Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Prostate ; 84(10): 977-990, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38654435

ABSTRACT

BACKGROUND: It is important to identify molecular features that improve prostate cancer (PCa) risk stratification before radical treatment with curative intent. Molecular analysis of historical diagnostic formalin-fixed paraffin-embedded (FFPE) prostate biopsies from cohorts with post-radiotherapy (RT) long-term clinical follow-up has been limited. Utilizing parallel sequencing modalities, we performed a proof-of-principle sequencing analysis of historical diagnostic FFPE prostate biopsies. We compared patients with (i) stable PCa (sPCa) postprimary or salvage RT, (ii) progressing PCa (pPCa) post-RT, and (iii) de novo metastatic PCa (mPCa). METHODS: A cohort of 19 patients with diagnostic prostate biopsies (n = 6 sPCa, n = 5 pPCa, n = 8 mPCa) and mean 4 years 10 months follow-up (diagnosed 2009-2016) underwent nucleic acid extraction from demarcated malignancy. Samples underwent 3'RNA sequencing (3'RNAseq) (n = 19), nanoString analysis (n = 12), and Illumina 850k methylation (n = 8) sequencing. Bioinformatic analysis was performed to coherently identify differentially expressed genes and methylated genomic regions (MGRs). RESULTS: Eighteen of 19 samples provided useable 3'RNAseq data. Principal component analysis (PCA) demonstrated similar expression profiles between pPCa and mPCa cases, versus sPCa. Coherently differentially methylated probes between these groups identified ~600 differentially MGRs. The top 50 genes with increased expression in pPCa patients were associated with reduced progression-free survival post-RT (p < 0.0001) in an external cohort. CONCLUSIONS: 3'RNAseq, nanoString and 850k-methylation analyses are each achievable from historical FFPE diagnostic pretreatment prostate biopsies, unlocking the potential to utilize large cohorts of historic clinical samples. Profiling similarities between individuals with pPCa and mPCa suggests biological similarities and historical radiological staging limitations, which warrant further investigation.


Subject(s)
Disease Progression , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Aged , Middle Aged , Biopsy , Genomics , Prostate/pathology , Neoplasm Metastasis , Cohort Studies
2.
Clin Cancer Res ; 27(9): 2459-2469, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33597271

ABSTRACT

PURPOSE: Tumor hypoxia fuels an aggressive tumor phenotype and confers resistance to anticancer treatments. We conducted a clinical trial to determine whether the antimalarial drug atovaquone, a known mitochondrial inhibitor, reduces hypoxia in non-small cell lung cancer (NSCLC). PATIENTS AND METHODS: Patients with NSCLC scheduled for surgery were recruited sequentially into two cohorts: cohort 1 received oral atovaquone at the standard clinical dose of 750 mg twice daily, while cohort 2 did not. Primary imaging endpoint was change in tumor hypoxic volume (HV) measured by hypoxia PET-CT. Intercohort comparison of hypoxia gene expression signatures using RNA sequencing from resected tumors was performed. RESULTS: Thirty patients were evaluable for hypoxia PET-CT analysis, 15 per cohort. Median treatment duration was 12 days. Eleven (73.3%) atovaquone-treated patients had meaningful HV reduction, with median change -28% [95% confidence interval (CI), -58.2 to -4.4]. In contrast, median change in untreated patients was +15.5% (95% CI, -6.5 to 35.5). Linear regression estimated the expected mean HV was 55% (95% CI, 24%-74%) lower in cohort 1 compared with cohort 2 (P = 0.004), adjusting for cohort, tumor volume, and baseline HV. A key pharmacodynamics endpoint was reduction in hypoxia-regulated genes, which were significantly downregulated in atovaquone-treated tumors. Data from multiple additional measures of tumor hypoxia and perfusion are presented. No atovaquone-related adverse events were reported. CONCLUSIONS: This is the first clinical evidence that targeting tumor mitochondrial metabolism can reduce hypoxia and produce relevant antitumor effects at the mRNA level. Repurposing atovaquone for this purpose may improve treatment outcomes for NSCLC.


Subject(s)
Atovaquone/pharmacology , Gene Expression Regulation, Neoplastic , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Tumor Hypoxia/drug effects , Tumor Hypoxia/genetics , Atovaquone/therapeutic use , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Energy Metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Molecular Imaging , Positron Emission Tomography Computed Tomography , STAT3 Transcription Factor/metabolism
3.
EMBO J ; 39(6): e103777, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32090359

ABSTRACT

Research on non-coding RNA (ncRNA) is a rapidly expanding field. Providing an official gene symbol and name to ncRNA genes brings order to otherwise potential chaos as it allows unambiguous communication about each gene. The HUGO Gene Nomenclature Committee (HGNC, www.genenames.org) is the only group with the authority to approve symbols for human genes. The HGNC works with specialist advisors for different classes of ncRNA to ensure that ncRNA nomenclature is accurate and informative, where possible. Here, we review each major class of ncRNA that is currently annotated in the human genome and describe how each class is assigned a standardised nomenclature.


Subject(s)
Genome, Human/genetics , RNA, Untranslated/classification , Terminology as Topic , Humans , RNA, Untranslated/genetics
4.
Hum Mutat ; 39(12): 1847-1853, 2018 12.
Article in English | MEDLINE | ID: mdl-30199583

ABSTRACT

Ataxia Telangiectasia and Rad3 related (ATR) is one of the main regulators of the DNA damage response. It coordinates cell cycle checkpoint activation, replication fork stability, restart and origin firing to maintain genome integrity. Mutations of the ATR gene have been reported in Seckel patients, who suffer from a rare genetic disease characterized by severe microcephaly and growth retardation. Here, we report the case of a Seckel patient with compound heterozygous mutations in ATR. One allele has an intronic mutation affecting splicing of neighboring exons, the other an exonic missense mutation, producing the variant p.Lys1665Asn, of unknown pathogenicity. We have modeled this novel missense mutation, as well as a previously described missense mutation p.Met1159Ile, and assessed their effect on ATR function. Interestingly, our data indicate that both missense mutations have no direct effect on protein function, but rather result in defective ATR splicing. These results emphasize the importance of splicing mutations in Seckel Syndrome.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Dwarfism/genetics , Microcephaly/genetics , Mutation, Missense , RNA Splicing , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Chickens , Dwarfism/metabolism , Exons , Humans , Introns , Microcephaly/metabolism , Exome Sequencing
5.
RNA Biol ; 15(1): 17-25, 2018 01 02.
Article in English | MEDLINE | ID: mdl-28876172

ABSTRACT

Much evidence is now accumulating that, in addition to their general role in splicing, the components of the core splicing machinery have extensive regulatory potential. In particular, recent evidence has demonstrated that de-regulation of these factors cause the highest extent of alternative splicing changes compared to de-regulation of the classical splicing regulators. This lack of a general inhibition of splicing resonates the differential splicing effects observed in different disease pathologies associated with specific mutations targeting core spliceosomal components. In this review we will summarize what is currently known regarding the involvement of core spliceosomal U-snRNP complexes in perturbed tissue development and human diseases and argue for the existence of a compensatory mechanism enabling cells to cope with drastic perturbations in core splicing components. This system maintains the correct balance of spliceosomal snRNPs through differential expression of variant (v)U-snRNPs.


Subject(s)
Alternative Splicing/genetics , Cell Nucleus/genetics , Ribonucleoproteins, Small Nuclear/genetics , Spliceosomes/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Organ Specificity , RNA Splicing/genetics
6.
Nucleic Acids Res ; 44(22): 10960-10973, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27536002

ABSTRACT

The U1 small nuclear (sn)RNA (U1) is a multifunctional ncRNA, known for its pivotal role in pre-mRNA splicing and regulation of RNA 3' end processing events. We recently demonstrated that a new class of human U1-like snRNAs, the variant (v)U1 snRNAs (vU1s), also participate in pre-mRNA processing events. In this study, we show that several human vU1 genes are specifically upregulated in stem cells and participate in the regulation of cell fate decisions. Significantly, ectopic expression of vU1 genes in human skin fibroblasts leads to increases in levels of key pluripotent stem cell mRNA markers, including NANOG and SOX2. These results reveal an important role for vU1s in the control of key regulatory networks orchestrating the transitions between stem cell maintenance and differentiation. Moreover, vU1 expression varies inversely with U1 expression during differentiation and cell re-programming and this pattern of expression is specifically de-regulated in iPSC-derived motor neurons from Spinal Muscular Atrophy (SMA) type 1 patient's. Accordingly, we suggest that an imbalance in the vU1/U1 ratio, rather than an overall reduction in Uridyl-rich (U)-snRNAs, may contribute to the specific neuromuscular disease phenotype associated with SMA.


Subject(s)
Human Embryonic Stem Cells/physiology , Induced Pluripotent Stem Cells/physiology , RNA, Small Nuclear/genetics , Cells, Cultured , Gene Expression Regulation , Humans , RNA, Small Nuclear/metabolism , Spinal Muscular Atrophies of Childhood/genetics , Transcriptome , Up-Regulation
7.
Nucleic Acids Res ; 43(9): 4721-32, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25897131

ABSTRACT

The conventional model for splicing involves excision of each intron in one piece; we demonstrate this inaccurately describes splicing in many human genes. First, after switching on transcription of SAMD4A, a gene with a 134 kb-long first intron, splicing joins the 3' end of exon 1 to successive points within intron 1 well before the acceptor site at exon 2 is made. Second, genome-wide analysis shows that >60% of active genes yield products generated by such intermediate intron splicing. These products are present at ∼15% the levels of primary transcripts, are encoded by conserved sequences similar to those found at canonical acceptors, and marked by distinctive structural and epigenetic features. Finally, using targeted genome editing, we demonstrate that inhibiting the formation of these splicing intermediates affects efficient exon-exon splicing. These findings greatly expand the functional and regulatory complexity of the human transcriptome.


Subject(s)
Introns , RNA Splicing , Cells, Cultured , Exons , Human Umbilical Vein Endothelial Cells/metabolism , Humans , RNA Splice Sites , Repressor Proteins/genetics , Transcription, Genetic
8.
PLoS One ; 9(9): e107654, 2014.
Article in English | MEDLINE | ID: mdl-25233083

ABSTRACT

The Herpes Simplex Virus 1 (HSV-1)-encoded ICP22 protein plays an important role in viral infection and affects expression of host cell genes. ICP22 is known to reduce the global level of serine (Ser)2 phosphorylation of the Tyr1Ser2Pro3Thr4Ser5Pro6Ser7 heptapeptide repeats comprising the carboxy-terminal domain (CTD) of the large subunit of RNA polymerase (pol) II. Accordingly, ICP22 is thought to associate with and inhibit the activity of the positive-transcription elongation factor b (P-TEFb) pol II CTD Ser2 kinase. We show here that ICP22 causes loss of CTD Ser2 phosphorylation from pol II engaged in transcription of protein-coding genes following ectopic expression in HeLa cells and that recombinant ICP22 interacts with the CDK9 subunit of recombinant P-TEFb. ICP22 also interacts with pol II in vitro. Residues 193 to 256 of ICP22 are sufficient for interaction with CDK9 and inhibition of pol II CTD Ser2 phosphorylation but do not interact with pol II. These results indicate that discrete regions of ICP22 interact with either CDK9 or pol II and that ICP22 interacts directly with CDK9 to inhibit expression of host cell genes.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Herpesvirus 1, Human/genetics , Immediate-Early Proteins/metabolism , Positive Transcriptional Elongation Factor B/antagonists & inhibitors , RNA Polymerase II/antagonists & inhibitors , Cell Line, Tumor , HeLa Cells , Humans , Phosphorylation , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Protein Kinases/metabolism , RNA Polymerase II/genetics , Transcription, Genetic/genetics
9.
Nucleic Acids Res ; 42(1): 264-75, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24097444

ABSTRACT

RNA polymerase II transcribes both protein coding and non-coding RNA genes and, in yeast, different mechanisms terminate transcription of the two gene types. Transcription termination of mRNA genes is intricately coupled to cleavage and polyadenylation, whereas transcription of small nucleolar (sno)/small nuclear (sn)RNA genes is terminated by the RNA-binding proteins Nrd1, Nab3 and Sen1. The existence of an Nrd1-like pathway in humans has not yet been demonstrated. Using the U1 and U2 genes as models, we show that human snRNA genes are more similar to mRNA genes than yeast snRNA genes with respect to termination. The Integrator complex substitutes for the mRNA cleavage and polyadenylation specificity factor complex to promote cleavage and couple snRNA 3'-end processing with termination. Moreover, members of the associated with Pta1 (APT) and cleavage factor I/II complexes function as transcription terminators for human snRNA genes with little, if any, role in snRNA 3'-end processing. The gene-specific factor, proximal sequence element-binding transcription factor (PTF), helps clear the U1 and U2 genes of nucleosomes, which provides an easy passage for pol II, and the negative elongation factor facilitates termination at the end of the genes where nucleosome levels increase. Thus, human snRNA genes may use chromatin structure as an additional mechanism to promote efficient transcription termination in vivo.


Subject(s)
RNA, Small Nuclear/genetics , Transcription Termination, Genetic , mRNA Cleavage and Polyadenylation Factors/metabolism , Chromatin/chemistry , HeLa Cells , Humans , RNA 3' End Processing , RNA, Small Nuclear/biosynthesis , RNA, Small Nuclear/metabolism , Transcription Factors/physiology
10.
Genome Res ; 23(2): 281-91, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23070852

ABSTRACT

Human U1 small nuclear (sn)RNA, required for splicing of pre-mRNA, is encoded by genes on chromosome 1 (1p36). Imperfect copies of these U1 snRNA genes, also located on chromosome 1 (1q12-21), were thought to be pseudogenes. However, many of these "variant" (v)U1 snRNA genes produce fully processed transcripts. Using antisense oligonucleotides to block the activity of a specific vU1 snRNA in HeLa cells, we have identified global transcriptome changes following interrogation of the Affymetrix Human Exon ST 1.0 array. Our results indicate that this vU1 snRNA regulates expression of a subset of target genes at the level of pre-mRNA processing. This is the first indication that variant U1 snRNAs have a biological function in vivo. Furthermore, some vU1 snRNAs are packaged into unique ribonucleoproteins (RNPs), and many vU1 snRNA genes are differentially expressed in human embryonic stem cells (hESCs) and HeLa cells, suggesting developmental control of RNA processing through expression of different sets of vU1 snRNPs.


Subject(s)
Alternative Splicing , Gene Expression Regulation , RNA, Small Nuclear/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Molecular Sequence Data , Pseudogenes , RNA, Small Nuclear/metabolism , Ribonucleoproteins/metabolism , Transcription, Genetic
12.
Mol Cell Biol ; 29(14): 4002-13, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19451231

ABSTRACT

The negative elongation factor NELF is a key component of an early elongation checkpoint generally located within 100 bp of the transcription start site of protein-coding genes. Negotiation of this checkpoint and conversion to productive elongation require phosphorylation of the carboxy-terminal domain of RNA polymerase II (pol II), NELF, and DRB sensitivity-inducing factor (DSIF) by positive transcription elongation factor b (P-TEFb). P-TEFb is dispensable for transcription of the noncoding U2 snRNA genes, suggesting that a NELF-dependent checkpoint is absent. However, we find that NELF at the end of the 800-bp U2 gene transcription unit and RNA interference-mediated knockdown of NELF causes a termination defect. NELF is also associated 800 bp downstream of the transcription start site of the beta-actin gene, where a "late" P-TEFb-dependent checkpoint occurs. Interestingly, both genes have an extended nucleosome-depleted region up to the NELF-dependent control point. In both cases, transcription through this region is P-TEFb independent, implicating chromatin in the formation of the terminator/checkpoint. Furthermore, CTCF colocalizes with NELF on the U2 and beta-actin genes, raising the possibility that it helps the positioning and/or function of the NELF-dependent control point on these genes.


Subject(s)
Actins/genetics , Actins/metabolism , Chromatin/genetics , Chromatin/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Binding Sites , CCCTC-Binding Factor , DNA Polymerase II/chemistry , DNA Polymerase II/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Humans , Models, Biological , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Phosphorylation , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription, Genetic , Transcriptional Elongation Factors/antagonists & inhibitors , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
13.
Biochem Soc Trans ; 36(Pt 4): 590-4, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18631122

ABSTRACT

In addition to protein-coding genes, mammalian pol II (RNA polymerase II) transcribes independent genes for some non-coding RNAs, including the spliceosomal U1 and U2 snRNAs (small nuclear RNAs). snRNA genes differ from protein-coding genes in several key respects and some of the mechanisms involved in expression are gene-type-specific. For example, snRNA gene promoters contain an essential PSE (proximal sequence element) unique to these genes, the RNA-encoding regions contain no introns, elongation of transcription is P-TEFb (positive transcription elongation factor b)-independent and RNA 3'-end formation is directed by a 3'-box rather than a cleavage and polyadenylation signal. However, the CTD (C-terminal domain) of pol II closely couples transcription with RNA 5' and 3' processing in expression of both gene types. Recently, it was shown that snRNA promoter-specific recognition of the 3'-box RNA processing signal requires a novel phosphorylation mark on the pol II CTD. This new mark plays a critical role in the recruitment of the snRNA gene-specific RNA-processing complex, Integrator. These new findings provide the first example of a phosphorylation mark on the CTD heptapeptide that can be read in a gene-type-specific manner, reinforcing the notion of a CTD code. Here, we review the control of expression of snRNA genes from initiation to termination of transcription.


Subject(s)
Gene Expression Regulation/genetics , RNA, Small Nuclear/genetics , Animals , Humans , Protein Binding , RNA Polymerase II/metabolism , RNA, Small Nuclear/metabolism , Transcription, Genetic/genetics
14.
Science ; 318(5857): 1777-9, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-18079403

ABSTRACT

RNA polymerase II (Pol II) transcribes genes that encode proteins and noncoding small nuclear RNAs (snRNAs). The carboxyl-terminal repeat domain (CTD) of the largest subunit of mammalian RNA Pol II, comprising tandem repeats of the heptapeptide consensus Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7, is required for expression of both gene types. We show that mutation of serine-7 to alanine causes a specific defect in snRNA gene expression. We also present evidence that phosphorylation of serine-7 facilitates interaction with the snRNA gene-specific Integrator complex. These findings assign a biological function to this amino acid and highlight a gene type-specific requirement for a residue within the CTD heptapeptide, supporting the existence of a CTD code.


Subject(s)
Gene Expression Regulation , RNA Polymerase II/metabolism , RNA, Small Nuclear/genetics , Serine/metabolism , Transcription, Genetic , Alanine , Amino Acid Sequence , Cell Line , Consensus Sequence , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Mutation , Oligopeptides/chemistry , Oligopeptides/metabolism , Phosphorylation , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Templates, Genetic
15.
Genomics ; 90(3): 407-15, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17583472

ABSTRACT

Transcriptional regulation in higher eukaryotes frequently involves long-range interactions, up to tens of hundreds of kilobases away, of a number of cis-acting regulatory DNA elements. Using the chromosome conformation capture technique we have analyzed the expression of a small 2.5-kb gene, CD68, in different human cell types and show for the first time that short-range interactions may also be critical. In human monocytes, which produce high levels of CD68 mRNA, the gene is characterized by intramolecular ligations between the promoter and the 3' intervening region. In cells that poorly express the gene a change in architecture is apparent whereby the promoter preferentially associates with the terminator region only. Furthermore, alterations in CD68 gene structure are associated with failings in mRNA splicing and changes with the phosphorylation status of RNA Pol II across the gene. We propose that short-range intrachromosomal interactions may form the basis of coordinated control of monocyte-specific gene regulation.


Subject(s)
Antigens, CD/biosynthesis , Antigens, CD/physiology , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/physiology , Gene Expression Regulation , Cell Line , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA Polymerase II/metabolism , HL-60 Cells , Humans , Jurkat Cells , Models, Genetic , Myeloid Cells/metabolism , Phosphorylation , RNA, Messenger/metabolism , Transcription, Genetic
16.
Genomics ; 84(6): 1030-40, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15533720

ABSTRACT

This study has investigated the transcriptional regulation of the Emr1 gene in murine macrophages and defined an enhancer element within the proximal promoter that is necessary for Emr1 expression in myeloid cells. This element consists of an extended purine-rich sequence (PuRS) of 83 consecutive purine residues containing 9 GGAA sequences, the core binding sequence for members of the Ets family of transcription factors. The Ets factor PU.1 associates with this PuRS element both in vitro and in vivo. Using a standard BLAST search we identified similar PuRS elements in other myeloid and nonmyeloid genes. All PuRS elements tested confer enhancer activity onto a heterologous promoter and chromatin immunoprecipitation experiments revealed that PU.1 associates in vivo with the PuRS elements from the genes expressed in myeloid cells. Our results provide evidence that extended purine-rich sequence elements may constitute a new transcription regulatory motif and that PU.1 association is a prerequisite for macrophage-specific expression.


Subject(s)
Gene Expression Regulation , Macrophages/metabolism , Membrane Glycoproteins/genetics , Mucins/genetics , Myeloid Cells/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Trans-Activators/metabolism , Animals , Base Sequence , Calcium-Binding Proteins , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic , Humans , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Mucins/metabolism , Proto-Oncogene Proteins/genetics , Purines/chemistry , Receptors, G-Protein-Coupled , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Sequence Homology, Nucleic Acid , Trans-Activators/genetics , Transcription, Genetic
17.
J Biol Chem ; 278(24): 21909-19, 2003 Jun 13.
Article in English | MEDLINE | ID: mdl-12676954

ABSTRACT

CD68 is a transmembrane glycoprotein expressed in all cells of the mononuclear phagocyte lineage including monocytes and tissue resident macrophages. Deletion analysis of the 5'-flanking sequences of the gene demonstrated that the proximal -150-bp sequence of the CD68 promoter exhibits high level promoter activity in macrophages. Mutations that abolish Ets factor binding at positions -106 and -89 reduce promoter activity in macrophages to 12 and 30%, respectively. Band shift experiments show that PU.1 associates with the -89 site whereas, Elf-1 preferentially binds the -106 Ets binding site and enhances CD68 activity in vitro. Furthermore, chromatin immunoprecipitation experiments confirm that Elf-1 and PU.1 associate with the CD68 proximal promoter in vivo in THP-1 cells. PU.1 does not bind to the CD68 promoter alone but instead forms heterocomplexes with members of the interferon regulatory factor family (IRF) including IRF-4 and IRF-8. IRF-4 and IRF-8 typically mediate transcriptional activation when associated with PU.1 on composite elements. However, our data show that PU.1/IRF-4 and IRF-8 heterocomplexes down-regulate CD68 promoter activity in macrophages and repression is dependent on the integrity of both the IRF and PU.1 half-sites of this composite element. Chromatin immunoprecipitation data reveal that neither IRF-4 nor IRF-8 associate with the CD68 proximal promoter in macrophages in vivo but IRF-4 is associated with the promoter in B lymphocytes. We propose that expression of CD68 in myeloid cells requires the Ets transcription factors Elf-1 and PU.1 and CD68 expression is down-regulated in lymphoid cells by combinatorial interactions between PU.1 and IRF-4.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , B-Lymphocytes/metabolism , Base Sequence , Binding Sites , Blotting, Western , COS Cells , Cell Line , Down-Regulation , Genes, Reporter , Genetic Vectors , HL-60 Cells , Humans , Interferon Regulatory Factors , Lymphocytes/metabolism , Macrophages/metabolism , Mice , Models, Genetic , Molecular Sequence Data , Mutation , Nuclear Proteins , Plasmids/metabolism , Polymerase Chain Reaction , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Time Factors , Trans-Activators/metabolism , Transcription, Genetic , Transcriptional Activation , Tumor Cells, Cultured , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...