162 research outputs found

    Functional Conservation of Cis-Regulatory Elements of Heat-Shock Genes over Long Evolutionary Distances

    Get PDF
    Transcriptional control of gene regulation is an intricate process that requires precise orchestration of a number of molecular components. Studying its evolution can serve as a useful model for understanding how complex molecular machines evolve. One way to investigate evolution of transcriptional regulation is to test the functions of cis-elements from one species in a distant relative. Previous results suggested that few, if any, tissue-specific promoters from Drosophila are faithfully expressed in C. elegans. Here we show that, in contrast, promoters of fly and human heat-shock genes are upregulated in C. elegans upon exposure to heat. Inducibility under conditions of heat shock may represent a relatively simple “on-off” response, whereas complex expression patterns require integration of multiple signals. Our results suggest that simpler aspects of regulatory logic may be retained over longer periods of evolutionary time, while more complex ones may be diverging more rapidly

    DNA Topoisomerase II Modulates Insulator Function in Drosophila

    Get PDF
    Insulators are DNA sequences thought to be important for the establishment and maintenance of cell-type specific nuclear architecture. In Drosophila there are several classes of insulators that appear to have unique roles in gene expression. The mechanisms involved in determining and regulating the specific roles of these insulator classes are not understood. Here we report that DNA Topoisomerase II modulates the activity of the Su(Hw) insulator. Downregulation of Topo II by RNAi or mutations in the Top2 gene result in disruption of Su(Hw) insulator function. This effect is mediated by the Mod(mdg4)2.2 protein, which is a unique component of the Su(Hw) insulator complex. Co-immunoprecipitation and yeast two-hybrid experiments show that Topo II and Mod(mdg4)2.2 proteins directly interact. In addition, mutations in Top2 cause a slight decrease of Mod(mdg4)2.2 transcript but have a dramatic effect on Mod(mdg4)2.2 protein levels. In the presence of proteasome inhibitors, normal levels of Mod(mdg4)2.2 protein and its binding to polytene chromosomes are restored. Thus, Topo II is required to prevent Mod(mdg4)2.2 degradation and, consequently, to stabilize Su(Hw) insulator-mediated chromatin organization

    Retrotransposons and the evolution of mammalian gene expression

    Full text link
    Transposable elements, and retroviral-like elements in particular, are a rich potential source of genetic variation within a host's genome. Many mutations of endogenous genes in phylogenetically diverse organisms are due to insertion of elements that affect gene expression by altering the normal pattern of regulation. While few such associations are known to have been maintained over time, two recently elucidated examples suggest transposable elements may have a significant impact in evolution of gene expression. The first example, concerning the mouse sex-limited protein ( Slp ), clearly establishes that ancient retroviral enhancer sequences now confer hormonal dependence on the adjacent gene. The second example shows that within the human amylase gene family, salivary specific expression has arisen due to inserted sequences, deriving perhaps from a conjunction of two retrotransposable elements.Peer Reviewedhttp://deepblue.lib.umich.edu/bitstream/2027.42/42800/1/10709_2004_Article_BF00133720.pd

    CpG binding protein (CFP1) occupies open chromatin regions of active genes, including enhancers and non-CpG islands

    Get PDF
    Additional file 1. Fig. S1: Analysis of CFP1 binding at individual loci and CpG islands (CGIs). (A-B) Analysis of CFP1 binding at the human α-globin locus in expressing and non-expressing cells. (A) Real-Time PCR analysis of immunoprecipitated chromatin using CFP1 antibody in human erythroblasts (red) and B-lymphocytes (blue). The y-axis represents enrichment over the input DNA, normalised to a control sequence in the human 18S gene. The x-axis represents the positions of Taqman probes used. The coding sequence is represented by the three exons (Promoter/Ex1, Ex2, Ex3) of the α-globin genes. 218 and hBact denote control sequences adjacent to the CpG islands of the human LUC7L (218) and ACTB promoters. Error bars correspond to ± 1 SD from at least two independent ChIPs. (B) Real-Time PCR analysis of immunoprecipitated chromatin using the CFP1 antibody indicated in humanised erythroblasts (normal, +MCS-R2 (left) and mutant, MCS-R2 (right). The y-axis represents enrichment over the input DNA, normalised to a control sequence in the mouse GAPDH gene. CpG Act denotes additional control sequence at the CGI of the mouse ACTB gene. The amplicons highlighted in red represent deleted regions in the humanised mice, for which no PCR signal is observed. Error bars correspond to ± 1 SD from at least two independent ChIPs. (C) CFP1 ChIP signal intensity in the top 200 peaks, by antibody and by cell type. Abcam, ab56035 antibody. Roeder, main antibody used in this study. (D) Analysis of CGI (green) and non-CGI (blue) transcription start sites (1-kb window, centred on TSS). Gene symbols shown with CpG content of individual loci in parentheses. Greek letters represent individual globin genes. Fig. S2: Peak overlaps of CFP1 and marks of active and repressed chromatin in transcription start sites (TSSs). Peaks were detected by MACS2. Venn diagrams show that CFP1 peaks within 1-kb of TSSs are strongly associated with H3K4me3 histone mark and poorly associated with H3K27me3 repressive histone mark. Cell types are (A) ERY and (B) EBV. Public data sets: * NCBI GEO GSE36985, ** NCBI GEO GSE50893. Fig. S3: UCSC tracks showing CFP1 and other ChIP signals in gene loci in erythroblasts (ERY) and EBV-transformed B-lymphoblasts (EBV). Hg38 coordinates for multiple genes, CpG islands (CGI, green boxes), and putative regulatory regions (blue boxes) are shown. CFP1 signals are shown in dark reds, inputs in grey, histone H3 signals in blues and open chromatin marks in greens. All ChIP pileups are scaled to 1x coverage genome-wide and shown in a range 0–50, except CFP1 (Roeder) is shown with extended range and H3K27me3 graphs scaled by 2x. (A) Tissue-specific binding of CFP1 to CGI promoters of tissue-specifically expressed genes. Left (chr16), CGI promoters of active genes in alpha globin locus are CFP1-bound in ERY, and unbound in EBV. Flanking regions are included, with known tissue-specific enhancers. Right (chr6), first seven exons of IRF4 locus, active in EBV and inactive in ERY, with CFP1 binding to CGI promoter in EBV only. (B) CGI promoters of housekeeping genes are CFP1 bound and unmarked by H3K27me3. Left (chr7), ACTB locus. Right (chr16), LUC7L locus. (C) CGI promoter of RHBDF1 locus (chr16) has H3K27me3 mark and the absence of CFP1 binding in both ERY and EBV. Fig. S4: Western blot analysis of CGBP (CFP1) expression in mouse and human erythroid and human lymphoid cell types. Whole cell extracts (20 µg) were loaded in each lane (1) mouse ES, (2) U-MEL, (3) I-MEL, (4) mouse primary erythroblasts and (5) human primary T lymphocytes and (6) human primary erythroblasts and separated on a 10% SDS-polyacrylamide gel. CFP1 antibody was used at a 1:1000 dilution. Fig. S5: Similar cell type-specific CFP1 read depth at CGI TSS of HBA1 gene and non-CGI TSS of HBB gene. Upper two tracks use the main antibody, and second two tracks use the commercial antibody. Coordinates are from the hg38 human genome build. Read depths are averaged in 50 bp bins and normalised to 1x genome-wide coverage. Blue boxes, known regulatory regions; green box, CGI. Fig. S6: Distribution of TrxG components in erythroid cells. Green indicates CGI and blue indicates other putative regulatory regions. All loci transcribed right to left. Pileups are shown scaled to 1x genome coverage, with full scale 0–50x depth. (A) Housekeeping genes ACTB, left (chr7), and LUC7L, right (chr16). (B) β-globin locus (chr11), (C) Non-expressed RHBDF1 locus (chr16). Fig. S7: Overlap of TrxG subunit ChIP peaks in a high-confidence subset of regions. SET1A complexes are represented by CFP1-SET1A colocalisation. MLL1/2 complexes are represented by Menin, and MLL3/4 complexes are represented by UTX, respectively. HCF1 is found in SET1A/B and MLL1/2 complexes, and RBBP5 is a member of SET1A/B and MLL1/2/3/4 complexes. Red outline (4220 peaks) shows strong colocalisation of Menin and CFP1-SET1A, accounting for the vast majority (99.5%) of 4242 CFP1-SET1A and half (50.0%) of 8432 Menin peak regions. Majority (87.0%, 2089/2400 peaks) of HCF1 (blue region) is accounted for by approximately half (49.5%, 2089/4220) of regions of Menin-SET1A-CFP1 colocalisation. Regions where either SET1A-CFP1 or Menin or both are colocalised with HCF1 (blue dashed line) accounts for nearly all (99.6%, 2390/2400) HCF1 regions, suggesting that HCF1 bound to DNA is primarily present as part of SET1A/B or MLL1/2 complexes. Fig. S8: Chromatin accessibility in TSSs and enhancers in erythroid cells as measured by ATAC-seq and DNase-seq. 1x-normalised, input-subtracted signals from ATAC-seq and DNase were averaged in a 2-kb window about TSSs and putative enhancers. Z-score transformed values for ATAC-seq and DNase-seq at a given locus were averaged. Fig. S9: Relationship of CFP1 signal to three predictive factors in top-decile open chromatin regions. A linear combination of CpG density and SET1A and H3K4me3 ChIP signals explains a substantial fraction of variation in CFP1 ChIP signal. Table S1: Bias of CFP1 for CGI TSSs in cell types and gene classes. Table S2: Bias of CFP1 for housekeeping gene TSSs. Table S3: Motifs associated with CFP1 peaks. Table S4: Dependence of CFP1 ChIP signal in erythroid cells on covariates putatively associated with its binding. Table S5: Analysis of variance of CFP1 signal in top-decile open chromatin regions surrounding TSSs and putative enhancers
    corecore