf1HepKO livers resulted in early and sustained activation of proregenerative genes and enhanced liver regeneration. This was attributed to redistribution of H3K27me3 from promoters to transposons, effectively silencing them and, consequently, alleviating repression of liver regeneration genes, priming them for expression in Uhrf1HepKO livers. Thus, epigenetic compensation safeguards the genome against transposon activation, indirectly affecting gene regulation.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , DNA Methylation , Epigenomics , Gene Expression Regulation , Hepatocytes/cytology , Liver Regeneration , Ubiquitin-Protein Ligases/physiology , Animals , Gene Expression Profiling , Hepatocytes/physiology , Male , Mice , Mice, Knockout
13.
Oncogene ; 38(28): 5686-5699, 2019 07.
Article in English | MEDLINE | ID: mdl-31043707

ABSTRACT

UHRF1 is an important epigenetic regulator that belongs to the UHRF family. Overexpression of UHRF1 has been found in many kinds of tumors and its overexpression is associated with poor prognosis and short survival in certain cancer types. However, its function in renal cell carcinoma (RCC) is not clear. Here we report that RCC tumor tissues had obviously higher UHRF1 expression than normal renal tissues. Downregulation of UHRF1 by siRNA or shRNA in RCC cell lines resulted in decreased cell viability, inhibited cell migration and invasion, and increased apoptosis. UHRF1 knockdown RCC xenografts also resulted in obviously inhibited tumor growth in vivo. After downregulation of UHRF1 in RCC cells, the expression of TXNIP was upregulated. In addition, after UHRF1 and TXNIP were simultaneously downregulated, cell viability and cell invasion increased, whereas cell apoptosis decreased compared with UHRF1 single downregulated cells. We also showed that UHRF1 could recruit HDAC1 to the TXNIP promoter and mediate the deacetylation of histone H3K9, resulting in the inhibition of TXNIP expression. Our results confirm that UHRF1 has oncogenic function in RCC and UHRF1 may promote tumor progression through epigenetic regulation of TXNIP. UHRF1 might be used as a therapeutic target for RCC treatment.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Carcinoma, Renal Cell/pathology , Carrier Proteins/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/physiology , Kidney Neoplasms/pathology , Ubiquitin-Protein Ligases/physiology , Acetylation , Animals , Apoptosis , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Movement , Down-Regulation , Heterografts , Histones/metabolism , Humans , Kidney Neoplasms/genetics , Mice , Mice, Nude , Neoplasm Invasiveness
14.
Biochim Biophys Acta Gene Regul Mech ; 1862(1): 96-106, 2019 01.
Article in English | MEDLINE | ID: mdl-30550771

ABSTRACT

The myeloid master regulator CCAAT enhancer-binding protein alpha (CEBPA) is known as a pioneer factor. In this study, we report the CEBPA cistrome of THP-1 human monocytes after stimulation with the vitamin D receptor (VDR) ligand 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) for 2, 8 and 24 h. About a third of the genomic VDR binding sites co-located with those of CEBPA. In parallel, the binding strength of 5% of the CEBPA cistrome, i.e. some 1500 sites, is significantly (p < 0.001) affected by 1,25(OH)2D3. Transcriptome-wide analysis after CEBPA silencing indicated that the pioneer factor enhances both the basal expression and ligand inducibility of 70 vitamin D target genes largely involved in lipid signaling and metabolism. In contrast, CEBPA suppresses 82 vitamin D target genes many of which are related to the modulation of T cell activity by monocytes. The inducibility of the promoter-specific histone marker H3K4me3 distinguishes the former class of genes from the latter. Moreover, prominent occupancy of the myeloid pioneer factor PU.1 on 1,25(OH)2D3-sensitive CEBPA enhancers mechanistically explains the dichotomy of vitamin D target genes. In conclusion, CEBPA supports vitamin D signaling concerning actions of the innate immune system, but uses the antagonism with PU.1 for suppressing possible overreactions of adaptive immunity.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Receptors, Calcitriol/metabolism , Vitamin D/analogs & derivatives , Adaptive Immunity , Humans , Immunity, Innate , Monocytes/metabolism , Proto-Oncogene Proteins/physiology , Signal Transduction , THP-1 Cells , Trans-Activators/physiology , Vitamin D/metabolism
15.
PLoS Genet ; 14(10): e1007643, 2018 10.
Article in English | MEDLINE | ID: mdl-30335751

ABSTRACT

The Fanconi Anemia (FA) pathway is important for repairing interstrand crosslinks (ICLs) between the Watson-Crick strands of the DNA double helix. An initial and essential stage in the repair process is the detection of the ICL. Here, we report the identification of UHRF2, a paralogue of UHRF1, as an ICL sensor protein. UHRF2 is recruited to ICLs in the genome within seconds of their appearance. We show that UHRF2 cooperates with UHRF1, to ensure recruitment of FANCD2 to ICLs. A direct protein-protein interaction is formed between UHRF1 and UHRF2, and between either UHRF1 and UHRF2, and FANCD2. Importantly, we demonstrate that the essential monoubiquitination of FANCD2 is stimulated by UHRF1/UHRF2. The stimulation is mediating by a retention of FANCD2 on chromatin, allowing for its monoubiquitination by the FA core complex. Taken together, we uncover a mechanism of ICL sensing by UHRF2, leading to FANCD2 recruitment and retention at ICLs, in turn facilitating activation of FANCD2 by monoubiquitination.


Subject(s)
DNA Repair/physiology , Fanconi Anemia Complementation Group D2 Protein/physiology , Ubiquitin-Protein Ligases/physiology , Amino Acid Sequence , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , DNA/metabolism , DNA Damage/physiology , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Protein Interaction Domains and Motifs , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
16.
Haematologica ; 103(8): 1269-1277, 2018 08.
Article in English | MEDLINE | ID: mdl-29773596

ABSTRACT

Maturation of granulocytes is dependent on controlled gene expression by myeloid lineage restricted transcription factors. CEBPE is one of the essential transcription factors required for granulocytic differentiation. Identification of downstream targets of CEBPE is vital to understand better its role in terminal granulopoiesis. In this study, we have identified Card10 as a novel target of CEBPE. We show that CEBPE binds to regulatory elements upstream of the murine Card10 locus, and expression of CARD10 is significantly reduced in Cebpe knock-out mice. Silencing Card10 in a human cell line and in murine primary cells impaired granulopoiesis, affecting expression of genes involved in myeloid cell development and function. Taken together, our data demonstrate for the first time that Card10 is expressed in granulocytes and is a direct target of CEBPE with functions extending to myeloid differentiation.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Cell Differentiation , Granulocytes/cytology , Animals , Binding Sites , Cell Line , Cells, Cultured , Gene Expression Regulation , Granulocytes/metabolism , Humans , Mice , Myeloid Cells , Protein Binding , Transcription Factors/genetics
18.
Immunity ; 48(2): 364-379.e8, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466759

ABSTRACT

Neutrophils are specialized innate cells that require constant replenishment from proliferative bone marrow (BM) precursors as a result of their short half-life. Although it is established that neutrophils are derived from the granulocyte-macrophage progenitor (GMP), the differentiation pathways from GMP to functional mature neutrophils are poorly defined. Using mass cytometry (CyTOF) and cell-cycle-based analysis, we identified three neutrophil subsets within the BM: a committed proliferative neutrophil precursor (preNeu) which differentiates into non-proliferating immature neutrophils and mature neutrophils. Transcriptomic profiling and functional analysis revealed that preNeu require the C/EBPε transcription factor for their generation from the GMP, and their proliferative program is substituted by a gain of migratory and effector function as they mature. preNeus expand under microbial and tumoral stress, and immature neutrophils are recruited to the periphery of tumor-bearing mice. In summary, our study identifies specialized BM granulocytic populations that ensure supply under homeostasis and stress responses.


Subject(s)
Bone Marrow Cells/physiology , Neutrophils/physiology , Animals , Bone Marrow Cells/immunology , CCAAT-Enhancer-Binding Proteins/physiology , Cell Lineage , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Expression Profiling , Humans , Mice , Neoplasms, Experimental/immunology , Neutrophils/immunology
20.
Compr Physiol ; 7(2): 635-674, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28333384

ABSTRACT

Adipocytes are the defining cell type of adipose tissue. Once considered a passive participant in energy storage, adipose tissue is now recognized as a dynamic organ that contributes to several important physiological processes, such as lipid metabolism, systemic energy homeostasis, and whole-body insulin sensitivity. Therefore, understanding the mechanisms involved in its development and function is of great importance. Adipocyte differentiation is a highly orchestrated process which can vary between different fat depots as well as between the sexes. While hormones, miRNAs, cytoskeletal proteins, and many other effectors can modulate adipocyte development, the best understood regulators of adipogenesis are the transcription factors that inhibit or promote this process. Ectopic expression and knockdown approaches in cultured cells have been widely used to understand the contribution of transcription factors to adipocyte development, providing a basis for more sophisticated in vivo strategies to examine adipogenesis. To date, over two dozen transcription factors have been shown to play important roles in adipocyte development. These transcription factors belong to several families with many different DNA-binding domains. While peroxisome proliferator-activated receptor gamma (PPARγ) is undoubtedly the most important transcriptional modulator of adipocyte development in all types of adipose tissue, members of the CCAAT/enhancer-binding protein, Krüppel-like transcription factor, signal transducer and activator of transcription, GATA, early B cell factor, and interferon-regulatory factor families also regulate adipogenesis. The importance of PPARγ activity is underscored by several covalent modifications that modulate its activity and its ability to modulate adipocyte development. This review will primarily focus on the transcriptional control of adipogenesis in white fat cells and on the mechanisms involved in this fine-tuned developmental process. © 2017 American Physiological Society. Compr Physiol 7:635-674, 2017.


Subject(s)
Adipogenesis/physiology , Transcription, Genetic/physiology , Adipocytes/cytology , Adipocytes/physiology , Adipogenesis/genetics , Animals , CCAAT-Enhancer-Binding Proteins/physiology , Gene Expression Regulation/physiology , Humans , Kruppel-Like Transcription Factors/physiology , Metabolic Diseases/physiopathology , Models, Biological , PPAR gamma/physiology , Phosphorylation/physiology , Receptors, Steroid/physiology , STAT Transcription Factors/physiology , Serine/metabolism , Sumoylation/physiology , Ubiquitination/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...