Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta ; 1859(3): 496-509, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26689747

ABSTRACT

H1 linker histones are key chromatin architectural proteins facilitating the formation of higher order chromatin structures. The H1 family constitutes the most heterogeneous group of histone proteins, with eleven non-allelic H1 variants in mammals. H1 variants differ in their biochemical properties and exhibit significant sequence divergence from one another, yet most of them are highly conserved during evolution from mouse to human. H1 variants are differentially regulated during development and their cellular compositions undergo dramatic changes in embryogenesis, gametogenesis, tissue maturation and cellular differentiation. As a group, H1 histones are essential for mouse development and proper stem cell differentiation. Here we summarize our current knowledge on the expression and functions of H1 variants in mammalian development and stem cell differentiation. Their diversity, sequence conservation, complex expression and distinct functions suggest that H1s mediate chromatin reprogramming and contribute to the large variations and complexity of chromatin structure and gene expression in the mammalian genome.


Subject(s)
Embryonic Development , Gametogenesis , Histones/physiology , Stem Cells/cytology , Amino Acid Sequence , Animals , Cell Differentiation , Chromatin/chemistry , Humans , Mice , Molecular Sequence Data
2.
Cancer Res ; 74(22): 6463-73, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25205099

ABSTRACT

Ovarian cancer is a deadly gynecologic malignancy for which novel biomarkers and therapeutic targets are imperative for improving survival. Previous studies have suggested the expression pattern of linker histone variants as potential biomarkers for ovarian cancer. To investigate the role of histone H1 in ovarian cancer cells, we characterize individual H1 variants and overexpress one of the major somatic H1 variants, H1.3, in the OVCAR-3 epithelial ovarian cancer cell line. We find that overexpression of H1.3 decreases the growth rate and colony formation of OVCAR-3 cells. We identify histone H1.3 as a specific repressor for the noncoding oncogene H19. Overexpression of H1.3 suppresses H19 expression, and knockdown of H1.3 increases its expression in multiple ovarian epithelial cancer cell lines. Furthermore, we demonstrate that histone H1.3 overexpression leads to increased occupancy of H1.3 at the H19 regulator region encompassing the imprinting control region (ICR), concomitant with increased DNA methylation and reduced occupancy of the insulator protein CTCF at the ICR. Finally, we demonstrate that H1.3 overexpression and H19 knockdown synergistically decrease the growth rate of ovarian cancer cells. Our findings suggest that H1.3 dramatically inhibits H19 expression, which contributes to the suppression of epithelial ovarian carcinogenesis.


Subject(s)
Cell Proliferation , Histones/physiology , Ovarian Neoplasms/pathology , RNA, Long Noncoding/physiology , Cell Line, Tumor , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Humans , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics
3.
J Clin Invest ; 124(7): 3187-99, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24865430

ABSTRACT

In atherosclerosis, plaques preferentially develop in arterial regions of disturbed blood flow (d-flow), which alters endothelial gene expression and function. Here, we determined that d-flow regulates genome-wide DNA methylation patterns in a DNA methyltransferase-dependent (DNMT-dependent) manner. Induction of d-flow by partial carotid ligation surgery in a murine model induced DNMT1 in arterial endothelium. In cultured endothelial cells, DNMT1 was enhanced by oscillatory shear stress (OS), and reduction of DNMT with either the inhibitor 5-aza-2'-deoxycytidine (5Aza) or siRNA markedly reduced OS-induced endothelial inflammation. Moreover, administration of 5Aza reduced lesion formation in 2 mouse models of atherosclerosis. Using both reduced representation bisulfite sequencing (RRBS) and microarray, we determined that d-flow in the carotid artery resulted in hypermethylation within the promoters of 11 mechanosensitive genes and that 5Aza treatment restored normal methylation patterns. Of the identified genes, HoxA5 and Klf3 encode transcription factors that contain cAMP response elements, suggesting that the methylation status of these loci could serve as a mechanosensitive master switch in gene expression. Together, our results demonstrate that d-flow controls epigenomic DNA methylation patterns in a DNMT-dependent manner, which in turn alters endothelial gene expression and induces atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/metabolism , DNA Methylation , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/physiopathology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Decitabine , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Homeodomain Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/physiopathology , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regional Blood Flow , Stress, Mechanical , Transcription Factors
4.
Nat Genet ; 46(2): 176-181, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24362818

ABSTRACT

Follicular lymphoma is an incurable malignancy, with transformation to an aggressive subtype representing a critical event during disease progression. Here we performed whole-genome or whole-exome sequencing on 10 follicular lymphoma-transformed follicular lymphoma pairs followed by deep sequencing of 28 genes in an extension cohort, and we report the key events and evolutionary processes governing tumor initiation and transformation. Tumor evolution occurred through either a 'rich' or 'sparse' ancestral common progenitor clone (CPC). We identified recurrent mutations in linker histone, JAK-STAT signaling, NF-κB signaling and B cell developmental genes. Longitudinal analyses identified early driver mutations in chromatin regulator genes (CREBBP, EZH2 and KMT2D (MLL2)), whereas mutations in EBF1 and regulators of NF-κB signaling (MYD88 and TNFAIP3) were gained at transformation. Collectively, this study provides new insights into the genetic basis of follicular lymphoma and the clonal dynamics of transformation and suggests that personalizing therapies to target key genetic alterations in the CPC represents an attractive therapeutic strategy.


Subject(s)
Cell Transformation, Neoplastic/genetics , Disease Progression , Genomics/methods , Lymphoma, Follicular/genetics , Lymphoma, Follicular/physiopathology , Base Sequence , CREB-Binding Protein/genetics , Cluster Analysis , Cohort Studies , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Exome/genetics , High-Throughput Nucleotide Sequencing , Histones/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Molecular Sequence Annotation , Molecular Sequence Data , Mutagenesis , Mutation/genetics , Myeloid Differentiation Factor 88/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Phylogeny , Polycomb Repressive Complex 2/genetics , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA , Trans-Activators/genetics , Tumor Necrosis Factor alpha-Induced Protein 3
5.
PLoS Genet ; 9(4): e1003417, 2013.
Article in English | MEDLINE | ID: mdl-23633960

ABSTRACT

H1 linker histones facilitate higher-order chromatin folding and are essential for mammalian development. To achieve high-resolution mapping of H1 variants H1d and H1c in embryonic stem cells (ESCs), we have established a knock-in system and shown that the N-terminally tagged H1 proteins are functionally interchangeable to their endogenous counterparts in vivo. H1d and H1c are depleted from GC- and gene-rich regions and active promoters, inversely correlated with H3K4me3, but positively correlated with H3K9me3 and associated with characteristic sequence features. Surprisingly, both H1d and H1c are significantly enriched at major satellites, which display increased nucleosome spacing compared with bulk chromatin. While also depleted at active promoters and enriched at major satellites, overexpressed H1(0) displays differential binding patterns in specific repetitive sequences compared with H1d and H1c. Depletion of H1c, H1d, and H1e causes pericentric chromocenter clustering and de-repression of major satellites. These results integrate the localization of an understudied type of chromatin proteins, namely the H1 variants, into the epigenome map of mouse ESCs, and we identify significant changes at pericentric heterochromatin upon depletion of this epigenetic mark.


Subject(s)
Chromatin/genetics , Embryonic Stem Cells , Heterochromatin/genetics , Histones/genetics , Animals , Chromatin Assembly and Disassembly/genetics , Chromosome Mapping , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Gene Knock-In Techniques , Histone-Lysine N-Methyltransferase , Mice
6.
PLoS Genet ; 8(5): e1002691, 2012.
Article in English | MEDLINE | ID: mdl-22589736

ABSTRACT

Pluripotent embryonic stem cells (ESCs) are known to possess a relatively open chromatin structure; yet, despite efforts to characterize the chromatin signatures of ESCs, the role of chromatin compaction in stem cell fate and function remains elusive. Linker histone H1 is important for higher-order chromatin folding and is essential for mammalian embryogenesis. To investigate the role of H1 and chromatin compaction in stem cell pluripotency and differentiation, we examine the differentiation of embryonic stem cells that are depleted of multiple H1 subtypes. H1c/H1d/H1e triple null ESCs are more resistant to spontaneous differentiation in adherent monolayer culture upon removal of leukemia inhibitory factor. Similarly, the majority of the triple-H1 null embryoid bodies (EBs) lack morphological structures representing the three germ layers and retain gene expression signatures characteristic of undifferentiated ESCs. Furthermore, upon neural differentiation of EBs, triple-H1 null cell cultures are deficient in neurite outgrowth and lack efficient activation of neural markers. Finally, we discover that triple-H1 null embryos and EBs fail to fully repress the expression of the pluripotency genes in comparison with wild-type controls and that H1 depletion impairs DNA methylation and changes of histone marks at promoter regions necessary for efficiently silencing pluripotency gene Oct4 during stem cell differentiation and embryogenesis. In summary, we demonstrate that H1 plays a critical role in pluripotent stem cell differentiation, and our results suggest that H1 and chromatin compaction may mediate pluripotent stem cell differentiation through epigenetic repression of the pluripotency genes.


Subject(s)
Cell Differentiation/genetics , Chromatin , Embryonic Stem Cells , Epigenesis, Genetic/genetics , Histones , Animals , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , DNA Methylation , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Histones/antagonists & inhibitors , Histones/genetics , Histones/metabolism , Mice , Neurites/metabolism , Neurons , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...