Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Genomics ; 111(6): 1713-1719, 2019 12.
Article in English | MEDLINE | ID: mdl-30503747

ABSTRACT

Mammalian imprinted domains are regulated through small genomic regions termed Imprinting Control Regions (ICRs). In the current study, the evolution patterns of the ICRs of Peg3 and H19-imprinted domains were analyzed using the genomic sequences derived from a large number of mammals. The results indicated that multiple YY1 and CTCF binding sites are localized within the Peg3 and H19-ICR in all the mammals tested. The numbers of YY1 and CTCF binding sites are variable among individual species, yet positively correlate with the presence of tandem repeats within the Peg3 and H19-ICRs. Thus, multiple YY1 and CTCF binding sites within the respective ICRs may have been maintained through tandem repeats/duplications. The unit lengths of tandem repeats are also non-random and locus-specific, 140 and 400 bp for the Peg3 and H19-ICRs. Overall, both Peg3 and H19-ICRs may have co-evolved with two unique features, multiple transcription factor binding sites and tandem repeats.


Subject(s)
Evolution, Molecular , Genetic Loci , Genomic Imprinting , Kruppel-Like Transcription Factors/genetics , RNA, Long Noncoding/genetics , Animals , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Humans , Kruppel-Like Transcription Factors/biosynthesis , RNA, Long Noncoding/biosynthesis , Response Elements
2.
Nucleic Acids Res ; 43(22): 10689-99, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26338779

ABSTRACT

Many imprinted genes are often epigenetically affected in human cancers due to their functional linkage to insulin and insulin-like growth factor signaling pathways. Thus, the current study systematically characterized the epigenetic instability of imprinted genes in multiple human cancers. First, the survey results from TCGA (The Cancer Genome Atlas) revealed that the expression levels of the majority of imprinted genes are downregulated in primary tumors compared to normal cells. These changes are also accompanied by DNA methylation level changes in several imprinted domains, such as the PEG3, MEST and GNAS domains. Second, these DNA methylation level changes were further confirmed manually using several sets of cancer DNA. According to the results, the Imprinting Control Regions of the PEG3, MEST and GNAS domains are indeed affected in breast, lung and ovarian cancers. This DNA methylation survey also revealed that evolutionarily conserved cis-regulatory elements within these imprinted domains are very variable in both normal and cancer cells. Overall, this study highlights the epigenetic instability of imprinted domains in human cancers and further suggests its potential use as cancer biomarkers.


Subject(s)
Gene Expression Regulation, Neoplastic , Genomic Imprinting , Neoplasms/genetics , DNA Methylation , Humans , Regulatory Elements, Transcriptional
3.
Anal Biochem ; 494: 82-4, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26617129

ABSTRACT

Mammalian genomes contain many unknown alternative first exons and promoters. Thus, we have modified the existing 5'RACE (5' rapid amplification of cDNA ends) approach into a next-generation sequencing (NGS)-based new protocol that can identify these alternative promoters. This protocol has incorporated two main ideas: (i) 5'RACE starting from the known second exons of genes and (ii) NGS-based sequencing of the subsequent cDNA products. This protocol also provides a bioinformatics strategy that processes the sequence reads from NGS runs. This protocol has successfully identified several alternative promoters for an imprinted gene, PEG3. Overall, this NGS-based 5'RACE protocol is a sensitive and reliable method for detecting low-abundant transcripts and promoters.


Subject(s)
DNA, Complementary/chemistry , High-Throughput Nucleotide Sequencing , Animals , DNA, Complementary/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Promoter Regions, Genetic , Sequence Analysis, DNA
4.
Proc Natl Acad Sci U S A ; 110(1): 276-81, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23248301

ABSTRACT

The Th2 locus control region (LCR) has been shown to be important in efficient and coordinated cytokine gene regulation during Th2 cell differentiation. However, the molecular mechanism for this is poorly understood. To study the molecular mechanism of the Th2 LCR, we searched for proteins binding to it. We discovered that transcription factor YY1 bound to the LCR and the entire Th2 cytokine locus in a Th2-specific manner. Retroviral overexpression of YY1 induced Th2 cytokine expression. CD4-specific knockdown of YY1 in mice caused marked reduction in Th2 cytokine expression, repressed chromatin remodeling, decreased intrachromosomal interactions, and resistance in an animal model of asthma. YY1 physically associated with GATA-binding protein-3 (GATA3) and is required for GATA3 binding to the locus. YY1 bound to the regulatory elements in the locus before GATA3 binding. Thus, YY1 cooperates with GATA3 and is required for regulation of the Th2 cytokine locus and Th2 cell differentiation.


Subject(s)
Asthma/immunology , Cell Differentiation/immunology , Th2 Cells/immunology , YY1 Transcription Factor/immunology , Animals , Chromatin Assembly and Disassembly/immunology , Chromatin Immunoprecipitation , Cytokines , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , GATA3 Transcription Factor/metabolism , Immunoblotting , Immunoprecipitation , Locus Control Region/genetics , Locus Control Region/immunology , Luciferases , Mice , Mice, Transgenic , Oligonucleotides/genetics , Real-Time Polymerase Chain Reaction , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
5.
Genomics ; 105(2): 108-15, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25451679

ABSTRACT

Aebp2 encodes an evolutionarily conserved zinc finger protein that has not been well studied so far, yet recent studies indicated that this gene is closely associated with the Polycomb Repressive Complex 2 (PRC2). Thus, the current study characterized the basic aspects of this gene, including alternative promoters and protein isoforms. According to the results, Aebp2 is controlled through three alternative promoters, deriving three different transcripts encoding the embryonic (32 kDa) and somatic (52 kDa) forms. Chromatin Immuno-Precipitation (ChIP) experiments revealed that AEBP2 binds to its own promoter as well as the promoters of Jarid2 and Snai2. While the embryonic form acts as a transcriptional repressor for Snai2, the somatic form functions as a transcriptional activator for Jarid2, Aebp2 and Snai2. Cell migration assays also demonstrated that the Aebp2 somatic form has an enhancing activity in cell migration. This is consistent with the functional association of Aebp2 with migratory neural crest cells. These results suggest that the two protein isoforms of AEBP2 may have opposite functions for the PcG target genes, and may play significant roles in cell migration during development.


Subject(s)
Cell Movement/physiology , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Transcriptional Activation , Animals , Base Sequence , Gene Expression Regulation, Developmental , Mice , Molecular Sequence Data , Neural Crest/metabolism , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Protein Isoforms/metabolism , Repressor Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism
6.
Genomics ; 104(6 Pt B): 538-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25277721

ABSTRACT

In the current study, we have used HT-TREBS to individually analyze the DNA methylation pattern of 4799 IAP LTR retrotransposons in embryonic stem, somatic and Neuro2A cells. According to the results, half of the loci within this family show constant methylation patterns between the three cell types whereas the remaining half display variable levels of methylation. About half of the variably methylated IAP LTRs tend to be hypomethylated in ES cells, and nearly all in this group are hypomethylated in Neuro2A cells. The observed hypomethylation in both cell types occur in a non-uniform, locus-specific manner and to various degrees of severity, with some of them being easily detectible by COBRA. Overall, this study demonstrates the feasibility of HT-TREBS to study DNA methylation changes at retrotransposons in a locus-specific manner in multiple cell types and further suggests the potential utility of this technique in developing epigenetic biomarkers for tracking disease progression.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Neoplasms/genetics , Retroelements , Terminal Repeat Sequences , Animals , Cell Line, Tumor , Genome , Mice , Mice, Inbred C57BL , Neoplasms/metabolism , Sequence Analysis, DNA/methods
7.
Hum Mol Genet ; 21(12): 2677-87, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22394678

ABSTRACT

The imprinting and transcription of the 500 kb genomic region surrounding the mouse Peg3 is predicted to be regulated by the Peg3-differentially methylated region (DMR). In the current study, this prediction was tested using a mutant mouse line lacking this potential imprinting control region (ICR). At the organismal level, paternal and maternal transmission of this knockout (KO) allele caused either reduced or increased growth rates in the mouse, respectively. In terms of the imprinting control, the paternal transmission of the KO allele resulted in bi-allelic expression of the normally maternally expressed Zim2, whereas the maternal transmission switched the transcriptionally dominant allele for Zfp264 (paternal to maternal). However, the allele-specific DNA methylation patterns of the DMRs of Peg3, Zim2 and Zim3 were not affected in the mice that inherited the KO allele either paternally or maternally. In terms of the transcriptional control, the paternal transmission caused a dramatic down-regulation in Peg3 expression, but overall up-regulation in the other nearby imprinted genes. Taken together, deletion of the Peg3-DMR caused global changes in the imprinting and transcription of the Peg3 domain, confirming that the Peg3-DMR is an ICR for this imprinted domain.


Subject(s)
DNA Methylation , Genomic Imprinting , Kruppel-Like Transcription Factors/genetics , Alleles , Animals , Animals, Newborn , Binding Sites/genetics , Blotting, Western , Brain/metabolism , Crosses, Genetic , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Gene Expression Profiling , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Zinc Fingers/genetics
8.
Hum Mol Genet ; 20(7): 1353-62, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21233130

ABSTRACT

Zfp42/Rex1 (reduced expression gene 1) is a well-known stem-cell marker that has been duplicated from YY1 in the eutherian lineage. In the current study, we characterized the in vivo roles of Rex1 using a mutant mouse line disrupting its transcription. In contrast to the ubiquitous expression of YY1, Rex1 is expressed only during spermatogenesis and early embryogenesis and also in a very limited area of the placenta. Yet, the gene dosage of Rex1 is very critical for the survival of the late-stage embryos and neonates. This delayed phenotypic consequence suggests potential roles for Rex1 in establishing and maintaining unknown epigenetic modifications. Consistently, Rex1-null blastocysts display hypermethylation in the differentially methylated regions (DMRs) of Peg3 and Gnas imprinted domains, which are known to contain YY1 binding sites. Further analyses confirmed in vivo binding of Rex1 only to the unmethylated allele of these two regions. Thus, Rex1 may function as a protector for these DMRs against DNA methylation. Overall, the functional connection of Rex1 to genomic imprinting represents another case where newly made genes have co-evolved with lineage-specific phenomena.


Subject(s)
Alleles , Blastocyst/metabolism , DNA Methylation/physiology , Genomic Imprinting/physiology , Transcription Factors/metabolism , Animals , Blastocyst/cytology , Chromogranins , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Mutant Strains , Transcription Factors/genetics
9.
Nucleic Acids Res ; 39(17): e116, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21724599

ABSTRACT

The identification of genome-wide cis-regulatory modules (CRMs) and characterization of their associated epigenetic features are fundamental steps toward the understanding of gene regulatory networks. Although integrative analysis of available genome-wide information can provide new biological insights, the lack of novel methodologies has become a major bottleneck. Here, we present a comprehensive analysis tool called combinatorial CRM decoder (CCD), which utilizes the publicly available information to identify and characterize genome-wide CRMs in a species of interest. CCD first defines a set of the epigenetic features which is significantly associated with a set of known CRMs as a code called 'trace code', and subsequently uses the trace code to pinpoint putative CRMs throughout the genome. Using 61 genome-wide data sets obtained from 17 independent mouse studies, CCD successfully catalogued ∼12 600 CRMs (five distinct classes) including polycomb repressive complex 2 target sites as well as imprinting control regions. Interestingly, we discovered that ∼4% of the identified CRMs belong to at least two different classes named 'multi-functional CRM', suggesting their functional importance for regulating spatiotemporal gene expression. From these examples, we show that CCD can be applied to any potential genome-wide datasets and therefore will shed light on unveiling genome-wide CRMs in various species.


Subject(s)
Algorithms , Regulatory Elements, Transcriptional , Animals , Genomics/methods , Histones/metabolism , Mice , Software , Transcription Factors/metabolism
10.
Nucleic Acids Res ; 37(9): 2940-50, 2009 May.
Article in English | MEDLINE | ID: mdl-19293275

ABSTRACT

AEBP2 is a zinc finger protein that has been shown to interact with the mammalian Polycomb Repression Complex 2 (PRC2). In the current study, we characterized this unknown protein and tested its potential targeting roles for the PRC2. AEBP2 is an evolutionarily well-conserved gene that is found in the animals ranging from flying insects to mammals. The transcription of mammalian AEBP2 is driven by two alternative promoters and produces at least two isoforms of the protein. These isoforms show developmental stage-specific expression patterns: the adult-specific larger form (51 kDa) and the embryo-specific smaller form (32 kDa). The AEBP2 protein binds to a DNA-binding motif with an unusual bipartite structure, CTT(N)15-23cagGCC with lower-case being less critical. A large fraction of AEBP2's target loci also map closely to the known target loci of the PRC2. In fact, many of these loci are co-occupied by the two proteins, AEBP2 and SUZ12. This suggests that AEBP2 is most likely a targeting protein for the mammalian PRC2 complex.


Subject(s)
DNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Alternative Splicing , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Chromatin Immunoprecipitation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Exons , Insecta/genetics , Male , Mice , Molecular Sequence Data , Nuclear Proteins , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Protein Isoforms/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Sequence Alignment , Vertebrates/genetics
11.
Nucleic Acids Res ; 37(6): 2003-13, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19208640

ABSTRACT

We have developed a new bioinformatics approach called ECMFinder (Evolutionary Conserved Motif Finder). This program searches for a given DNA motif within the entire genome of one species and uses the gene association information of a potential transcription factor-binding site (TFBS) to screen the homologous regions of a second and third species. If multiple species have this potential TFBS in homologous positions, this program recognizes the identified TFBS as an evolutionary conserved motif (ECM). This program outputs a list of ECMs, which can be uploaded as a Custom Track in the UCSC genome browser and can be visualized along with other available data. The feasibility of this approach was tested by searching the genomes of three mammals (human, mouse and cow) with the DNA-binding motifs of YY1 and CTCF. This program successfully identified many clustered YY1- and CTCF-binding sites that are conserved among these species but were previously undetected. In particular, this program identified CTCF-binding sites that are located close to the Dlk1, Magel2 and Cdkn1c imprinted genes. Individual ChIP experiments confirmed the in vivo binding of the YY1 and CTCF proteins to most of these newly discovered binding sites, demonstrating the feasibility and usefulness of ECMFinder.


Subject(s)
DNA-Binding Proteins/metabolism , Genomics/methods , Regulatory Elements, Transcriptional , Repressor Proteins/metabolism , Software , YY1 Transcription Factor/metabolism , Algorithms , Animals , Base Sequence , Binding Sites , CCCTC-Binding Factor , Cattle , Conserved Sequence , Genome, Human , Humans , Mice , Molecular Sequence Data
12.
Nucleic Acids Res ; 37(17): 5656-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19628663

ABSTRACT

Unusual clusters of YY1 binding sites are located within several differentially methylated regions (DMRs), including Xist, Nespas and Peg3, which all become methylated during oogenesis. In this study, we performed conditional YY1 knockdown (KD) to investigate YY1's roles in DNA methylation of these DMRs. Reduced levels of YY1 during spermatogenesis did not cause any major change in these DMRs although the same YY1 KD caused hypermethylation in these DMRs among a subset of aged mice. However, YY1 KD during oogenesis resulted in the loss of DNA methylation on Peg3 and Xist, but there were no changes on Nespas and H19. Continued YY1 KD from oogenesis to the blastocyst stage caused further loss in DNA methylation on Peg3. Consequently, high incidents of lethality were observed among embryos that had experienced the reduced levels of YY1 protein. Overall, the current study suggests that YY1 likely plays a role in the de novo DNA methylation of the DMRs of Peg3 and Xist during oogenesis and also in the maintenance of unmethylation status of these DMRs during spermatogenesis.


Subject(s)
DNA Methylation , Kruppel-Like Transcription Factors/genetics , Oogenesis/genetics , RNA, Untranslated/genetics , Spermatogenesis/genetics , YY1 Transcription Factor/physiology , Animals , Blastocyst/metabolism , Female , Gene Knockdown Techniques , Germ Cells/growth & development , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Transgenic , RNA, Long Noncoding , RNA, Untranslated/metabolism , YY1 Transcription Factor/genetics
13.
PLoS One ; 16(5): e0252354, 2021.
Article in English | MEDLINE | ID: mdl-34048454

ABSTRACT

Peg3 (Paternally expressed gene 3) is an imprinted gene encoding a DNA-binding protein that is a well-known transcriptional repressor. Previous studies have shown that the mutant phenotypes of Peg3 are associated with the over-expression of genes involved in lipid metabolism. In the current study, we investigated four potential downstream genes of Peg3, which were identified through ChIP-seq data: Acly, Fasn, Idh1, and Hmgcr. In vivo binding of PEG3 to the promoter region of these key genes involved in lipogenesis was subsequently confirmed through individual ChIP experiments. We observed the opposite response of Acly expression levels against the variable gene dosages of Peg3, involving 0x, 1x, and 2x Peg3. This suggests the transcriptional repressor role of Peg3 in the expression levels of Acly. Another set of analyses showed a sex-biased response in the expression levels of Acly, Fasn, and Idh1 against 0x Peg3 with higher levels in female and lower levels in male mammary glands. These results overall highlight that Peg3 may be involved in regulating the expression levels of several key genes in adipogenesis.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Lipogenesis/physiology , Mammary Glands, Human/metabolism , Animals , Female , Genomic Imprinting , Humans , Kruppel-Like Transcription Factors/genetics , Lipogenesis/genetics , Male , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism
14.
Hum Mol Genet ; 17(3): 391-401, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17977899

ABSTRACT

The YY1 transcription factor is predicted to control several imprinted domains, including the Peg3, Gnas and Xist/Tsix regions. To test this possibility, we have used RNA interference strategies to generate transgenic mouse lines that express reduced levels of the cellular YY1 protein. As predicted, lowering YY1 levels resulted in global expression changes in these three imprinted domains. In neonatal brains, most imprinted genes of the Peg3 domain were up-regulated. In the Gnas domain, Nespas was down-regulated, whereas three other imprinted transcripts were up-regulated, including Nesp, Gnasxl and Exon1A. In the Xist/Tsix domain, no obvious change was detected in the expression levels of the two genes in female mice. However, male mice showed low-level coordinated, up- and down-regulation of Xist and Tsix, respectively, suggesting potential de-repression of Xist in a subset of male cell populations. YY1 knockdown also changed the methylation levels at the imprinting control regions (ICRs) of these domains in a target-specific manner. In addition, breeding experiments indicated that the birth weights of 20% of the transgenic females were much lower than those of normal female littermates. We surmise that this gender-specific outcome is caused by the YY1 knockdown effect on the Xist locus of females. In sum, these results demonstrate that YY1 indeed functions as a trans factor for transcriptional regulation and DNA methylation of these imprinted domains in vivo.


Subject(s)
Genomic Imprinting , YY1 Transcription Factor/antagonists & inhibitors , YY1 Transcription Factor/genetics , Animals , Animals, Newborn , Base Sequence , Birth Weight/genetics , Brain/metabolism , Chromogranins , DNA Methylation , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Transgenic , Nucleic Acid Conformation , Pregnancy , RNA/chemistry , RNA/genetics , RNA Interference , RNA, Long Noncoding , RNA, Untranslated/genetics , Sex Characteristics , YY1 Transcription Factor/deficiency
15.
Genomics ; 93(2): 152-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18950698

ABSTRACT

The DNA-binding sites of YY1 located within two newly identified downstream genes of YY1, Peg3 (GGCGCCATnTT) and Xist (CCGCCATnTT), are longer than the known motif of YY1 (CGCCATnTT). Gel shift assays indicated that these DNA-binding sites are previously unnoticed, longer motifs of YY1. Independent DNA-binding motif studies further confirmed that YY1 recognizes a longer sequence (GCCGCCATTTTG) as its consensus motif. This longer motif exhibited higher affinity to the YY1 protein than the known motif. Another DNA-binding motif study was also performed using a protein containing three amino acid substitutions in the first zinc finger unit of YY1, mimicking the zinc finger domain of pho (a drosophila homologue of YY1). The substitutions cause the weakening of DNA-binding specificity at both 5'- and 3'-sides of the longer motif, yielding a much shorter sequence (GCCAT) as a consensus motif. This indicates that the intact first finger unit is required for recognition of the longer motif of YY1. Also, this shortening suggests that the DNA recognition by YY1 is mediated through the concerted, but not modular, contribution by its four zinc finger units.


Subject(s)
DNA/metabolism , YY1 Transcription Factor/chemistry , YY1 Transcription Factor/genetics , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites/genetics , Cattle , Consensus Sequence , DNA/genetics , Dogs , Humans , Macaca mulatta , Mammals/genetics , Mice , Molecular Sequence Data , Pan troglodytes , Protein Structure, Tertiary/genetics , Rabbits , Sequence Homology, Amino Acid , YY1 Transcription Factor/metabolism , Zinc Fingers/genetics
16.
BMC Genomics ; 10: 576, 2009 Dec 03.
Article in English | MEDLINE | ID: mdl-19958529

ABSTRACT

BACKGROUND: YY1 is an epigenetic regulator for a large number of mammalian genes. While performing genome-wide YY1 binding motif searches, we discovered that the olfactory receptor (OLFR) genes have an unusual cluster of YY1 binding sites within their coding regions. The statistical significance of this observation was further analyzed. RESULTS: About 45% of the olfactory genes in the mouse have a range of 4-8 YY1 binding sites within their respective 1 kb coding regions. Statistical analyses indicate that this enrichment of YY1 motifs has likely been driven by unknown selection pressures at the DNA level, but not serendipitously by some peptides enriched within the OLFR genes. Similar patterns are also detected in the OLFR genes of all mammals analyzed, but not in the OLFR genes of the fish lineage, suggesting a mammal-specific phenomenon. CONCLUSION: YY1, or YY1-related transcription factors, may help regulate olfactory receptor genes. Furthermore, the protein-coding regions of vertebrate genes can contain cis-regulatory elements for transcription factor binding as well as codons.


Subject(s)
Conserved Sequence , DNA/genetics , DNA/metabolism , Receptors, Odorant/genetics , Regulatory Sequences, Nucleic Acid/genetics , YY1 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites , Cattle , Computational Biology , Humans , Mice
17.
BMC Mol Biol ; 10: 85, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19712462

ABSTRACT

BACKGROUND: The transcription factor Yin Yang 1 (YY1) is a ubiquitously expressed, multifunctional protein that controls a large number of genes and biological processes in vertebrates. As a general transcription factor, the proper levels of YY1 protein need to be maintained for the normal function of cells and organisms. However, the mechanism for the YY1 homeostasis is currently unknown. RESULTS: The current study reports that the YY1 gene locus of all vertebrates contains a cluster of its own DNA-binding sites within the 1st intron. The intact structure of these DNA-binding sites is absolutely necessary for transcriptional activity of the YY1 promoter. In an inducible cell line system that over-expresses an exogenous YY1 gene, the overall increased levels of YY1 protein caused a reduction in transcription levels of the endogenous YY1 gene. Reversion to the normal levels of YY1 protein restored the transcriptional levels of the endogenous YY1 to normal levels. This homeostatic response was also mediated through its cluster of YY1 binding sites. CONCLUSION: Taken together, the transcriptional level of YY1 is self-regulated through its internal DNA-binding sites. This study identifies YY1 as the first known autoregulating transcription factor in mammalian genomes.


Subject(s)
Transcriptional Activation , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , Evolution, Molecular , Homeostasis , Humans , Introns , Mice , Molecular Sequence Data , Protein Binding
18.
Nucleic Acids Res ; 35(10): 3442-52, 2007.
Article in English | MEDLINE | ID: mdl-17478514

ABSTRACT

YY1 is a DNA-binding transcription factor found in both vertebrates and invertebrates. Database searches identified 62 YY1 related sequences from all the available genome sequences ranging from flying insects to human. These sequences are characterized by high levels of sequence conservation, ranging from 66% to 100% similarity, in the zinc finger DNA-binding domain of the predicted proteins. Phylogenetic analyses uncovered duplication events of YY1 in several different lineages, including flies, fish and mammals. Retroposition is responsible for generating one duplicate in flies, PHOL from PHO, and two duplicates in placental mammals, YY2 and Reduced Expression 1 (REX1) from YY1. DNA-binding motif studies have demonstrated that YY2 still binds to the same consensus sequence as YY1 but with much lower affinity. In contrast, REX1 binds to DNA motifs divergent from YY1, but the binding motifs of REX1 and YY1 share some similarity at their core regions (5'-CCAT-3'). This suggests that the two duplicates, YY2 and REX1, although generated through similar retroposition events have undergone different selection schemes to adapt to new roles in placental mammals. Overall, the conservation of YY2 and REX1 in all placental mammals predicts that each duplicate has co-evolved with some unique features of eutherian mammals.


Subject(s)
DNA-Binding Proteins/chemistry , Evolution, Molecular , Retroelements , YY1 Transcription Factor/chemistry , YY1 Transcription Factor/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , DNA-Binding Proteins/genetics , Diptera/genetics , Electrophoretic Mobility Shift Assay , Gene Duplication , Genomics , Humans , Invertebrates/genetics , Kruppel-Like Transcription Factors/chemistry , Mammals/genetics , Molecular Sequence Data , Sequence Homology, Amino Acid , Transcription Factors/chemistry , Vertebrates/genetics , YY1 Transcription Factor/classification
19.
Methods Mol Biol ; 1908: 219-228, 2019.
Article in English | MEDLINE | ID: mdl-30649731

ABSTRACT

High-throughput targeted repeat element bisulfite sequencing (HT-TREBS) is designed to assay the methylation level of individual retrotransposon loci of a targeted family, in a locus-specific manner, and on a genome-wide scale. Briefly, genomic DNA is sheared and ligated to Ion Torrent A adaptors (with methylated cytosines), followed by bisulfite-conversion, and amplification with primers designed to bind the targeted retrotransposon. Since the primers carry the Ion Torrent P1 adaptor as a 5'-extension, the amplified library is ready to be size-selected and sequenced on a next-generation sequencing platform. Once sequenced, each retrotransposon is mapped to a particular genomic locus, which is achieved through ensuring at least a 10-bp overlap with flanking unique sequence, followed by the calculation of methylation levels of the mapped retrotransposon using a BiQ Analyzer HT. A complete protocol for library construction as well as the bioinformatics for HT-TREBS is described in this chapter.


Subject(s)
DNA Methylation , High-Throughput Nucleotide Sequencing/methods , Retroelements , Sequence Analysis, DNA/methods , Epigenomics/methods , Genetic Loci , Humans , Sulfites
20.
PLoS One ; 14(10): e0224287, 2019.
Article in English | MEDLINE | ID: mdl-31639176

ABSTRACT

The parental allele specificity of mammalian imprinted genes has been evolutionarily well conserved, although its functional constraints and associated mechanisms are not fully understood. In the current study, we generated a mouse mutant with switched active alleles driving the switch from paternal-to-maternal expression for Peg3 and the maternal-to-paternal expression for Zim1. The expression levels of Peg3 and Zim1, but not the spatial expression patterns, within the brain showed clear differences between wild type and mutant animals. We identified putative enhancers localized upstream of Peg3 that displayed allele-biased DNA methylation, and that also participate in allele-biased chromosomal conformations with regional promoters. Most importantly, these data suggest for the first time that long-distance enhancers may contribute to allelic expression within imprinted domains through allele-biased interactions with regional promoters.


Subject(s)
Alleles , Enhancer Elements, Genetic/genetics , Genomic Imprinting , Kruppel-Like Transcription Factors/genetics , Animals , Chromosomes, Mammalian/genetics , DNA Methylation , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Enzymologic/genetics , Male , Mice , Mutation , Nerve Tissue Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL