42 results on '"Galina N. Filippova"'
Search Results
2. Trans- and cis-acting effects of Firre on epigenetic features of the inactive X chromosome
- Author
-
He Fang, Giancarlo Bonora, Jordan P. Lewandowski, Jitendra Thakur, Galina N. Filippova, Steven Henikoff, Jay Shendure, Zhijun Duan, John L. Rinn, Xinxian Deng, William S. Noble, and Christine M. Disteche
- Subjects
Science - Abstract
Firre encodes a lncRNA involved in nuclear organization in mammals. Here, the authors find that allelic deletion of Firre on the active X chromosome (Xa) results in dose-dependent loss of histone H3K27me3 on the inactive X chromosome (Xi), along with other trans-acting effects, including disruption of the perinuclear location and minor dysregulation of gene expression.
- Published
- 2020
- Full Text
- View/download PDF
3. SMCHD1 regulates a limited set of gene clusters on autosomal chromosomes
- Author
-
Amanda G. Mason, Roderick C. Slieker, Judit Balog, Richard J. L. F. Lemmers, Chao-Jen Wong, Zizhen Yao, Jong-Won Lim, Galina N. Filippova, Enrico Ne, Rabi Tawil, Bas T. Heijmans, Stephen J. Tapscott, and Silvère M. van der Maarel
- Subjects
SMCHD1 ,FSHD ,Chromatin ,Methylation ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Background Facioscapulohumeral muscular dystrophy (FSHD) is in most cases caused by a contraction of the D4Z4 macrosatellite repeat on chromosome 4 (FSHD1) or by mutations in the SMCHD1 or DNMT3B gene (FSHD2). Both situations result in the incomplete epigenetic repression of the D4Z4-encoded retrogene DUX4 in somatic cells, leading to the aberrant expression of DUX4 in the skeletal muscle. In mice, Smchd1 regulates chromatin repression at different loci, having a role in CpG methylation establishment and/or maintenance. Methods To investigate the global effects of harboring heterozygous SMCHD1 mutations on DNA methylation in humans, we combined 450k methylation analysis on mononuclear monocytes from female heterozygous SMCHD1 mutation carriers and unaffected controls with reduced representation bisulfite sequencing (RRBS) on FSHD2 and control myoblast cell lines. Candidate loci were then evaluated for SMCHD1 binding using ChIP-qPCR and expression was evaluated using RT-qPCR. Results We identified a limited number of clustered autosomal loci with CpG hypomethylation in SMCHD1 mutation carriers: the protocadherin (PCDH) cluster on chromosome 5, the transfer RNA (tRNA) and 5S rRNA clusters on chromosome 1, the HOXB and HOXD clusters on chromosomes 17 and 2, respectively, and the D4Z4 repeats on chromosomes 4 and 10. Furthermore, minor increases in RNA expression were seen in FSHD2 myoblasts for some of the PCDHβ cluster isoforms, tRNA isoforms, and a HOXB isoform in comparison to controls, in addition to the previously reported effects on DUX4 expression. SMCHD1 was bound at DNAseI hypersensitivity sites known to regulate the PCDHβ cluster and at the chromosome 1 tRNA cluster, with decreased binding in SMCHD1 mutation carriers at the PCDHβ cluster sites. Conclusions Our study is the first to investigate the global methylation effects in humans resulting from heterozygous mutations in SMCHD1. Our results suggest that SMCHD1 acts as a repressor on a limited set of autosomal gene clusters, as an observed reduction in methylation associates with a loss of SMCHD1 binding and increased expression for some of the loci.
- Published
- 2017
- Full Text
- View/download PDF
4. CTCF Haploinsufficiency Destabilizes DNA Methylation and Predisposes to Cancer
- Author
-
Christopher J. Kemp, James M. Moore, Russell Moser, Brady Bernard, Matt Teater, Leslie E. Smith, Natalia A. Rabaia, Kay E. Gurley, Justin Guinney, Stephanie E. Busch, Rita Shaknovich, Victor V. Lobanenkov, Denny Liggitt, Ilya Shmulevich, Ari Melnick, and Galina N. Filippova
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Epigenetic alterations, particularly in DNA methylation, are ubiquitous in cancer, yet the molecular origins and the consequences of these alterations are poorly understood. CTCF, a DNA-binding protein that regulates higher-order chromatin organization, is frequently altered by hemizygous deletion or mutation in human cancer. To date, a causal role for CTCF in cancer has not been established. Here, we show that Ctcf hemizygous knockout mice are markedly susceptible to spontaneous, radiation-, and chemically induced cancer in a broad range of tissues. Ctcf+/− tumors are characterized by increased aggressiveness, including invasion, metastatic dissemination, and mixed epithelial/mesenchymal differentiation. Molecular analysis of Ctcf+/− tumors indicates that Ctcf is haploinsufficient for tumor suppression. Tissues with hemizygous loss of CTCF exhibit increased variability in CpG methylation genome wide. These findings establish CTCF as a prominent tumor-suppressor gene and point to CTCF-mediated epigenetic stability as a major barrier to neoplastic progression.
- Published
- 2014
- Full Text
- View/download PDF
5. CTCF-mediated insulation and chromatin environment modulate Car5b escape from X inactivation
- Author
-
He Fang, Ana R. Tronco, Giancarlo Bonora, Truong Nguyen, Jitendra Thakur, Joel B. Berletch, Galina N. Filippova, Steven Henikoff, Jay Shendure, William S. Noble, Christine M. Disteche, and Xinxian Deng
- Subjects
Article - Abstract
BackgroundThe number and escape levels of genes that escape X chromosome inactivation (XCI) in female somatic cells vary among tissues and cell types, potentially contributing to specific sex differences. Here we investigate the role of CTCF, a master chromatin conformation regulator, in regulating escape from XCI. CTCF binding profiles and epigenetic features were systematically examined at constitutive and facultative escape genes using mouse allelic systems to distinguish the inactive X (Xi) and active X (Xa) chromosomes.ResultsWe found that escape genes are located inside domains flanked by convergent arrays of CTCF binding sites, consistent with the formation of loops. In addition, strong and divergent CTCF binding sites often located at the boundaries between escape genes and adjacent neighbors subject to XCI would help insulate domains. Facultative escapees show clear differences in CTCF binding dependent on their XCI status in specific cell types/tissues. Concordantly, deletion but not inversion of a CTCF binding site at the boundary between the facultative escape geneCar5band its silent neighborSiah1bresulted in loss ofCar5bescape. Reduced CTCF binding and enrichment of a repressive mark overCar5bin cells with a boundary deletion indicated loss of looping and insulation. In mutant lines in which either the Xi-specific compact structure or its H3K27me3 enrichment was disrupted, escape genes showed an increase in gene expression and associated active marks, supporting the roles of the 3D Xi structure and heterochromatic marks in constraining levels of escape.ConclusionOur findings indicate that escape from XCI is modulated both by looping and insulation of chromatin via convergent arrays of CTCF binding sites and by compaction and epigenetic features of the surrounding heterochromatin.
- Published
- 2023
6. Trans- and cis-acting effects of Firre on epigenetic features of the inactive X chromosome
- Author
-
Jordan P. Lewandowski, He Fang, Christine M. Disteche, Xinxian Deng, Galina N. Filippova, John L. Rinn, Steven Henikoff, Zhijun Duan, Giancarlo Bonora, William Stafford Noble, Jitendra K. Thakur, and Jay Shendure
- Subjects
Male ,0301 basic medicine ,General Physics and Astronomy ,Epigenesis, Genetic ,Histones ,0302 clinical medicine ,X Chromosome Inactivation ,Gene expression ,Transgenes ,X chromosome ,Genome ,Multidisciplinary ,biology ,Polycomb Repressive Complex 2 ,Chromatin ,Up-Regulation ,Cell biology ,Histone ,Female ,RNA, Long Noncoding ,Epigenetics ,DNA, Complementary ,X Chromosome ,Science ,Methylation ,Chromatin structure ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Animals ,Gene ,Alleles ,Cell Nucleus ,Nuclear organization ,Base Sequence ,Lysine ,RNA ,General Chemistry ,Mice, Inbred C57BL ,Gene Ontology ,030104 developmental biology ,Genetic Loci ,Long non-coding RNAs ,biology.protein ,Ectopic expression ,Gene Deletion ,030217 neurology & neurosurgery - Abstract
Firre encodes a lncRNA involved in nuclear organization. Here, we show that Firre RNA expressed from the active X chromosome maintains histone H3K27me3 enrichment on the inactive X chromosome (Xi) in somatic cells. This trans-acting effect involves SUZ12, reflecting interactions between Firre RNA and components of the Polycomb repressive complexes. Without Firre RNA, H3K27me3 decreases on the Xi and the Xi-perinucleolar location is disrupted, possibly due to decreased CTCF binding on the Xi. We also observe widespread gene dysregulation, but not on the Xi. These effects are measurably rescued by ectopic expression of mouse or human Firre/FIRRE transgenes, supporting conserved trans-acting roles. We also find that the compact 3D structure of the Xi partly depends on the Firre locus and its RNA. In common lymphoid progenitors and T-cells Firre exerts a cis-acting effect on maintenance of H3K27me3 in a 26 Mb region around the locus, demonstrating cell type-specific trans- and cis-acting roles of this lncRNA., Firre encodes a lncRNA involved in nuclear organization in mammals. Here, the authors find that allelic deletion of Firre on the active X chromosome (Xa) results in dose-dependent loss of histone H3K27me3 on the inactive X chromosome (Xi), along with other trans-acting effects, including disruption of the perinuclear location and minor dysregulation of gene expression.
- Published
- 2020
7. Small noncoding RNAs in FSHD2 muscle cells reveal both DUX4- and SMCHD1-specific signatures
- Author
-
Jong-Won Lim, Stephen J. Tapscott, Galina N. Filippova, Chao-Jen Wong, Rabi Tawil, Daniel G. Miller, Silvère M. van der Maarel, and Zizhen Yao
- Subjects
0301 basic medicine ,Small RNA ,Chromosomal Proteins, Non-Histone ,Muscle Fibers, Skeletal ,Biology ,Myoblasts ,03 medical and health sciences ,RNA, Transfer ,DUX4 ,microRNA ,Gene expression ,Genetics ,Humans ,Myocyte ,Molecular Biology ,Genetics (clinical) ,Homeodomain Proteins ,Regulation of gene expression ,RNA, Ribosomal, 5S ,Reproducibility of Results ,RNA ,Cell Differentiation ,Articles ,General Medicine ,Non-coding RNA ,Muscular Dystrophy, Facioscapulohumeral ,Cell biology ,MicroRNAs ,030104 developmental biology ,Gene Expression Regulation ,Case-Control Studies ,Mutation ,RNA, Small Untranslated - Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is caused by insufficient epigenetic repression of D4Z4 macrosatellite repeat where DUX4, an FSHD causing gene is embedded. There are two forms of FSHD, FSHD1 with contraction of D4Z4 repeat and FSHD2 with chromatin compaction defects mostly due to SMCHD1 mutation. Previous reports showed DUX4-induced gene expression changes as well as changes in microRNA expression in FSHD muscle cells. However, a genome wide analysis of small noncoding RNAs that might be regulated by DUX4 or by mutations in SMCHD1 has not been reported yet. Here, we identified several types of small noncoding RNAs including known microRNAs that are differentially expressed in FSHD2 muscle cells compared to control. Although fewer small RNAs were differentially expressed during muscle differentiation in FSHD2 cells compared to controls, most of the known myogenic microRNAs, such as miR1, miR133a and miR206 were induced in both FSHD2 and control muscle cells during differentiation. Our small RNA sequencing data analysis also revealed both DUX4- and SMCHD1-specific changes in FSHD2 muscle cells. Six FSHD2 microRNAs were affected by DUX4 overexpression in control myoblasts, whereas increased expression of tRNAs and 5S rRNAs in FSHD2 muscle cells was largely recapitulated in SMCHD1-depleted control myoblasts. Altogether, our studies suggest that the small noncoding RNA transcriptome changes in FSHD2 might be different from those in FSHD1 and that these differences may provide new diagnostic and therapeutic tools specific to FSHD2.
- Published
- 2018
8. SMCHD1 regulates a limited set of gene clusters on autosomal chromosomes
- Author
-
Galina N. Filippova, Judit Balog, Roderick C. Slieker, Enrico Ne, Richard J.L.F. Lemmers, Silvère M. van der Maarel, Rabi Tawil, Jong-Won Lim, Stephen J. Tapscott, Amanda G. Mason, Chao Jen Wong, Zizhen Yao, B.T. Heijmans, Epidemiology and Data Science, APH - Aging & Later Life, and APH - Health Behaviors & Chronic Diseases
- Subjects
0301 basic medicine ,Heterozygote ,lcsh:Diseases of the musculoskeletal system ,Chromosomal Proteins, Non-Histone ,Biology ,Methylation ,Myoblasts ,03 medical and health sciences ,0302 clinical medicine ,DUX4 ,Humans ,Orthopedics and Sports Medicine ,Molecular Biology ,Gene ,Cells, Cultured ,Genetics ,FSHD ,Autosome ,SMCHD1 ,Research ,Cell Biology ,DNA Methylation ,Molecular biology ,Muscular Dystrophy, Facioscapulohumeral ,Chromatin ,030104 developmental biology ,Chromosome 4 ,CpG site ,Genetic Loci ,Multigene Family ,DNA methylation ,Mutation ,CpG Islands ,Female ,lcsh:RC925-935 ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Background Facioscapulohumeral muscular dystrophy (FSHD) is in most cases caused by a contraction of the D4Z4 macrosatellite repeat on chromosome 4 (FSHD1) or by mutations in the SMCHD1 or DNMT3B gene (FSHD2). Both situations result in the incomplete epigenetic repression of the D4Z4-encoded retrogene DUX4 in somatic cells, leading to the aberrant expression of DUX4 in the skeletal muscle. In mice, Smchd1 regulates chromatin repression at different loci, having a role in CpG methylation establishment and/or maintenance. Methods To investigate the global effects of harboring heterozygous SMCHD1 mutations on DNA methylation in humans, we combined 450k methylation analysis on mononuclear monocytes from female heterozygous SMCHD1 mutation carriers and unaffected controls with reduced representation bisulfite sequencing (RRBS) on FSHD2 and control myoblast cell lines. Candidate loci were then evaluated for SMCHD1 binding using ChIP-qPCR and expression was evaluated using RT-qPCR. Results We identified a limited number of clustered autosomal loci with CpG hypomethylation in SMCHD1 mutation carriers: the protocadherin (PCDH) cluster on chromosome 5, the transfer RNA (tRNA) and 5S rRNA clusters on chromosome 1, the HOXB and HOXD clusters on chromosomes 17 and 2, respectively, and the D4Z4 repeats on chromosomes 4 and 10. Furthermore, minor increases in RNA expression were seen in FSHD2 myoblasts for some of the PCDHβ cluster isoforms, tRNA isoforms, and a HOXB isoform in comparison to controls, in addition to the previously reported effects on DUX4 expression. SMCHD1 was bound at DNAseI hypersensitivity sites known to regulate the PCDHβ cluster and at the chromosome 1 tRNA cluster, with decreased binding in SMCHD1 mutation carriers at the PCDHβ cluster sites. Conclusions Our study is the first to investigate the global methylation effects in humans resulting from heterozygous mutations in SMCHD1. Our results suggest that SMCHD1 acts as a repressor on a limited set of autosomal gene clusters, as an observed reduction in methylation associates with a loss of SMCHD1 binding and increased expression for some of the loci. Electronic supplementary material The online version of this article (doi:10.1186/s13395-017-0129-7) contains supplementary material, which is available to authorized users.
- Published
- 2017
9. Trans- and cis-acting effects of the lncRNA Firre on epigenetic and structural features of the inactive X chromosome
- Author
-
He Fang, John L. Rinn, Jitendra K. Thakur, Jay Shendure, Galina N. Filippova, Christine M. Disteche, Jordan P. Lewandowski, Zhijun Duan, Steven Henikoff, Xinxian Deng, Giancarlo Bonora, and William Stafford Noble
- Subjects
Histone ,biology ,Heterochromatin ,CTCF ,biology.protein ,RNA ,Ectopic expression ,Epigenetics ,Gene ,Chromatin ,Cell biology - Abstract
Firre encodes a lncRNA involved in nuclear organization in mammals. Here we find that Firre RNA is transcribed from the active X chromosome (Xa) and exerts trans-acting effects on the inactive X chromosome (Xi). Allelic deletion of Firre on the Xa in a mouse hybrid fibroblast cell line results in a dramatic loss of the histone modification H3K27me3 and of components of the PRC2 complex on the Xi as well as the disruption of the perinucleolar location of the Xi. These features are measurably rescued by ectopic expression of a mouse or human Firre/FIRRE cDNA transgene, strongly supporting a conserved trans-acting role of the Firre transcript in maintaining the Xi heterochromatin environment. Surprisingly, CTCF occupancy is decreased on the Xi upon loss of Firre RNA, but is partially recovered by ectopic transgene expression, suggesting a functional link between Firre RNA and CTCF in maintenance of epigenetic features and/or location of the Xi. Loss of Firre RNA results in dysregulation of genes implicated in cell division and development, but not in reactivation of genes on the Xi, which retains its bipartite structure despite some changes in chromatin contact distribution. Allelic deletion or inversion of Firre on the Xi causes localized redistribution of chromatin contacts, apparently dependent on the orientation of CTCF binding sites clustered at the locus. Thus, the Firre locus and its RNA have roles in the maintenance of epigenetic features and structure of the Xi.
- Published
- 2019
- Full Text
- View/download PDF
10. Allele-Specific Gene Regulation by Kdm6a
- Author
-
Galina N. Filippova, Christine M. Disteche, Joel B. Berletch, He Fang, Nicolas Pease, and Wenxiu Ma
- Subjects
Genetics ,Regulation of gene expression ,Histone ,biology.protein ,Promoter ,Biology ,Allele ,Genomic imprinting ,Gene ,Gene knockout ,X-inactivation ,Chromatin - Abstract
SUMMARYKDM6A demethylates the repressive histone mark H3K27me3 and thus plays an important role in developmental gene regulation. KDM6A expression is female-biased due to escape from X inactivation, suggesting that this protein may play a role in sex differences. Here, we report that maternal and paternal alleles of a subset of mouse genes are differentially regulated by KDM6A. Knockouts of Kdm6a in male and female embryonic stem cells derived from F1 hybrid mice from reciprocal interspecific crosses resulted in preferential downregulation of maternal alleles of a number of genes implicated in development. Moreover, the majority of these genes exhibited a maternal allele expression bias, which was observed in both reciprocal crosses. Promoters of genes downregulated on maternal but not paternal alleles demonstrated a loss of chromatin accessibility, while the expected increase in H3K27me3 levels occurred only at promoters of genes downregulated on paternal but not maternal alleles. These results illustrate parent-of-origin mechanisms of gene regulation by KDM6A, consistent with histone demethylation-dependent and -independent activities.
- Published
- 2019
11. CTCF Expression is Essential for Somatic Cell Viability and Protection Against Cancer
- Author
-
John E.J. Rasko, Crystal Semaan, Galina N. Filippova, Kinsha Baidya, Cynthia Metierre, Yue Feng, Dmitri Loukinov, Victor V. Lobanenkov, and Charles G. Bailey
- Subjects
0301 basic medicine ,CCCTC-Binding Factor ,Somatic cell ,lcsh:Chemistry ,Mice ,Genome editing ,Transcriptional regulation ,CRISPR ,RNA, Small Interfering ,lcsh:QH301-705.5 ,Spectroscopy ,Zinc finger ,Gene knockdown ,gene editing ,General Medicine ,tumour suppressor gene ,3. Good health ,Computer Science Applications ,Chromatin ,Cell biology ,endometrial cancer ,Female ,Haploinsufficiency ,Cell Survival ,Biology ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,medicine ,Animals ,Humans ,cancer ,Physical and Theoretical Chemistry ,Molecular Biology ,Transcription factor ,Gene ,CRISPR/Cas9 ,Cell Proliferation ,molecular_biology ,zinc finger ,Endometrial cancer ,Organic Chemistry ,Cancer ,medicine.disease ,CTCF ,Endometrial Neoplasms ,haploinsufficiency ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cancer research ,CRISPR-Cas Systems ,K562 Cells - Abstract
CCCTC-binding factor (CTCF) is a conserved transcription factor that performs diverse roles in transcriptional regulation and chromatin architecture. Cancer genome sequencing reveals diverse acquired mutations in CTCF, which we have shown functions as a tumour suppressor gene. While CTCF is essential for embryonic development, little is known of its absolute requirement in somatic cells and the consequences of CTCF haploinsufficiency. We examined the consequences of CTCF depletion in immortalised human and mouse cells using shRNA knockdown and CRISPR/Cas9 genome editing as well as examined the growth and development of heterozygous Ctcf (Ctcf+/&minus, ) mice. We also analysed the impact of CTCF haploinsufficiency by examining gene expression changes in CTCF-altered endometrial carcinoma. Knockdown and CRISPR/Cas9-mediated editing of CTCF reduced the cellular growth and colony-forming ability of K562 cells. CTCF knockdown also induced cell cycle arrest and a pro-survival response to apoptotic insult. However, in p53 shRNA-immortalised Ctcf+/&minus, MEFs we observed the opposite: increased cellular proliferation, colony formation, cell cycle progression, and decreased survival after apoptotic insult compared to wild-type MEFs. CRISPR/Cas9-mediated targeting in Ctcf+/&minus, MEFs revealed a predominance of in-frame microdeletions in Ctcf in surviving clones, however protein expression could not be ablated. Examination of CTCF mutations in endometrial cancers showed locus-specific alterations in gene expression due to CTCF haploinsufficiency, in concert with downregulation of tumour suppressor genes and upregulation of estrogen-responsive genes. Depletion of CTCF expression imparts a dramatic negative effect on normal cell function. However, CTCF haploinsufficiency can have growth-promoting effects consistent with known cancer hallmarks in the presence of additional genetic hits. Our results confirm the absolute requirement for CTCF expression in somatic cells and provide definitive evidence of CTCF&rsquo, s role as a haploinsufficient tumour suppressor gene. CTCF genetic alterations in endometrial cancer indicate that gene dysregulation is a likely consequence of CTCF loss, contributing to, but not solely driving cancer growth.
- Published
- 2018
12. A Single-Cell Atlas of In Vivo Mammalian Chromatin Accessibility
- Author
-
Frank J. Steemers, William S DeWitt, Galina N. Filippova, Cole Trapnell, Lena Christiansen, Delasa Aghamirzaie, Andrew J. Hill, Darren A. Cusanovich, Joel B. Berletch, Choli Lee, Hannah A. Pliner, David F. Read, Riza M. Daza, Xingfan Huang, Christine M. Disteche, Samuel G. Regalado, and Jay Shendure
- Subjects
0301 basic medicine ,Epigenomics ,Male ,Cell type ,ATAC-seq ,Computational biology ,Biology ,Genome ,General Biochemistry, Genetics and Molecular Biology ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,Mice ,Animals ,Cluster Analysis ,Humans ,Epigenetics ,Gene ,Transcription factor ,Mammals ,Genome, Human ,Chromatin ,Mice, Inbred C57BL ,030104 developmental biology ,Gene Expression Regulation ,Single-Cell Analysis ,Genome-Wide Association Study ,Transcription Factors - Abstract
We applied a combinatorial indexing assay, sci-ATAC-seq, to profile genome-wide chromatin accessibility in ∼100,000 single cells from 13 adult mouse tissues. We identify 85 distinct patterns of chromatin accessibility, most of which can be assigned to cell types, and ∼400,000 differentially accessible elements. We use these data to link regulatory elements to their target genes, to define the transcription factor grammar specifying each cell type, and to discover in vivo correlates of heterogeneity in accessibility within cell types. We develop a technique for mapping single cell gene expression data to single-cell chromatin accessibility data, facilitating the comparison of atlases. By intersecting mouse chromatin accessibility with human genome-wide association summary statistics, we identify cell-type-specific enrichments of the heritability signal for hundreds of complex traits. These data define the in vivo landscape of the regulatory genome for common mammalian cell types at single-cell resolution.
- Published
- 2018
13. CTCF Haploinsufficiency Destabilizes DNA Methylation and Predisposes to Cancer
- Author
-
Denny Liggitt, Kay E. Gurley, James M. Moore, Russell Moser, Galina N. Filippova, Victor V. Lobanenkov, Leslie E. Smith, Stephanie E. Busch, Brady Bernard, Natalia A. Rabaia, Ari Melnick, Justin Guinney, Ilya Shmulevich, Christopher J. Kemp, Rita Shaknovich, and Matt Teater
- Subjects
CCCTC-Binding Factor ,Mice, Transgenic ,Haploinsufficiency ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,Genetic Predisposition to Disease ,Epigenetics ,lcsh:QH301-705.5 ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Mutation ,Cancer ,DNA Methylation ,medicine.disease ,Survival Analysis ,3. Good health ,Chromatin ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Repressor Proteins ,lcsh:Biology (General) ,CTCF ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,Protein Binding - Abstract
SummaryEpigenetic alterations, particularly in DNA methylation, are ubiquitous in cancer, yet the molecular origins and the consequences of these alterations are poorly understood. CTCF, a DNA-binding protein that regulates higher-order chromatin organization, is frequently altered by hemizygous deletion or mutation in human cancer. To date, a causal role for CTCF in cancer has not been established. Here, we show that Ctcf hemizygous knockout mice are markedly susceptible to spontaneous, radiation-, and chemically induced cancer in a broad range of tissues. Ctcf+/− tumors are characterized by increased aggressiveness, including invasion, metastatic dissemination, and mixed epithelial/mesenchymal differentiation. Molecular analysis of Ctcf+/− tumors indicates that Ctcf is haploinsufficient for tumor suppression. Tissues with hemizygous loss of CTCF exhibit increased variability in CpG methylation genome wide. These findings establish CTCF as a prominent tumor-suppressor gene and point to CTCF-mediated epigenetic stability as a major barrier to neoplastic progression.
- Published
- 2014
14. CTCF physically links cohesin to chromatin
- Author
-
Ruedi Aebersold, Jeffrey A. Ranish, Eric D. Rubio, David J Reiss, Piri Welcsh, Nitin S. Baliga, Christine M. Disteche, Anton Krumm, and Galina N. Filippova
- Subjects
Proteomics ,CCCTC-Binding Factor ,Chromatin Immunoprecipitation ,Chromosomal Proteins, Non-Histone ,Centromere ,Molecular Sequence Data ,Cell Cycle Proteins ,Biology ,Mass Spectrometry ,Genomic Imprinting ,Jurkat Cells ,Mice ,Insulin-Like Growth Factor II ,Animals ,Chromosomes, Human ,Humans ,Sister chromatids ,Amino Acid Sequence ,Alleles ,Genetics ,Multidisciplinary ,Cohesin ,Nuclear Proteins ,3T3 Cells ,Genomics ,Biological Sciences ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,Establishment of sister chromatid cohesion ,CTCF ,Insulator Elements ,Chromatid ,biological phenomena, cell phenomena, and immunity ,Chromatin immunoprecipitation - Abstract
Cohesin is required to prevent premature dissociation of sister chromatids after DNA replication. Although its role in chromatid cohesion is well established, the functional significance of cohesin's association with interphase chromatin is not clear. Using a quantitative proteomics approach, we show that the STAG1 (Scc3/SA1) subunit of cohesin interacts with the CCTC-binding factor CTCF bound to the c-myc insulator element. Both allele-specific binding of CTCF and Scc3/SA1 at the imprinted IGF2/H19 gene locus and our analyses of human DM1 alleles containing base substitutions at CTCF-binding motifs indicate that cohesin recruitment to chromosomal sites depends on the presence of CTCF. A large-scale genomic survey using ChIP-Chip demonstrates that Scc3/SA1 binding strongly correlates with the CTCF-binding site distribution in chromosomal arms. However, some chromosomal sites interact exclusively with CTCF, whereas others interact with Scc3/SA1 only. Furthermore, immunofluorescence microscopy and ChIP-Chip experiments demonstrate that CTCF associates with both centromeres and chromosomal arms during metaphase. These results link cohesin to gene regulatory functions and suggest an essential role for CTCF during sister chromatid cohesion. These results have implications for the functional role of cohesin subunits in the pathogenesis of Cornelia de Lange syndrome and Roberts syndromes.
- Published
- 2008
15. An antisense transcript spanning the CGG repeat region of FMR1 is upregulated in premutation carriers but silenced in full mutation individuals
- Author
-
Randi J Hagerman, Galina N. Filippova, R. Scott Hansen, Paula D. Ladd, Stephen J. Tapscott, Leslie E. Smith, James M. Moore, Sara A. Georges, Natalia A. Rabaia, and Flora Tassone
- Subjects
Untranslated region ,CCCTC-Binding Factor ,Heterozygote ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system diseases ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,Fragile X Mental Retardation Protein ,Mice ,Open Reading Frames ,Trinucleotide Repeats ,Cricetinae ,Genetics ,Transcriptional regulation ,medicine ,Animals ,Humans ,RNA, Antisense ,Tissue Distribution ,Amino Acid Sequence ,Gene Silencing ,Cloning, Molecular ,Allele ,Molecular Biology ,Gene ,Cells, Cultured ,Genetics (clinical) ,Mutation ,Binding Sites ,Base Sequence ,Alternative splicing ,Brain ,General Medicine ,Molecular biology ,Up-Regulation ,nervous system diseases ,Antisense RNA ,DNA-Binding Proteins ,Repressor Proteins ,Alternative Splicing ,Antisense Orientation ,Peptides - Abstract
Expansion of the polymorphic CGG repeats within the 5'-UTR of the FMR1 gene is associated with variable transcriptional regulation of FMR1. Here we report a novel gene, ASFMR1, overlapping the CGG repeat region of FMR1 and transcribed in the antisense orientation. The ASFMR1 transcript is spliced, polyadenylated and exported to the cytoplasm. Similar to FMR1, ASFMR1 is upregulated in individuals with premutation alleles and is not expressed from full mutation alleles. Moreover, it exhibits premutation-specific alternative splicing. Taken together, these observations suggest that in addition to FMR1, ASFMR1 may contribute to the variable phenotypes associated with the CGG repeat expansion.
- Published
- 2007
16. DICER/AGO-dependent epigenetic silencing of D4Z4 repeats enhanced by exogenous siRNA suggests mechanisms and therapies for FSHD
- Author
-
Lauren Snider, Rabi Tawil, Galina N. Filippova, Frank Rigo, Jong-Won Lim, Silvère M. van der Maarel, Stephen J. Tapscott, C. Frank Bennett, and Zizhen Yao
- Subjects
Ribonuclease III ,Small interfering RNA ,Biology ,Cell Line ,Epigenesis, Genetic ,Histones ,RNA interference ,Genetics ,Gene silencing ,Humans ,Gene Silencing ,RNA, Small Interfering ,Molecular Biology ,Genetics (clinical) ,Homeodomain Proteins ,Gene knockdown ,General Medicine ,Articles ,Argonaute ,Chromatin ,Muscular Dystrophy, Facioscapulohumeral ,RNA silencing ,Gene Expression Regulation ,Epigenetic Repression ,Argonaute Proteins ,Cancer research ,biology.protein ,RNA Interference ,Transcription Initiation Site ,Dicer ,Microsatellite Repeats - Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is caused by the aberrant expression of the DUX4 transcription factor in skeletal muscle. The DUX4 retrogene is encoded in the D4Z4 macrosatellite repeat array, and smaller array size or a mutation in the SMCHD1 gene results in inefficient epigenetic repression of DUX4 in skeletal muscle, causing FSHD1 and FSHD2, respectively. Previously we showed that the entire D4Z4 repeat is bi-directionally transcribed with the generation of small si- or miRNA-like fragments and suggested that these might suppress DUX4 expression through the endogenous RNAi pathway. Here we show that exogenous siRNA targeting the region upstream of the DUX4 transcription start site suppressed DUX4 mRNA expression and increased both H3K9 methylation and AGO2 recruitment. In contrast, similarly targeted MOE-gapmer antisense oligonucleotides that degrade RNA but do not engage the RNAi pathway did not repress DUX4 expression. In addition, knockdown of DICER or AGO2 using either siRNA or MOE-gapmer chemistries resulted in the induction of DUX4 expression in control muscle cells that normally do not express DUX4, indicating that the endogenous RNAi pathway is necessary to maintain repression of DUX4 in control muscle cells. Together these data demonstrate a role of the endogenous RNAi pathway in repeat-mediated epigenetic repression of the D4Z4 macrosatellite repeat, and show that enhancing the activity of this pathway by supplying exogenous siRNA oligonucleotides represents a potential therapeutic approach to silencing DUX4 in FSHD.
- Published
- 2015
17. Antisense Transcription and Heterochromatin at the DM1 CTG Repeats Are Constrained by CTCF
- Author
-
Erwin Analau, Galina N. Filippova, Cortlandt P. Thienes, Sarah E. Mahoney, Stephen J. Tapscott, and Diane H. Cho
- Subjects
musculoskeletal diseases ,CCCTC-Binding Factor ,congenital, hereditary, and neonatal diseases and abnormalities ,Transcription, Genetic ,Chromosomal Proteins, Non-Histone ,Heterochromatin ,Quantitative Trait Loci ,Insulator (genetics) ,Biology ,Methylation ,Histones ,Trinucleotide Repeats ,Transcription (biology) ,Humans ,Constitutive heterochromatin ,Nucleosome ,RNA, Antisense ,Promoter Regions, Genetic ,Molecular Biology ,Cells, Cultured ,Homeodomain Proteins ,Genetics ,Cell Biology ,Fibroblasts ,DNA-Binding Proteins ,Repressor Proteins ,CTCF ,DNA methylation ,Insulator Elements ,Protein Binding - Abstract
Summary Prior studies of the DM1 locus have shown that the CTG repeats are a component of a CTCF-dependent insulator element and that repeat expansion results in conversion of the region to heterochromatin. We now show that the DM1 insulator is maintained in a local heterochromatin context: an antisense transcript emanating from the adjacent SIX5 regulatory region extends into the insulator element and is converted into 21 nucleotide (nt) fragments with associated regional histone H3 lysine 9 (H3-K9) methylation and HP1γ recruitment that is embedded within a region of euchromatin-associated H3 lysine 4 (H3-K4) methylation. CTCF restricts the extent of the antisense RNA at the wild-type (wt) DM1 locus and constrains the H3-K9 methylation to the nucleosome associated with the CTG repeat, whereas the expanded allele in congenital DM1 is associated with loss of CTCF binding, spread of heterochromatin, and regional CpG methylation.
- Published
- 2005
18. Thyroid hormone-regulated enhancer blocking: cooperation of CTCF and thyroid hormone receptor
- Author
-
Constanze Bonifer, Les J. Burke, Fiona A. Myers, Colyn Crane-Robinson, Alan W. Thorne, Victor V. Lobanenkov, Galina N. Filippova, Marcus Lutz, Pascal Lefevre, and Rainer Renkawitz
- Subjects
CCCTC-Binding Factor ,Thyroid Hormones ,Molecular Sequence Data ,Electrophoretic Mobility Shift Assay ,Enhancer RNAs ,General Biochemistry, Genetics and Molecular Biology ,Histones ,Histone H4 ,Sequence Homology, Nucleic Acid ,Animals ,Humans ,Enhancer ,Molecular Biology ,Receptors, Thyroid Hormone ,Thyroid hormone receptor ,Base Sequence ,General Immunology and Microbiology ,biology ,General Neuroscience ,Acetylation ,Articles ,Molecular biology ,Chromatin ,DNA-Binding Proteins ,Repressor Proteins ,Enhancer Elements, Genetic ,Histone ,CTCF ,biology.protein ,K562 Cells ,Chickens ,Chromatin immunoprecipitation ,Transcription Factors - Abstract
The highly conserved, ubiquitously expressed, zinc finger protein CTCF is involved in enhancer blocking, a mechanism crucial for shielding genes from illegitimate enhancer effects. Interestingly, CTCF-binding sites are often flanked by thyroid hormone response elements (TREs), as at the chicken lysozyme upstream silencer. Here we identify a similar composite site positioned upstream of the human c-myc gene. For both elements, we demonstrate that thyroid hormone abrogates enhancer blocking. Relief of enhancer blocking occurs even though CTCF remains bound to the lysozyme chromatin. Furthermore, chromatin immunoprecipitation analysis of the lysozyme upstream region revealed that histone H4 is acetylated at the CTCF-binding site. Loss of enhancer blocking by the addition of T3 led to increased histone acetylation, not only at the CTCF site, but also at the enhancer and the promoter. Thus, when TREs are adjacent to CTCF-binding sites, thyroid hormone can regulate enhancer blocking, thereby providing a new property for what was previously thought to be constitutive enhancer shielding by CTCF.
- Published
- 2003
19. Escape from X inactivation
- Author
-
Galina N. Filippova, Karen D. Tsuchiya, and Christine M. Disteche
- Subjects
Genetics ,X Chromosome ,Dosage compensation ,Models, Genetic ,Gene Expression ,Genetic Variation ,Biology ,X-inactivation ,Chromatin ,Evolution, Molecular ,Dosage Compensation, Genetic ,Y Chromosome ,Animals ,Humans ,Gene silencing ,XIST ,Epigenetics ,Molecular Biology ,Skewed X-inactivation ,Genetics (clinical) ,X chromosome - Abstract
Although the process of X inactivation in mammalian cells silences the majority of genes on the inactivated X chromosome, some genes escape this chromosome-wide silencing. Genes that escape X inactivation present a unique opportunity to study the process of silencing and the mechanisms that protect some genes from being turned off. In this review, we will discuss evolutionary aspects of escape from X inactivation, in relation to the divergence of the sex chromosomes. Molecular characteristics, expression, and epigenetic modifications of genes that escape will be presented, including their developmental regulation and the implications of chromatin domains along the X chromosome in modeling the escape process.
- Published
- 2002
20. CTCF-binding sites flank CTG/CAG repeats and form a methylation-sensitive insulator at the DM1 locus
- Author
-
Bennett H. Penn, Victor V. Lobanenkov, Cortlandt P. Thienes, Ying Jia Hu, James M. Moore, Galina N. Filippova, Diane H. Cho, Stephen J. Tapscott, and Todd R. Klesert
- Subjects
musculoskeletal diseases ,CCCTC-Binding Factor ,congenital, hereditary, and neonatal diseases and abnormalities ,Molecular Sequence Data ,Locus (genetics) ,Protein Serine-Threonine Kinases ,Insulator (genetics) ,Biology ,Myotonic dystrophy ,Myotonin-Protein Kinase ,Cell Line ,Trinucleotide Repeats ,Sequence Homology, Nucleic Acid ,Genetics ,medicine ,Humans ,Myotonic Dystrophy ,Nuclear Matrix ,Gene ,Homeodomain Proteins ,Binding Sites ,Cell-Free System ,Myotonin-protein kinase ,Methylation ,DNA Methylation ,medicine.disease ,Molecular biology ,Nucleosomes ,nervous system diseases ,DNA-Binding Proteins ,Repressor Proteins ,CTCF ,DNA methylation ,CpG Islands ,Transcription Factors - Abstract
An expansion of a CTG repeat at the DM1 locus causes myotonic dystrophy (DM) by altering the expression of the two adjacent genes, DMPK and SIX5, and through a toxic effect of the repeat-containing RNA. Here we identify two CTCF-binding sites that flank the CTG repeat and form an insulator element between DMPK and SIX5. Methylation of these sites prevents binding of CTCF, indicating that the DM1 locus methylation in congenital DM would disrupt insulator function. Furthermore, CTCF-binding sites are associated with CTG/CAG repeats at several other loci. We suggest a general role for CTG/CAG repeats as components of insulator elements at multiple sites in the human genome.
- Published
- 2001
21. A widely expressed transcription factor with multiple DNA sequence specificity,CTCF, is localized at chromosome segment 16q22.1 within one of the smallest regions of overlap for common deletions in breast and prostate cancers
- Author
-
Paul E. Neiman, Victor V. Lobanenkov, Steve J. Collins, Elena Klenova, Linda Meincke, Galina N. Filippova, Norman A. Doggett, and Annika Lindblom
- Subjects
Loss of heterozygosity ,Genetics ,Regulation of gene expression ,Cancer Research ,CTCF ,Cancer research ,Locus (genetics) ,Ectopic expression ,Biology ,Candidate Tumor Suppressor Gene ,Gene dosage ,Gene - Abstract
The cellular protooncogene MYC encodes a nuclear transcription factor that is involved in regulating important cellular functions, including cell cycle progression, differentiation, and apoptosis. Dysregulated MYC expression appears critical to the development of various types of malignancies, and thus factors involved in regulating MYC expression may also play a key role in the pathogenesis of certain cancers. We have cloned one such MYC regulatory factor, termed CTCF, which is a highly evolutionarily conserved-11-zinc finger transcriptional factor possessing multiple DNA sequence specificity. CTCF binds to a number of important regulatory regions within the 5' noncoding sequence of the human MYC oncogene, and it can regulate its transcription in several experimental systems. CTCF mRNA is expressed in cells of multiple different lineages. Enforced ectopic expression of CTCF inhibits cell growth in culture. Southern blot analyses and fluorescence in situ hybridization (FISH) with normal human metaphase chromosomes showed that the human CTCF is a single-copy gene situated at chromosome locus 16q22. Cytogenetic studies have pointed out that chromosome abnormalities (deletions) at this locus frequently occur in many different human malignancies, suggesting the presence of one or more tumor suppressor genes in the region. To narrow down their localization, several loss of heterozygosity (LOH) studies of chromosome arm 16q in sporadic breast and prostate cancers have been carried out to define the most recurrent and smallest region(s) of overlap (SRO) for commonly deleted chromosome arm 16q material. For CTCF to be considered as a candidate tumor suppressor gene associated with tumorigenesis, it should localize within one of the SROs at 16q. Fine-mapping of CTCF has enabled us to assign the CTCF gene to about a 2 centiMorgan (cM) interval of 16q22.1 between the somatic cell hybrid breakpoints CY130(D) and CY4, which is between markers D16S186 (16AC16-101) and D16S496 (AFM214zg5). This relatively small region, containing the CTCF gene, overlaps the most frequently observed SROs for common chromosomal deletions found in sporadic breast and prostate tumors. In one of four analyzed paired DNA samples from primary breast cancer patients, we have detected a tumor-specific rearrangement of CTCF exons encoding the 11-zinc-finger domain. Therefore, taken together with other CTCF properties, localization of CTCF to a narrow cancer-associated chromosome region suggests that CTCF is a novel candidate tumor suppressor gene at 16q22.1.
- Published
- 1998
22. Facioscapulohumeral muscular dystrophy: consequences of chromatin relaxation
- Author
-
Rabi Tawil, Daniel G. Miller, Silvère M. van der Maarel, Stephen J. Tapscott, and Galina N. Filippova
- Subjects
musculoskeletal diseases ,muscular dystrophy ,congenital, hereditary, and neonatal diseases and abnormalities ,Transcription, Genetic ,DUX4 ,Biology ,germline ,Germline ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Facioscapulohumeral muscular dystrophy ,Animals ,Humans ,Muscle, Skeletal ,Gene ,Transcription factor ,030304 developmental biology ,Genetics ,Homeodomain Proteins ,0303 health sciences ,retrotransposon ,Skeletal muscle ,facioscapulohumeral ,medicine.disease ,Chromatin ,Muscular Dystrophy, Facioscapulohumeral ,Cell biology ,medicine.anatomical_structure ,Neurology ,Neurology (clinical) ,Stem cell ,030217 neurology & neurosurgery - Abstract
PURPOSE OF REVIEW In recent years, we have seen remarkable progress in our understanding of the disease mechanism underlying facioscapulohumeral muscular dystrophy (FSHD). The purpose of this review is to provide a comprehensive overview of our current understanding of the disease mechanism and to discuss the observations supporting the possibility of a developmental defect in this disorder. RECENT FINDINGS In the majority of cases, FSHD is caused by contraction of the D4Z4 repeat array (FSHD1). This results in local chromatin relaxation and stable expression of the DUX4 retrogene in skeletal muscle, but only when a polymorphic DUX4 polyadenylation signal is present. In some cases (FSHD2), D4Z4 chromatin relaxation and stable DUX4 expression occur in the absence of D4Z4 array contraction. DUX4 is a germline transcription factor and its expression in skeletal muscle leads to activation of early stem cell and germline programs and transcriptional activation of retroelements. SUMMARY Recent studies have provided a plausible disease mechanism for FSHD in which FSHD results from inappropriate expression of the germline transcription factor DUX4. The genes regulated by DUX4 suggest several mechanisms of muscle damage, and provide potential biomarkers and therapeutic targets that should be investigated in future studies.
- Published
- 2012
23. Asymmetric Bidirectional Transcription from the FSHD-Causing D4Z4 Array Modulates DUX4 Production
- Author
-
Gregory J. Block, Daniel G. Miller, Amanda M. Amell, Rabi Tawil, Natalia A. Rabaia, Galina N. Filippova, Lisa M. Petek, James M. Moore, Divya Narayanan, Ashlee E. Tyler, and Silvère M. van der Maarel
- Subjects
Transcription, Genetic ,Muscle Fibers, Skeletal ,Gene Expression ,lcsh:Medicine ,Biochemistry ,Muscular Dystrophies ,Mice ,0302 clinical medicine ,Molecular cell biology ,Transcription (biology) ,Genes, Reporter ,Nucleic Acids ,Sense (molecular biology) ,Transcriptional regulation ,Promoter Regions, Genetic ,lcsh:Science ,Cells, Cultured ,Genetics ,Regulation of gene expression ,0303 health sciences ,Multidisciplinary ,Chromatin ,Muscular Dystrophy, Facioscapulohumeral ,Neurology ,Autosomal Dominant ,Multigene Family ,Medicine ,Epigenetics ,Transcription Initiation Site ,Research Article ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Myoblasts, Skeletal ,Green Fluorescent Proteins ,Molecular Sequence Data ,DNA transcription ,Biology ,Molecular Genetics ,03 medical and health sciences ,DUX4 ,Animals ,Humans ,RNA, Antisense ,Gene Regulation ,Transcription factor ,Embryonic Stem Cells ,030304 developmental biology ,Homeodomain Proteins ,Binding Sites ,Base Sequence ,lcsh:R ,Promoter ,Human Genetics ,Gene Expression Regulation ,Haplotypes ,RNA processing ,Genetics of Disease ,Mutagenesis, Site-Directed ,lcsh:Q ,Gene Function ,030217 neurology & neurosurgery ,Microsatellite Repeats - Abstract
Facioscapulohumeral Disease (FSHD) is a dominantly inherited progressive myopathy associated with aberrant production of the transcription factor, Double Homeobox Protein 4 (DUX4). The expression of DUX4 depends on an open chromatin conformation of the D4Z4 macrosatellite array and a specific haplotype on chromosome 4. Even when these requirements are met, DUX4 transcripts and protein are only detectable in a subset of cells indicating that additional constraints govern DUX4 production. Since the direction of transcription, along with the production of non-coding antisense transcripts is an important regulatory feature of other macrosatellite repeats, we developed constructs that contain the non-coding region of a single D4Z4 unit flanked by genes that report transcriptional activity in the sense and antisense directions. We found that D4Z4 contains two promoters that initiate sense and antisense transcription within the array, and that antisense transcription predominates. Transcriptional start sites for the antisense transcripts, as well as D4Z4 regions that regulate the balance of sense and antisense transcripts were identified. We show that the choice of transcriptional direction is reversible but not mutually exclusive, since sense and antisense reporter activity was often present in the same cell and simultaneously upregulated during myotube formation. Similarly, levels of endogenous sense and antisense D4Z4 transcripts were upregulated in FSHD myotubes. These studies offer insight into the autonomous distribution of muscle weakness that is characteristic of FSHD.
- Published
- 2012
24. Loss of maternal CTCF is associated with peri-implantation lethality of Ctcf null embryos
- Author
-
Leslie E. Smith, Galina N. Filippova, Victor V. Lobanenkov, Natalia A. Rabaia, James M. Moore, Kay E. Gurley, Sara R. Fagerlie, Christopher J. Kemp, Christine M. Disteche, Steven J. Collins, and Dmitry Loukinov
- Subjects
Embryology ,CCCTC-Binding Factor ,Gene Expression ,Embryonic Development ,lcsh:Medicine ,Apoptosis ,Biology ,Molecular Genetics ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Molecular Cell Biology ,Genetics ,medicine ,Animals ,Inner cell mass ,Embryo Implantation ,Blastocyst ,lcsh:Science ,Alleles ,030304 developmental biology ,Mice, Knockout ,Zinc finger ,Regulation of gene expression ,0303 health sciences ,Multidisciplinary ,lcsh:R ,Computational Biology ,Embryo ,Molecular Development ,Molecular biology ,Cell biology ,Chromatin ,Repressor Proteins ,medicine.anatomical_structure ,CTCF ,030220 oncology & carcinogenesis ,lcsh:Q ,Gene Function ,Genomic imprinting ,Organism Development ,Research Article ,Developmental Biology - Abstract
CTCF is a highly conserved, multifunctional zinc finger protein involved in critical aspects of gene regulation including transcription regulation, chromatin insulation, genomic imprinting, X-chromosome inactivation, and higher order chromatin organization. Such multifunctional properties of CTCF suggest an essential role in development. Indeed, a previous report on maternal depletion of CTCF suggested that CTCF is essential for pre-implantation development. To distinguish between the effects of maternal and zygotic expression of CTCF, we studied pre-implantation development in mice harboring a complete loss of function Ctcf knockout allele. Although we demonstrated that homozygous deletion of Ctcf is early embryonically lethal, in contrast to previous observations, we showed that the Ctcf nullizygous embryos developed up to the blastocyst stage (E3.5) followed by peri-implantation lethality (E4.5-E5.5). Moreover, one-cell stage Ctcf nullizygous embryos cultured ex vivo developed to the 16-32 cell stage with no obvious abnormalities. Using a single embryo assay that allowed both genotype and mRNA expression analyses of the same embryo, we demonstrated that pre-implantation development of the Ctcf nullizygous embryos was associated with the retention of the maternal wild type Ctcf mRNA. Loss of this stable maternal transcript was temporally associated with loss of CTCF protein expression, apoptosis of the developing embryo, and failure to further develop an inner cell mass and trophoectoderm ex vivo. This indicates that CTCF expression is critical to early embryogenesis and loss of its expression rapidly leads to apoptosis at a very early developmental stage. This is the first study documenting the presence of the stable maternal Ctcf transcript in the blastocyst stage embryos. Furthermore, in the presence of maternal CTCF, zygotic CTCF expression does not seem to be required for pre-implantation development.
- Published
- 2012
25. CTCF regulates ataxin-7 expression through promotion of a convergently transcribed, antisense noncoding RNA
- Author
-
Bryce L. Sopher, Galina N. Filippova, Susan M. Sunkin, Cortlandt P. Thienes, Albert R. La Spada, Victor V. Pineda, Paula D. Ladd, Terry Gaasterland, James B. Hurley, and Randell T. Libby
- Subjects
CCCTC-Binding Factor ,Ataxin 7 ,RNA, Untranslated ,Neuroscience(all) ,BACE1-AS ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Chromatin remodeling ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Transcription (biology) ,Gene expression ,Tumor Cells, Cultured ,Animals ,Humans ,RNA, Antisense ,Promoter Regions, Genetic ,030304 developmental biology ,Genetics ,Ataxin-7 ,0303 health sciences ,General Neuroscience ,Chromosome Mapping ,Non-coding RNA ,Repressor Proteins ,Gene Expression Regulation ,CTCF ,Transcription Coactivator ,biology.protein ,030217 neurology & neurosurgery - Abstract
SummarySpinocerebellar ataxia type 7 (SCA7) is a neurodegenerative disorder caused by CAG/polyglutamine repeat expansions in the ataxin-7 gene. Ataxin-7 is a component of two different transcription coactivator complexes, and recent work indicates that disease protein normal function is altered in polyglutamine neurodegeneration. Given this, we studied how ataxin-7 gene expression is regulated. The ataxin-7 repeat and translation start site are flanked by binding sites for CTCF, a highly conserved multifunctional transcription regulator. When we analyzed this region, we discovered an adjacent alternative promoter and a convergently transcribed antisense noncoding RNA, SCAANT1. To understand how CTCF regulates ataxin-7 gene expression, we introduced ataxin-7 mini-genes into mice, and found that CTCF is required for SCAANT1 expression. Loss of SCAANT1 derepressed ataxin-7 sense transcription in a cis-dependent fashion and was accompanied by chromatin remodeling. Discovery of this pathway underscores the importance of altered epigenetic regulation for disease pathology at repeat loci exhibiting bidirectional transcription.Video Abstract
- Published
- 2011
26. FSHD: A Repeat Contraction Disease Finally Ready to Expand (Our Understanding of Its Pathogenesis)
- Author
-
Michael Kyba, Silvère M. van der Maarel, Richard J.L.F. Lemmers, Lauren Snider, Stephen J. Tapscott, Rabi Tawil, Carol B. Ware, Angelique M. Nelson, Galina N. Filippova, Daniel G. Miller, and Linda Geng
- Subjects
Male ,Cancer Research ,Developmental Biology/Germ Cells ,Cellular differentiation ,Fluorescent Antibody Technique ,Mice, SCID ,Developmental Biology/Molecular Development ,Mice ,0302 clinical medicine ,Facioscapulohumeral muscular dystrophy ,Myocyte ,Muscular dystrophy ,Genetics (clinical) ,Genetics and Genomics/Genetics of Disease ,Regulation of gene expression ,Genetics and Genomics/Medical Genetics ,0303 health sciences ,Reverse Transcriptase Polymerase Chain Reaction ,Muscular Dystrophy, Facioscapulohumeral ,Developmental Biology/Stem Cells ,medicine.anatomical_structure ,Perspective ,Female ,Chromosomes, Human, Pair 4 ,Research Article ,musculoskeletal diseases ,Adult ,congenital, hereditary, and neonatal diseases and abnormalities ,lcsh:QH426-470 ,Retroelements ,RNA Splicing ,Cell Biology/Developmental Molecular Mechanisms ,Blotting, Western ,Induced Pluripotent Stem Cells ,Molecular Sequence Data ,Neurological Disorders/Neuromuscular Diseases ,Biology ,Cell Line ,03 medical and health sciences ,DUX4 ,Genetics and Genomics/Epigenetics ,Genetics ,medicine ,Gene silencing ,Animals ,Humans ,Muscle, Skeletal ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Cell Biology/Gene Expression ,030304 developmental biology ,Repetitive Sequences, Nucleic Acid ,Homeodomain Proteins ,Muscle Cells ,Models, Genetic ,Gene Expression Profiling ,Skeletal muscle ,medicine.disease ,HCT116 Cells ,Molecular biology ,Mice, Inbred C57BL ,lcsh:Genetics ,Gene Expression Regulation ,030217 neurology & neurosurgery - Abstract
Each unit of the D4Z4 macrosatellite repeat contains a retrotransposed gene encoding the DUX4 double-homeobox transcription factor. Facioscapulohumeral dystrophy (FSHD) is caused by deletion of a subset of the D4Z4 units in the subtelomeric region of chromosome 4. Although it has been reported that the deletion of D4Z4 units induces the pathological expression of DUX4 mRNA, the association of DUX4 mRNA expression with FSHD has not been rigorously investigated, nor has any human tissue been identified that normally expresses DUX4 mRNA or protein. We show that FSHD muscle expresses a different splice form of DUX4 mRNA compared to control muscle. Control muscle produces low amounts of a splice form of DUX4 encoding only the amino-terminal portion of DUX4. FSHD muscle produces low amounts of a DUX4 mRNA that encodes the full-length DUX4 protein. The low abundance of full-length DUX4 mRNA in FSHD muscle cells represents a small subset of nuclei producing a relatively high abundance of DUX4 mRNA and protein. In contrast to control skeletal muscle and most other somatic tissues, full-length DUX4 transcript and protein is expressed at relatively abundant levels in human testis, most likely in the germ-line cells. Induced pluripotent (iPS) cells also express full-length DUX4 and differentiation of control iPS cells to embryoid bodies suppresses expression of full-length DUX4, whereas expression of full-length DUX4 persists in differentiated FSHD iPS cells. Together, these findings indicate that full-length DUX4 is normally expressed at specific developmental stages and is suppressed in most somatic tissues. The contraction of the D4Z4 repeat in FSHD results in a less efficient suppression of the full-length DUX4 mRNA in skeletal muscle cells. Therefore, FSHD represents the first human disease to be associated with the incomplete developmental silencing of a retrogene array normally expressed early in development., Author Summary Facioscapulohumeral muscular dystrophy is caused by the deletion of a subset of D4Z4 macrosatellite repeats on chromosome 4. Each repeat contains a retrogene encoding the double-homeobox factor DUX4. We show that this retrogene is normally expressed in human testis, most likely the germ-line cells, and pluripotent stem cells. DUX4 expression is epigenetically suppressed in differentiated tissues and the residual DUX4 transcripts are spliced to remove the carboxyterminal domain that has been associated with cell toxicity. In FSHD individuals, the expression of the full-length DUX4 transcript is not completely suppressed in skeletal muscle, and possibly other differentiated tissues, and results in a small percentage of cells expressing relatively abundant amounts of the full-length DUX4 mRNA and protein. We therefore propose that FSHD is caused by the inefficient developmental suppression of the DUX4 retrogene and that the residual expression of the full-length DUX4 in skeletal muscle is sufficient to cause the disease. Therefore, FSHD represents the first human disease to be associated with the incomplete developmental silencing of a retrogene array that is normally expressed early in development.
- Published
- 2010
27. RNA transcripts, miRNA-sized fragments and proteins produced from D4Z4 units: new candidates for the pathophysiology of facioscapulohumeral dystrophy
- Author
-
Galina N. Filippova, Daniel G. Miller, Lisa Maves, Amy Asawachaicharn, Silvère M. van der Maarel, Lisa M. Petek, Lauren Snider, Sara T. Winokur, Rabi Tawil, Richard J.L.F. Lemmers, Linda Geng, Stephen J. Tapscott, and Ashlee E. Tyler
- Subjects
Untranslated region ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,RNA, Untranslated ,Polyadenylation ,Molecular Sequence Data ,Biology ,Muscle Development ,Myoblasts ,Mice ,DUX4 ,Transcription (biology) ,Gene expression ,Genetics ,medicine ,Facioscapulohumeral muscular dystrophy ,Animals ,Humans ,Amino Acid Sequence ,RNA, Messenger ,Molecular Biology ,Genetics (clinical) ,Cells, Cultured ,Zebrafish ,Repetitive Sequences, Nucleic Acid ,Homeodomain Proteins ,Base Sequence ,Intron ,RNA ,General Medicine ,Articles ,medicine.disease ,Muscular Dystrophy, Facioscapulohumeral ,Alternative Splicing - Abstract
Deletion of a subset of the D4Z4 macrosatellite repeats in the subtelomeric region of chromosome 4q causes facioscapulohumeral muscular dystrophy (FSHD) when occurring on a specific haplotype of 4qter (4qA161). Several genes have been examined as candidates for causing FSHD, including the DUX4 homeobox gene in the D4Z4 repeat, but none have been definitively shown to cause the disease, nor has the full extent of transcripts from the D4Z4 region been carefully characterized. Using strand-specific RT-PCR, we have identified several sense and antisense transcripts originating from the 4q D4Z4 units in wild-type and FSHD muscle cells. Consistent with prior reports, we find that the DUX4 transcript from the last (most telomeric) D4Z4 unit is polyadenylated and has two introns in its 3-prime untranslated region. In addition, we show that this transcript generates (i) small si/miRNA-sized fragments, (ii) uncapped, polyadenylated 3-prime fragments that encode the conserved C-terminal portion of DUX4 and (iii) capped and polyadenylated mRNAs that contain the double-homeobox domain of DUX4 but splice-out the C-terminal portion. Transfection studies demonstrate that translation initiation at an internal methionine can produce the C-terminal polypeptide and developmental studies show that this peptide inhibits myogenesis at a step between MyoD transcription and the activation of MyoD target genes. Together, we have identified new sense and anti-sense RNA transcripts, novel mRNAs and mi/siRNA-sized RNA fragments generated from the D4Z4 units that are new candidates for the pathophysiology of FSHD.
- Published
- 2009
28. CTCF cis-regulates trinucleotide repeat instability in an epigenetic manner: a novel basis for mutational hot spot determination
- Author
-
Michelle M. Axford, John D. Cleary, Rachel Lau, Bryce L. Sopher, Christopher E. Pearson, Victor V. Pineda, James M. Moore, Galina N. Filippova, Katharine A. Hagerman, Sandy L. Baccam, Albert R. La Spada, Diane H. Cho, Stephen J. Tapscott, and Randell T. Libby
- Subjects
Genome instability ,Male ,Cancer Research ,CCCTC-Binding Factor ,Regulatory Sequences, Nucleic Acid ,Epigenesis, Genetic ,Mice ,0302 clinical medicine ,Genetics (clinical) ,Genetics ,Genetics and Genomics/Medical Genetics ,0303 health sciences ,Mice, Inbred C3H ,DNA-Binding Proteins ,DNA methylation ,Spinocerebellar ataxia ,Female ,Research Article ,lcsh:QH426-470 ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Chromatin remodeling ,Genomic Instability ,03 medical and health sciences ,Microsatellite Repeat ,Genetics and Genomics/Epigenetics ,medicine ,Animals ,Humans ,Spinocerebellar Ataxias ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Ataxin-7 ,Binding Sites ,DNA Methylation ,medicine.disease ,Mice, Inbred C57BL ,Repressor Proteins ,lcsh:Genetics ,Genetics and Genomics/Disease Models ,Neurological Disorders/Neurogenetics ,CTCF ,Mutation ,Trinucleotide repeat expansion ,Genomic imprinting ,Trinucleotide Repeat Expansion ,030217 neurology & neurosurgery - Abstract
At least 25 inherited disorders in humans result from microsatellite repeat expansion. Dramatic variation in repeat instability occurs at different disease loci and between different tissues; however, cis-elements and trans-factors regulating the instability process remain undefined. Genomic fragments from the human spinocerebellar ataxia type 7 (SCA7) locus, containing a highly unstable CAG tract, were previously introduced into mice to localize cis-acting “instability elements,” and revealed that genomic context is required for repeat instability. The critical instability-inducing region contained binding sites for CTCF—a regulatory factor implicated in genomic imprinting, chromatin remodeling, and DNA conformation change. To evaluate the role of CTCF in repeat instability, we derived transgenic mice carrying SCA7 genomic fragments with CTCF binding-site mutations. We found that CTCF binding-site mutation promotes triplet repeat instability both in the germ line and in somatic tissues, and that CpG methylation of CTCF binding sites can further destabilize triplet repeat expansions. As CTCF binding sites are associated with a number of highly unstable repeat loci, our findings suggest a novel basis for demarcation and regulation of mutational hot spots and implicate CTCF in the modulation of genetic repeat instability., Author Summary The human genome contains many repetitive sequences. In 1991, we discovered that excessive lengthening of a three-nucleotide (trinucleotide) repeat sequence could cause a human genetic disease. We now know that this unique type of genetic mutation, known as a “repeat expansion,” occurs in at least 25 different diseases, including inherited neurological disorders such as the fragile X syndrome of mental retardation, myotonic muscular dystrophy, and Huntington's disease. An interesting feature of repeat expansion mutations is that they are genetically unstable, meaning that the repeat expansion changes in length when transmitted from parent to offspring. Thus, expanded repeats violate one major tenet of genetics—i.e., that any given sequence has a low likelihood for mutation. For expanded repeats, the likelihood of further mutation approaches 100%. Understanding why expanded repeats are so mutable has been a challenging problem for genetics research. In this study, we implicate the CTCF protein in the repeat expansion process by showing that mutation of a CTCF binding site, next to an expanded repeat sequence, increases genetic instability in mice. CTCF is an important regulatory factor that controls the expression of genes. As binding sites for CTCF are associated with many repeat sequences, CTCF may play a role in regulating genetic instability in various repeat diseases—not just the one we studied.
- Published
- 2008
29. Genetics and epigenetics of the multifunctional protein CTCF
- Author
-
Galina N, Filippova
- Subjects
Male ,CCCTC-Binding Factor ,Gene Expression Regulation, Developmental ,DNA Methylation ,Models, Biological ,Epigenesis, Genetic ,DNA-Binding Proteins ,Repressor Proteins ,Genomic Imprinting ,X Chromosome Inactivation ,Neoplasms ,Animals ,Humans ,Female ,Genes, Tumor Suppressor ,Repetitive Sequences, Nucleic Acid - Abstract
Recent advances in studying long-range chromatin interactions have shifted focus from the transcriptional regulation by nearby regulatory elements to recognition of the role of higher-order chromatin organization within the nucleus. These advances have also suggested that CCCTC-binding factor (CTCF), a known chromatin insulator protein, may play a central role in mediating long-range chromatin interactions, directing DNA segments into transcription factories and/or facilitating interactions with other DNA regions. Several models that describe possible mechanisms for multiple functions of CTCF in establishment and maintenance of epigenetic programs are now emerging. Epigenetics plays an important role in normal development and disease including cancer. CTCF involvement in multiple aspects of epigenetic regulation, including regulation of genomic imprinting and X-chromosome inactivation, has been well established. More recently, CTCF was found to play a role in regulation of noncoding transcription and establishing local chromatin structure at the repetitive elements in mammalian genomes, suggesting a new epigenetic basis for several repeat-associated genetic disorders. Emerging evidence also points to the role of CTCF deregulation in the epigenetic imbalance in cancer. These studies provide some of the important missing links in our understanding of epigenetic control of both development and cancer.
- Published
- 2007
30. Genetics and Epigenetics of the Multifunctional Protein CTCF
- Author
-
Galina N. Filippova
- Subjects
Genetics ,Transcription factories ,Regulation of gene expression ,CTCF ,DNA methylation ,Epigenetics ,Biology ,Insulator (genetics) ,Genomic imprinting ,Chromatin - Abstract
Recent advances in studying long-range chromatin interactions have shifted focus from the transcriptional regulation by nearby regulatory elements to recognition of the role of higher-order chromatin organization within the nucleus. These advances have also suggested that CCCTC-binding factor (CTCF), a known chromatin insulator protein, may play a central role in mediating long-range chromatin interactions, directing DNA segments into transcription factories and/or facilitating interactions with other DNA regions. Several models that describe possible mechanisms for multiple functions of CTCF in establishment and maintenance of epigenetic programs are now emerging. Epigenetics plays an important role in normal development and disease including cancer. CTCF involvement in multiple aspects of epigenetic regulation, including regulation of genomic imprinting and X-chromosome inactivation, has been well established. More recently, CTCF was found to play a role in regulation of noncoding transcription and establishing local chromatin structure at the repetitive elements in mammalian genomes, suggesting a new epigenetic basis for several repeat-associated genetic disorders. Emerging evidence also points to the role of CTCF deregulation in the epigenetic imbalance in cancer. These studies provide some of the important missing links in our understanding of epigenetic control of both development and cancer.
- Published
- 2007
31. Boundaries between chromosomal domains of X inactivation and escape bind CTCF and lack CpG methylation during early development
- Author
-
Galina N. Filippova, Mimi K. Cheng, Di Kim Nguyen, Ying J. Hu, Christine M. Disteche, Karen D. Tsuchiya, James M. Moore, and Jean Pierre Truong
- Subjects
Models, Molecular ,CCCTC-Binding Factor ,X Chromosome ,Eukaryotic Initiation Factor-2 ,Molecular Sequence Data ,Electrophoretic Mobility Shift Assay ,Insulator (genetics) ,Biology ,General Biochemistry, Genetics and Molecular Biology ,X-inactivation ,03 medical and health sciences ,Mice ,Dosage Compensation, Genetic ,Animals ,Humans ,Immunoprecipitation ,Histone H3 acetylation ,Molecular Biology ,Gene ,Cells, Cultured ,030304 developmental biology ,Genetics ,Histone Demethylases ,0303 health sciences ,Binding Sites ,030302 biochemistry & molecular biology ,Gene Expression Regulation, Developmental ,Proteins ,Acetylation ,Oxidoreductases, N-Demethylating ,Cell Biology ,Methylation ,DNA Methylation ,Embryo, Mammalian ,Chromatin ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Repressor Proteins ,CTCF ,Mutagenesis ,DNA methylation ,CpG Islands ,Developmental Biology - Abstract
Escape from X inactivation results in expression of genes embedded within inactive chromatin, suggesting the existence of boundary elements between domains. We report that the 5′ end of Jarid1c , a mouse escape gene adjacent to an inactivated gene, binds CTCF, displays high levels of histone H3 acetylation, and functions as a CTCF-dependent chromatin insulator. CpG island methylation at Jarid1c was very low during development and virtually absent at the CTCF sites, signifying that CTCF may influence DNA methylation and chromatin modifications. CTCF binding sites were also present at the 5′ end of two other escape genes, mouse Eif2s3x and human EIF2S3 , each adjacent to an inactivated gene, but not at genes embedded within large escape domains. Thus, CTCF was specifically bound to transition regions, suggesting a role in maintaining both X inactivation and escape domains. Furthermore, the evolution of X chromosome domains appears to be associated with repositioning of chromatin boundary elements.
- Published
- 2005
32. CTCF is conserved from Drosophila to humans and confers enhancer blocking of the Fab-8 insulator
- Author
-
Rainer Renkawitz, Britta Grewe, Rolf Ohlsson, Galina N. Filippova, Marek Bartkuhn, Renate Renkawitz-Pohl, Rüdiger Arnold, Adam Munhall, Elena M. Pugacheva, Dmitry Loukinov, Jumin Zhou, Hanlim Moon, Sheryl T. Smith, Les J. Burke, Qi Chen, and Victor V. Lobanenkov
- Subjects
CCCTC-Binding Factor ,Molecular Sequence Data ,Scientific Report ,Enhancer RNAs ,Biology ,Biochemistry ,DNA-binding protein ,Genetics ,Enhancer trap ,Animals ,Drosophila Proteins ,Humans ,Amino Acid Sequence ,Enhancer ,Molecular Biology ,Zinc finger ,Homeodomain Proteins ,Chromatin ,DNA-Binding Proteins ,Repressor Proteins ,Enhancer Elements, Genetic ,Gene Expression Regulation ,CTCF ,Drosophila ,Insulator Elements ,Drosophila Protein - Abstract
Eukaryotic transcriptional regulation often involves regulatory elements separated from the cognate genes by long distances, whereas appropriately positioned insulator or enhancer-blocking elements shield promoters from illegitimate enhancer action. Four proteins have been identified in Drosophila mediating enhancer blocking—Su(Hw), Zw5, BEAF32 and GAGA factor. In vertebrates, the single protein CTCF, with 11 highly conserved zinc fingers, confers enhancer blocking in all known chromatin insulators. Here, we characterize an orthologous CTCF factor in Drosophila with a similar domain structure, binding site specificity and transcriptional repression activity as in vertebrates. In addition, we demonstrate that one of the insulators (Fab-8) in the Drosophila Abdominal-B locus mediates enhancer blocking by dCTCF. Therefore, the enhancer-blocking protein CTCF and, most probably, the mechanism of enhancer blocking mediated by this remarkably versatile factor are conserved from Drosophila to humans.
- Published
- 2004
33. Functional phosphorylation sites in the C-terminal region of the multivalent multifunctional transcriptional factor CTCF
- Author
-
Ayman El-Kady, Igor Chernukhin, Paul E. Neiman, Robin E. Lee, Dolores Delgado, Victor V. Lobanenkov, Graham H. Goodwin, Elena M. Pugacheva, Herbert C. Morse, Elena Klenova, Galina N. Filippova, Javier León, and Dmitri Loukinov
- Subjects
CCCTC-Binding Factor ,Molecular Sequence Data ,Genes, myc ,Biology ,Insulator (genetics) ,Protein Serine-Threonine Kinases ,DNA-binding protein ,Cell Line ,Transcriptional regulation ,Animals ,Humans ,Amino Acid Sequence ,Phosphorylation ,Casein Kinase II ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Transcriptional Regulation ,Binding Sites ,Cell Biology ,Molecular biology ,DNA-Binding Proteins ,Repressor Proteins ,Amino Acid Substitution ,CTCF ,Mutation ,Casein kinase 2 ,Chickens ,Nuclear localization sequence ,Cell Division ,Transcription Factors - Abstract
CTCF is a widely expressed and highly conserved multi-Zn-finger (ZF) nuclear factor. Binding to various CTCF target sites (CTSs) is mediated by combinatorial contributions of different ZFs. Different CTSs mediate distinct CTCF functions in transcriptional regulation, including promoter repression or activation and hormone-responsive gene silencing. In addition, the necessary and sufficient core sequences of diverse enhancer-blocking (insulator) elements, including CpG methylation-sensitive ones, have recently been pinpointed to CTSs. To determine whether a posttranslational modification may modulate CTCF functions, we studied CTCF phosphorylation. We demonstrated that most of the modifications that occur at the carboxy terminus in vivo can be reproduced in vitro with casein kinase II (CKII). Major modification sites map to four serines within the S(604)KKEDS(609)S(610)DS(612)E motif that is highly conserved in vertebrates. Specific mutations of these serines abrogate phosphorylation of CTCF in vivo and CKII-induced phosphorylation in vitro. In addition, we showed that completely preventing phosphorylation by substituting all serines within this site resulted in markedly enhanced repression of the CTS-bearing vertebrate c-myc promoters, but did not alter CTCF nuclear localization or in vitro DNA-binding characteristics assayed with c-myc CTSs. Moreover, these substitutions manifested a profound effect on negative cell growth regulation by wild-type CTCF. CKII may thus be responsible for attenuation of CTCF activity, either acting on its own or by providing the signal for phosphorylation by other kinases and for CTCF-interacting protein partners.
- Published
- 2001
34. Characterization of the chicken CTCF genomic locus, and initial study of the cell cycle-regulated promoter of the gene
- Author
-
Galina N. Filippova, Gilbert Loring, Victor V. Lobanenkov, Graham H. Goodwin, Paul E. Neiman, Sara R. Fagerlie, Elena Klenova, and Leo Kretzner
- Subjects
CCCTC-Binding Factor ,Transcription, Genetic ,Response element ,Molecular Sequence Data ,Cell Cycle Proteins ,Biology ,Biochemistry ,Transcriptional regulation ,Animals ,Humans ,RNA, Messenger ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,Transcription factor ,Genetics ,Base Sequence ,YY1 ,Cell Cycle ,Promoter ,Cell Biology ,DNA ,Cell Cycle Gene ,DNA-Binding Proteins ,Repressor Proteins ,CTCF ,Chickens ,Transcription Factors - Abstract
CTCF is a multifunctional transcription factor encoded by a novel candidate tumor suppressor gene (Filippova, G. N., Lindblom, A., Meinke, L. J., Klenova, E. M., Neiman, P. E., Collins, S. J., Doggett, N. D., and Lobanenkov, V. V. (1998) Genes Chromosomes Cancer 22, 26–36). We characterized genomic organization of the chicken CTCF(chCTCF) gene, and studied the chCTCF promoter. Genomic locus of chCTCF contains a GC-rich untranslated exon separated from seven coding exons by a long intron. The 2-kilobase pair region upstream of the major transcription start site contains a CpG island marked by a “Not-knot” that includes sequence motifs characteristic of a TATA-less promoter of housekeeping genes. When fused upstream of a reporter chloramphenicol acetyltransferase gene, it acts as a strong transcriptional promoter in transient transfection experiments. The minimal 180-base pair chCTCF promoter region that is fully sufficient to confer high level transcriptional activity to the reporter contains high affinity binding element for the transcription factor YY1. This element is strictly conserved in chicken, mouse, and human CTCF genes. Mutations in the core nucleotides of the YY1 element reduce transcriptional activity of the minimal chCTCF promoter, indicating that the conserved YY1-binding sequence is critical for transcriptional regulation of vertebrate CTCF genes. We also noted in thechCTCF promoter several elements previously characterized in cell cycle-regulated genes, including the “cell cycle-dependent element” and “cell cycle gene homology region” motifs shown to be important for S/G2-specific up-regulation of cdc25C, cdc2, cyclin A, and Plk (polo-like kinase) gene promoters. Presence of the cell cycle-dependent element/cell cycle gene homology region element suggested that chCTCFexpression may be cell cycle-regulated. We show that both levels of the endogenous chCTCF mRNA, and the activity of the stably transfected chCTCF promoter constructs, increase in S/G2 cells.
- Published
- 1998
35. Mechanisms of cycloheximide-induced apoptosis in liver cells
- Author
-
Alice V Alessenko, Peter Ya Boikov, Galina N Filippova, Alexey V Khrenov, Anatoliy S Loginov, and Elena D Makarieva
- Subjects
PLCD3 ,Biophysics ,Apoptosis ,Cycloheximide ,Biology ,Biochemistry ,chemistry.chemical_compound ,Structural Biology ,In vivo ,Sphingosine ,Genetics ,Animals ,Rats, Wistar ,Molecular Biology ,Gene ,Protein kinase C ,Protein Synthesis Inhibitors ,p53 gene expression ,Cell Biology ,Cell biology ,c-myc gene expression ,Rats ,c-fos gene expression ,Microscopy, Electron ,chemistry ,Gene Expression Regulation ,Liver ,Cancer research ,Sphingomyelin ,c-jun gene expression - Abstract
Cycloheximide in sublethal doses caused apoptosis in liver cells in vivo, inducing c-myc, c-fos, c-jun and p53 genes and accumulation of sphingosine, a toxic product of the sphingomyelin cycle. These data support the hypothesis that continuous synthesis of labile protective proteins is required to restrain apoptosis in liver; sphingosine might be important in mediating cycloheximide-induced apoptosis as an endogenous modulator of protein kinase C activity.
- Published
- 1997
36. An exceptionally conserved transcriptional repressor, CTCF, employs different combinations of zinc fingers to bind diverged promoter sequences of avian and mammalian c-myc oncogenes
- Author
-
Y Dehner, Paul E. Neiman, Victor V. Lobanenkov, Sara R. Fagerlie, Steven J. Collins, Galina N. Filippova, Elena Klenova, C Myers, and G Goodwin
- Subjects
DNA, Complementary ,Molecular Sequence Data ,Genes, myc ,Repressor ,Biology ,Transfection ,Conserved sequence ,Cell Line ,Mice ,Species Specificity ,Animals ,Humans ,Amino Acid Sequence ,Binding site ,Promoter Regions, Genetic ,Molecular Biology ,Conserved Sequence ,Genetics ,Binding Sites ,Base Sequence ,Sequence Homology, Amino Acid ,Promoter ,Zinc Fingers ,Cell Biology ,DNA-binding domain ,Repressor Proteins ,Regulatory sequence ,CTCF ,Transcriptional Repressor CTCF ,Chickens ,Protein Binding ,Research Article - Abstract
We have isolated and analyzed human CTCF cDNA clones and show here that the ubiquitously expressed 11-zinc-finger factor CTCF is an exceptionally highly conserved protein displaying 93% identity between avian and human amino acid sequences. It binds specifically to regulatory sequences in the promoter-proximal regions of chicken, mouse, and human c-myc oncogenes. CTCF contains two transcription repressor domains transferable to a heterologous DNA binding domain. One CTCF binding site, conserved in mouse and human c-myc genes, is found immediately downstream of the major P2 promoter at a sequence which maps precisely within the region of RNA polymerase II pausing and release. Gel shift assays of nuclear extracts from mouse and human cells show that CTCF is the predominant factor binding to this sequence. Mutational analysis of the P2-proximal CTCF binding site and transient-cotransfection experiments demonstrate that CTCF is a transcriptional repressor of the human c-myc gene. Although there is 100% sequence identity in the DNA binding domains of the avian and human CTCF proteins, the regulatory sequences recognized by CTCF in chicken and human c-myc promoters are clearly diverged. Mutating the contact nucleotides confirms that CTCF binding to the human c-myc P2 promoter requires a number of unique contact DNA bases that are absent in the chicken c-myc CTCF binding site. Moreover, proteolytic-protection assays indicate that several more CTCF Zn fingers are involved in contacting the human CTCF binding site than the chicken site. Gel shift assays utilizing successively deleted Zn finger domains indicate that CTCF Zn fingers 2 to 7 are involved in binding to the chicken c-myc promoter, while fingers 3 to 11 mediate CTCF binding to the human promoter. This flexibility in Zn finger usage reveals CTCF to be a unique "multivalent" transcriptional factor and provides the first feasible explanation of how certain homologous genes (i.e., c-myc) of different vertebrate species are regulated by the same factor and maintain similar expression patterns despite significant promoter sequence divergence.
- Published
- 1996
37. Abstract 118: CTCF, a master epigenetic regulator, is a haploinsufficient tumor suppressor gene in multiple cell lineages
- Author
-
Leslie Smith, Denny Liggett, James M. Moore, Russell Moser, Christopher J. Kemp, Galina N. Filippova, and Kay E. Gurley
- Subjects
Genetics ,Cancer Research ,Tumor suppressor gene ,Cancer ,Environmental exposure ,Biology ,medicine.disease ,Oncology ,CTCF ,Knockout mouse ,DNA methylation ,medicine ,Cancer research ,Neoplastic transformation ,Epigenetics - Abstract
While changes in the epigenetic landscape are ubiquitous in cancer, the origin and causal effects of most of these changes on neoplastic progression are largely unknown. The DNA binding protein Ctcf, regulates a diverse array of epigenetic processes, including DNA methylation spreading, but is role in cancer is unclear. Here we show that Ctcf heterozygous knockout mice are remarkably susceptible to spontaneous, ionizing radiation, and chemically-induced neoplasia in a broad range of tissues. Analysis of lung tumors from Ctcf+/− mice establishes Ctcf as a haploinsufficient tumor suppressor gene whose loss cooperates with mutant Kras to accelerate tumor growth and malignant progression. Increased DNA methylation at Ctcf binding sites in normal tissues from Ctcf+/− mice indicates that reduction of Ctcf leads to epigenetic deregulation and this occurs prior to overt neoplastic transformation. This indicates epigenetic instability can predispose a broad range of tissues to neoplastic transformation. A mouse model of Ctcf regulated epigenetic stability provides a useful setting to dissect the mechanistic link between environmental exposure, and risk of cancer or other diseases with an epigenetic component. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 118. doi:1538-7445.AM2012-118
- Published
- 2012
38. CTCF Expression is Essential for Somatic Cell Viability and Protection Against Cancer
- Author
-
Charles G Bailey, Cynthia Metierre, Yue Feng, Kinsha Baidya, Galina N Filippova, Dmitri I Loukinov, Victor V Lobanenkov, Crystal Semaan, and John EJ Rasko
- Subjects
CTCF ,tumour suppressor gene ,haploinsufficiency ,zinc finger ,CRISPR/Cas9 ,cancer ,endometrial cancer ,gene editing ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
CCCTC-binding factor (CTCF) is a conserved transcription factor that performs diverse roles in transcriptional regulation and chromatin architecture. Cancer genome sequencing reveals diverse acquired mutations in CTCF, which we have shown functions as a tumour suppressor gene. While CTCF is essential for embryonic development, little is known of its absolute requirement in somatic cells and the consequences of CTCF haploinsufficiency. We examined the consequences of CTCF depletion in immortalised human and mouse cells using shRNA knockdown and CRISPR/Cas9 genome editing as well as examined the growth and development of heterozygous Ctcf (Ctcf+/−) mice. We also analysed the impact of CTCF haploinsufficiency by examining gene expression changes in CTCF-altered endometrial carcinoma. Knockdown and CRISPR/Cas9-mediated editing of CTCF reduced the cellular growth and colony-forming ability of K562 cells. CTCF knockdown also induced cell cycle arrest and a pro-survival response to apoptotic insult. However, in p53 shRNA-immortalised Ctcf+/− MEFs we observed the opposite: increased cellular proliferation, colony formation, cell cycle progression, and decreased survival after apoptotic insult compared to wild-type MEFs. CRISPR/Cas9-mediated targeting in Ctcf+/− MEFs revealed a predominance of in-frame microdeletions in Ctcf in surviving clones, however protein expression could not be ablated. Examination of CTCF mutations in endometrial cancers showed locus-specific alterations in gene expression due to CTCF haploinsufficiency, in concert with downregulation of tumour suppressor genes and upregulation of estrogen-responsive genes. Depletion of CTCF expression imparts a dramatic negative effect on normal cell function. However, CTCF haploinsufficiency can have growth-promoting effects consistent with known cancer hallmarks in the presence of additional genetic hits. Our results confirm the absolute requirement for CTCF expression in somatic cells and provide definitive evidence of CTCF’s role as a haploinsufficient tumour suppressor gene. CTCF genetic alterations in endometrial cancer indicate that gene dysregulation is a likely consequence of CTCF loss, contributing to, but not solely driving cancer growth.
- Published
- 2018
- Full Text
- View/download PDF
39. Loss of maternal CTCF is associated with peri-implantation lethality of Ctcf null embryos.
- Author
-
James M Moore, Natalia A Rabaia, Leslie E Smith, Sara Fagerlie, Kay Gurley, Dmitry Loukinov, Christine M Disteche, Steven J Collins, Christopher J Kemp, Victor V Lobanenkov, and Galina N Filippova
- Subjects
Medicine ,Science - Abstract
CTCF is a highly conserved, multifunctional zinc finger protein involved in critical aspects of gene regulation including transcription regulation, chromatin insulation, genomic imprinting, X-chromosome inactivation, and higher order chromatin organization. Such multifunctional properties of CTCF suggest an essential role in development. Indeed, a previous report on maternal depletion of CTCF suggested that CTCF is essential for pre-implantation development. To distinguish between the effects of maternal and zygotic expression of CTCF, we studied pre-implantation development in mice harboring a complete loss of function Ctcf knockout allele. Although we demonstrated that homozygous deletion of Ctcf is early embryonically lethal, in contrast to previous observations, we showed that the Ctcf nullizygous embryos developed up to the blastocyst stage (E3.5) followed by peri-implantation lethality (E4.5-E5.5). Moreover, one-cell stage Ctcf nullizygous embryos cultured ex vivo developed to the 16-32 cell stage with no obvious abnormalities. Using a single embryo assay that allowed both genotype and mRNA expression analyses of the same embryo, we demonstrated that pre-implantation development of the Ctcf nullizygous embryos was associated with the retention of the maternal wild type Ctcf mRNA. Loss of this stable maternal transcript was temporally associated with loss of CTCF protein expression, apoptosis of the developing embryo, and failure to further develop an inner cell mass and trophoectoderm ex vivo. This indicates that CTCF expression is critical to early embryogenesis and loss of its expression rapidly leads to apoptosis at a very early developmental stage. This is the first study documenting the presence of the stable maternal Ctcf transcript in the blastocyst stage embryos. Furthermore, in the presence of maternal CTCF, zygotic CTCF expression does not seem to be required for pre-implantation development.
- Published
- 2012
- Full Text
- View/download PDF
40. Asymmetric bidirectional transcription from the FSHD-causing D4Z4 array modulates DUX4 production.
- Author
-
Gregory J Block, Lisa M Petek, Divya Narayanan, Amanda M Amell, James M Moore, Natalia A Rabaia, Ashlee Tyler, Silvere M van der Maarel, Rabi Tawil, Galina N Filippova, and Daniel G Miller
- Subjects
Medicine ,Science - Abstract
Facioscapulohumeral Disease (FSHD) is a dominantly inherited progressive myopathy associated with aberrant production of the transcription factor, Double Homeobox Protein 4 (DUX4). The expression of DUX4 depends on an open chromatin conformation of the D4Z4 macrosatellite array and a specific haplotype on chromosome 4. Even when these requirements are met, DUX4 transcripts and protein are only detectable in a subset of cells indicating that additional constraints govern DUX4 production. Since the direction of transcription, along with the production of non-coding antisense transcripts is an important regulatory feature of other macrosatellite repeats, we developed constructs that contain the non-coding region of a single D4Z4 unit flanked by genes that report transcriptional activity in the sense and antisense directions. We found that D4Z4 contains two promoters that initiate sense and antisense transcription within the array, and that antisense transcription predominates. Transcriptional start sites for the antisense transcripts, as well as D4Z4 regions that regulate the balance of sense and antisense transcripts were identified. We show that the choice of transcriptional direction is reversible but not mutually exclusive, since sense and antisense reporter activity was often present in the same cell and simultaneously upregulated during myotube formation. Similarly, levels of endogenous sense and antisense D4Z4 transcripts were upregulated in FSHD myotubes. These studies offer insight into the autonomous distribution of muscle weakness that is characteristic of FSHD.
- Published
- 2012
- Full Text
- View/download PDF
41. Facioscapulohumeral dystrophy: incomplete suppression of a retrotransposed gene.
- Author
-
Lauren Snider, Linda N Geng, Richard J L F Lemmers, Michael Kyba, Carol B Ware, Angelique M Nelson, Rabi Tawil, Galina N Filippova, Silvère M van der Maarel, Stephen J Tapscott, and Daniel G Miller
- Subjects
Genetics ,QH426-470 - Abstract
Each unit of the D4Z4 macrosatellite repeat contains a retrotransposed gene encoding the DUX4 double-homeobox transcription factor. Facioscapulohumeral dystrophy (FSHD) is caused by deletion of a subset of the D4Z4 units in the subtelomeric region of chromosome 4. Although it has been reported that the deletion of D4Z4 units induces the pathological expression of DUX4 mRNA, the association of DUX4 mRNA expression with FSHD has not been rigorously investigated, nor has any human tissue been identified that normally expresses DUX4 mRNA or protein. We show that FSHD muscle expresses a different splice form of DUX4 mRNA compared to control muscle. Control muscle produces low amounts of a splice form of DUX4 encoding only the amino-terminal portion of DUX4. FSHD muscle produces low amounts of a DUX4 mRNA that encodes the full-length DUX4 protein. The low abundance of full-length DUX4 mRNA in FSHD muscle cells represents a small subset of nuclei producing a relatively high abundance of DUX4 mRNA and protein. In contrast to control skeletal muscle and most other somatic tissues, full-length DUX4 transcript and protein is expressed at relatively abundant levels in human testis, most likely in the germ-line cells. Induced pluripotent (iPS) cells also express full-length DUX4 and differentiation of control iPS cells to embryoid bodies suppresses expression of full-length DUX4, whereas expression of full-length DUX4 persists in differentiated FSHD iPS cells. Together, these findings indicate that full-length DUX4 is normally expressed at specific developmental stages and is suppressed in most somatic tissues. The contraction of the D4Z4 repeat in FSHD results in a less efficient suppression of the full-length DUX4 mRNA in skeletal muscle cells. Therefore, FSHD represents the first human disease to be associated with the incomplete developmental silencing of a retrogene array normally expressed early in development.
- Published
- 2010
- Full Text
- View/download PDF
42. CTCF cis-regulates trinucleotide repeat instability in an epigenetic manner: a novel basis for mutational hot spot determination.
- Author
-
Randell T Libby, Katharine A Hagerman, Victor V Pineda, Rachel Lau, Diane H Cho, Sandy L Baccam, Michelle M Axford, John D Cleary, James M Moore, Bryce L Sopher, Stephen J Tapscott, Galina N Filippova, Christopher E Pearson, and Albert R La Spada
- Subjects
Genetics ,QH426-470 - Abstract
At least 25 inherited disorders in humans result from microsatellite repeat expansion. Dramatic variation in repeat instability occurs at different disease loci and between different tissues; however, cis-elements and trans-factors regulating the instability process remain undefined. Genomic fragments from the human spinocerebellar ataxia type 7 (SCA7) locus, containing a highly unstable CAG tract, were previously introduced into mice to localize cis-acting "instability elements," and revealed that genomic context is required for repeat instability. The critical instability-inducing region contained binding sites for CTCF -- a regulatory factor implicated in genomic imprinting, chromatin remodeling, and DNA conformation change. To evaluate the role of CTCF in repeat instability, we derived transgenic mice carrying SCA7 genomic fragments with CTCF binding-site mutations. We found that CTCF binding-site mutation promotes triplet repeat instability both in the germ line and in somatic tissues, and that CpG methylation of CTCF binding sites can further destabilize triplet repeat expansions. As CTCF binding sites are associated with a number of highly unstable repeat loci, our findings suggest a novel basis for demarcation and regulation of mutational hot spots and implicate CTCF in the modulation of genetic repeat instability.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.