189 results on '"histone H3.3"'
Search Results
2. Histone H3.3 chaperone HIRA renders stress‐responsive genes poised for prospective lethal stresses in acquired tolerance.
- Author
-
Nagagaki, Yoshikazu, Kozakura, Yuji, Mahandaran, Theventhiran, Fumoto, Yukiko, Nakato, Ryuichiro, Shirahige, Katsuhiko, and Ishikawa, Fuyuki
- Subjects
- *
HEAT shock proteins , *HELA cells , *HORMESIS , *CHROMATIN , *FIBROBLASTS - Abstract
Appropriate responses to environmental challenges are imperative for the survival of all living organisms. Exposure to low‐dose stresses is recognized to yield increased cellular fitness, a phenomenon termed hormesis. However, our molecular understanding of how cells respond to low‐dose stress remains profoundly limited. Here we report that histone variant H3.3‐specific chaperone, HIRA, is required for acquired tolerance, where low‐dose heat stress exposure confers resistance to subsequent lethal heat stress. We found that human HIRA activates stress‐responsive genes, including HSP70, by depositing histone H3.3 following low‐dose stresses. These genes are also marked with histone H3 Lys‐4 trimethylation and H3 Lys‐9 acetylation, both active chromatin markers. Moreover, depletion of HIRA greatly diminished acquired tolerance, both in normal diploid fibroblasts and in HeLa cells. Collectively, our study revealed that HIRA is required for eliciting adaptive stress responses under environmental fluctuations and is a master regulator of stress tolerance. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. A knockout‐first model of H3f3a gene targeting leads to developmental lethality
- Author
-
Bush, Kelly, Cervantes, Vanessa, Yee, Jennifer Q, Klein, Rachel H, and Knoepfler, Paul S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Pediatric ,Genetics ,Biotechnology ,2.1 Biological and endogenous factors ,Generic health relevance ,Animals ,Female ,Mice ,Pregnancy ,Embryo ,Mammalian ,Fibroblasts ,Gene Targeting ,Histones ,Mice ,Inbred C57BL ,Mice ,Knockout ,Mutation ,development ,H3f3a ,H3f3b ,histone H3.3 ,mouse knockouts ,neurospheres ,Paediatrics and Reproductive Medicine ,Developmental Biology ,Biochemistry and cell biology - Abstract
Histone variant H3.3 is encoded by two genes, H3f3a and H3f3b, which can be expressed differentially depending on tissue type. Previous work in our lab has shown that knockout of H3f3b causes some neonatal lethality and infertility in mice, and chromosomal defects in mouse embryonic fibroblasts (MEFs). Studies of H3f3a and H3f3b null mice by others have produced generally similar phenotypes to what we found in our H3f3b nulls, but the relative impacts of the loss of either H3f3a or H3f3b have varied depending on the approach and genetic background. Here we used a knockout-first approach to target the H3f3a gene for inactivation in C57BL6 mice. Homozygous H3f3a targeting produced a lethal phenotype at or before birth. E13.5 null embryos had some potential morphological differences from WT littermates including smaller size and reduced head size. An E18.5 null embryo was smaller than its control littermates with several potential defects including small head and brain size as well as small lungs, which would be consistent with a late gestation lethal phenotype. Despite a reduction in H3.3 and total H3 protein levels, the only histone H3 post-translational modification in the small panel assessed that was significantly altered was the unique H3.3 mark phospho-Serine31, which was consistently increased in null neurospheres. H3f3a null neurospheres also exhibited consistent gene expression changes including in protocadherins. Overall, our findings are consistent with the model that there are differential, cell-type-specific contributions of H3f3a and H3f3b to H3.3 functions in epigenetic and developmental processes.
- Published
- 2023
4. Knockout tales: the versatile roles of histone H3.3 in development and disease
- Author
-
Klein, Rachel H and Knoepfler, Paul S
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Human Genome ,Brain Disorders ,Biotechnology ,Pediatric ,Genetics ,Cancer ,2.1 Biological and endogenous factors ,Generic health relevance ,Child ,Humans ,Histones ,Neoplasms ,Genomic Instability ,Brain ,Histone H3.3 ,H3f3a ,H3f3b ,Mouse knockouts - Abstract
Histone variant H3.3 plays novel roles in development as compared to canonical H3 proteins and is the most commonly mutated histone protein of any kind in human disease. Here we discuss how gene targeting studies of the two H3.3-coding genes H3f3a and H3f3b have provided important insights into H3.3 functions including in gametes as well as brain and lung development. Knockouts have also provided insights into the important roles of H3.3 in maintaining genomic stability and chromatin organization, processes that are also affected when H3.3 is mutated in human diseases such as pediatric tumors and neurodevelopmental syndromes. Overall, H3.3 is a unique histone linking development and disease via epigenomic machinery.
- Published
- 2023
5. Knockout tales: the versatile roles of histone H3.3 in development and disease
- Author
-
Rachel H. Klein and Paul S. Knoepfler
- Subjects
Histone H3.3 ,H3f3a ,H3f3b ,Mouse knockouts ,Genetics ,QH426-470 - Abstract
Abstract Histone variant H3.3 plays novel roles in development as compared to canonical H3 proteins and is the most commonly mutated histone protein of any kind in human disease. Here we discuss how gene targeting studies of the two H3.3-coding genes H3f3a and H3f3b have provided important insights into H3.3 functions including in gametes as well as brain and lung development. Knockouts have also provided insights into the important roles of H3.3 in maintaining genomic stability and chromatin organization, processes that are also affected when H3.3 is mutated in human diseases such as pediatric tumors and neurodevelopmental syndromes. Overall, H3.3 is a unique histone linking development and disease via epigenomic machinery.
- Published
- 2023
- Full Text
- View/download PDF
6. Dynamic changes in whole genome DNA methylation, chromatin and gene expression during mouse lens differentiation
- Author
-
William Chang, Yilin Zhao, Danielle Rayêe, Qing Xie, Masako Suzuki, Deyou Zheng, and Ales Cvekl
- Subjects
ATAC-seq ,Differentiation ,DNA methylation ,Gene regulation ,Histone H3.3 ,Lens ,Genetics ,QH426-470 - Abstract
Abstract Background Cellular differentiation is marked by temporally and spatially coordinated gene expression regulated at multiple levels. DNA methylation represents a universal mechanism to control chromatin organization and its accessibility. Cytosine methylation of CpG dinucleotides regulates binding of methylation-sensitive DNA-binding transcription factors within regulatory regions of transcription, including promoters and distal enhancers. Ocular lens differentiation represents an advantageous model system to examine these processes as lens comprises only two cell types, the proliferating lens epithelium and postmitotic lens fiber cells all originating from the epithelium. Results Using whole genome bisulfite sequencing (WGBS) and microdissected lenses, we investigated dynamics of DNA methylation and chromatin changes during mouse lens fiber and epithelium differentiation between embryos (E14.5) and newborns (P0.5). Histone H3.3 variant chromatin landscapes were also generated for both P0.5 lens epithelium and fibers by chromatin immunoprecipitation followed by next generation sequencing (ChIP-seq). Tissue-specific features of DNA methylation patterns are demonstrated via comparative studies with embryonic stem (ES) cells and neural progenitor cells (NPCs) at Nanog, Pou5f1, Sox2, Pax6 and Six3 loci. Comparisons with ATAC-seq and RNA-seq data demonstrate that reduced methylation is associated with increased expression of fiber cell abundant genes, including crystallins, intermediate filament (Bfsp1 and Bfsp2) and gap junction proteins (Gja3 and Gja8), marked by high levels of histone H3.3 within their transcribed regions. Interestingly, Pax6-binding sites exhibited predominantly DNA hypomethylation in lens chromatin. In vitro binding of Pax6 proteins showed Pax6’s ability to interact with sites containing one or two methylated CpG dinucleotides. Conclusions Our study has generated the first data on methylation changes between two different stages of mammalian lens development and linked these data with chromatin accessibility maps, presence of histone H3.3 and gene expression. Reduced DNA methylation correlates with expression of important genes involved in lens morphogenesis and lens fiber cell differentiation.
- Published
- 2023
- Full Text
- View/download PDF
7. Dominant effects of the histone mutant H3-L61R on Spt16-gene interactions in budding yeast
- Author
-
Alex Pablo-Kaiser, McKenzie G. Tucker, Grace A. Turner, Elijah G. Dilday, Avery G. Olmstead, Caroline L. Tackett, and Andrea A. Duina
- Subjects
histone h3 ,histone h3.3 ,fact complex ,spt16 ,yeast ,nucleosomes ,chromatin ,Genetics ,QH426-470 - Abstract
Recent studies have unveiled an association between an L61R substitution within the human histone H3.3 protein and the presentation of neurodevelopmental disorders in two patients. In both cases, the mutation responsible for this substitution is encoded by one allele of the H3F3A gene and, if this mutation is indeed responsible for the disease phenotypes, it must act in a dominant fashion since the genomes of these patients also harbour three other alleles encoding wild-type histone H3.3. In our previous work in yeast, we have shown that most amino acid substitutions at H3-L61 cause an accumulation of the Spt16 component of the yFACT histone chaperone complex at the 3’ end of transcribed genes, a defect we have attributed to impaired yFACT dissociation from chromatin following transcription. In those studies, however, the H3-L61R mutant had not been tested since it does not sustain viability when expressed as the sole source of histone H3 in cells. In the present work, we show that H3-L61R impairs proper Spt16 dissociation from genes when co-expressed with wild-type histone H3 in haploid cells as well as in diploid cells that express the mutant protein from one of four histone H3-encoding alleles. These results, combined with other studies linking loss of function mutations in human Spt16 and neurodevelopmental disorders, provide a possible molecular mechanism underlying the neurodevelopmental disorders seen in patients expressing the histone H3.3 L61R mutant.
- Published
- 2022
- Full Text
- View/download PDF
8. Multimodal MR imaging signatures to identify brain diffuse midline gliomas with H3 K27M mutation
- Author
-
Xiaorui Su, Yanhui Liu, Haoyu Wang, Ni Chen, Huaiqiang Sun, Xibiao Yang, Weina Wang, Simin Zhang, Xinyue Wan, Qiaoyue Tan, Qiang Yue, and Qiyong Gong
- Subjects
functional magnetic resonance imaging ,glioma ,histone H3.3 ,quantitative evaluation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Conventional MR imaging has limited value in identifying H3 K27M mutations. We aimed to investigate the capacity of quantitative MR imaging variables in identifying the H3 K27M mutation status of diffuse midline glioma. Materials and Methods Twenty‐three patients with H3 K27M mutation and thirty‐two wild‐type patients were recruited in this retrospective study, all of whom underwent multimodal MR imaging. Clinical data and quantitative MR imaging variables were explored by subgroup analysis stratified by age (juveniles and adults). Then, a logistic model for all patients was constructed to identify potential variables for predicting K27M mutation status. Besides, a retrospective validation set including 13 patients was recruited. The C‐index and F1 score were used to evaluate the performance of the prediction model. Results It turned out that patients with H3 K27M mutation were younger in the adult subgroup. In the mutation group, some relative apparent diffusion coefficient (rADC) histogram parameters and myo‐inositol/creatine plus phosphocreatine (Ins/tCr) ratio were lower than in the wild‐type group of both juveniles and adults (p
- Published
- 2022
- Full Text
- View/download PDF
9. Dynamic changes in whole genome DNA methylation, chromatin and gene expression during mouse lens differentiation.
- Author
-
Chang, William, Zhao, Yilin, Rayêe, Danielle, Xie, Qing, Suzuki, Masako, Zheng, Deyou, and Cvekl, Ales
- Subjects
DNA methylation ,GENE expression ,NUCLEOTIDE sequencing ,DNA methyltransferases ,IMMUNOPRECIPITATION ,CHROMATIN ,CYTOPLASMIC filaments - Abstract
Background: Cellular differentiation is marked by temporally and spatially coordinated gene expression regulated at multiple levels. DNA methylation represents a universal mechanism to control chromatin organization and its accessibility. Cytosine methylation of CpG dinucleotides regulates binding of methylation-sensitive DNA-binding transcription factors within regulatory regions of transcription, including promoters and distal enhancers. Ocular lens differentiation represents an advantageous model system to examine these processes as lens comprises only two cell types, the proliferating lens epithelium and postmitotic lens fiber cells all originating from the epithelium. Results: Using whole genome bisulfite sequencing (WGBS) and microdissected lenses, we investigated dynamics of DNA methylation and chromatin changes during mouse lens fiber and epithelium differentiation between embryos (E14.5) and newborns (P0.5). Histone H3.3 variant chromatin landscapes were also generated for both P0.5 lens epithelium and fibers by chromatin immunoprecipitation followed by next generation sequencing (ChIP-seq). Tissue-specific features of DNA methylation patterns are demonstrated via comparative studies with embryonic stem (ES) cells and neural progenitor cells (NPCs) at Nanog, Pou5f1, Sox2, Pax6 and Six3 loci. Comparisons with ATAC-seq and RNA-seq data demonstrate that reduced methylation is associated with increased expression of fiber cell abundant genes, including crystallins, intermediate filament (Bfsp1 and Bfsp2) and gap junction proteins (Gja3 and Gja8), marked by high levels of histone H3.3 within their transcribed regions. Interestingly, Pax6-binding sites exhibited predominantly DNA hypomethylation in lens chromatin. In vitro binding of Pax6 proteins showed Pax6's ability to interact with sites containing one or two methylated CpG dinucleotides. Conclusions: Our study has generated the first data on methylation changes between two different stages of mammalian lens development and linked these data with chromatin accessibility maps, presence of histone H3.3 and gene expression. Reduced DNA methylation correlates with expression of important genes involved in lens morphogenesis and lens fiber cell differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Chromatin mutations in pediatric high grade gliomas.
- Author
-
Voon, Hsiao P. J. and Wong, Lee H.
- Subjects
CHROMATIN ,GLIOMAS ,ANIMAL models in research - Abstract
Pediatric high grade gliomas (HGG) are lethal tumors which are currently untreatable. A number of recent studies have provided much needed insights into the mutations and mechanisms which drive oncogenesis in pediatric HGGs. It is now clear that mutations in chromatin proteins, particularly H3.3 and its associated chaperone complex (ATRX), are a hallmark feature of pediatric HGGs. We review the current literature on the normal roles of the ATRX/H3.3 complex and how these functions are disrupted by oncogenic mutations. We discuss the current clinical trials and pre-clinical models that target chromatin and DNA, and how these agents fit into the ATRX/H3.3 mutation model. As chromatin mutations are a relatively new discovery in pediatric HGGs, developing clear mechanistic insights are a key step to improving therapies for these tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. Dominant effects of the histone mutant H3-L61R on Spt16-gene interactions in budding yeast.
- Author
-
Pablo-Kaiser, Alex, Tucker, McKenzie G., Turner, Grace A., Dilday, Elijah G., Olmstead, Avery G., Tackett, Caroline L., and Duina, Andrea A.
- Subjects
MUTANT proteins ,YEAST ,HAPLOIDY ,ALLELES ,HISTONES ,GENOMES ,PHENOTYPES ,CHROMATIN-remodeling complexes - Abstract
Recent studies have unveiled an association between an L61R substitution within the human histone H3.3 protein and the presentation of neurodevelopmental disorders in two patients. In both cases, the mutation responsible for this substitution is encoded by one allele of the H3F3A gene and, if this mutation is indeed responsible for the disease phenotypes, it must act in a dominant fashion since the genomes of these patients also harbour three other alleles encoding wild-type histone H3.3. In our previous work in yeast, we have shown that most amino acid substitutions at H3-L61 cause an accumulation of the Spt16 component of the yFACT histone chaperone complex at the 3' end of transcribed genes, a defect we have attributed to impaired yFACT dissociation from chromatin following transcription. In those studies, however, the H3-L61R mutant had not been tested since it does not sustain viability when expressed as the sole source of histone H3 in cells. In the present work, we show that H3-L61R impairs proper Spt16 dissociation from genes when co-expressed with wild-type histone H3 in haploid cells as well as in diploid cells that express the mutant protein from one of four histone H3-encoding alleles. These results, combined with other studies linking loss of function mutations in human Spt16 and neurodevelopmental disorders, provide a possible molecular mechanism underlying the neurodevelopmental disorders seen in patients expressing the histone H3.3 L61R mutant. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
12. Identification of Plant Chromatin Interaction Networks Using IP-MS and co-IP.
- Author
-
Linares A, Härter NC, Rannou E, and Bischof S
- Subjects
- Immunoprecipitation methods, Mass Spectrometry methods, Protein Interaction Mapping methods, Protein Binding, Protein Interaction Maps, Proteomics methods, Chromatin metabolism, Chromatin genetics, Arabidopsis metabolism, Arabidopsis genetics, Arabidopsis Proteins metabolism
- Abstract
Proteins often act in concert to perform their function. Thus, the identification of protein complexes is crucial if we want to understand how they work. In this chapter, we present a highly sensitive protocol for the immunoprecipitation of nuclear chromatin-linked proteins in Arabidopsis thaliana that does not rely on time-consuming nuclei extraction. Interaction partners are identified using mass spectrometry and confirmed by co-immunoprecipitation. To help solubilize chromatin-bound proteins and eliminate nonspecific interactions of proteins binding the same DNA stretch, we include an enzymatic digestion step to remove DNA before immunoprecipitation. Our protocol offers a simplified process using optimized buffers, which facilitates quick and effective immunoprecipitation. The outcome is high-quality eluates that are ideal for identifying proteins through MS., (© 2025. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
13. Genome editing as a tool to explore transcriptional and epigenetic regulation in neural stem cells and brain cancer
- Author
-
Bressan, Raul Bardini, Smith, Andrew, and Pollard, Steven
- Subjects
618.92 ,paediatric glioblastoma ,pGBM ,CRISPR/Cas9 ,neural stem cells ,histone H3.3 - Abstract
Mammalian neural stem cell (NSC) lines provide a useful experimental model for basic and applied research across stem cell and developmental biology, regenerative medicine and neuroscience. NSCs are clonally expandable, genetically stable, and easily transfectable - experimental attributes compatible with functional genetic analyses. However, targeted genetic manipulations have not been reported for mammalian NSC lines. Here, we deploy the CRISPR/Cas9 technology and demonstrate a variety of diverse targeted genetic modifications in both mouse and human NSC lines such as: targeted transgene insertion at safe harbour loci; biallelic knockout of neurodevelopmental genes; knock-in of epitope tags and fluorescent reporters; and engineering of glioma driver mutations at endogenous proto-oncogenes. Leveraging these new optimised methods, we explored gene editing to model the earliest stages of paediatric gliomagenesis in primary human NSCs. Our data indicate that oncogenic mutations in histone H3.3 play a role in NSC transformation and may operate through suppression of replication induced senescence. By comparing cellular responses of NSC cultures from different compartments of the developing brain, we also identify differences in susceptibility to distinct H3.3 mutations that mirror the disease etiology. Altogether, our findings indicate that CRISPR/Cas9-assisted genome editing in NSC lines is a versatile tool to explore gene function in CNS development and cancer biology. Our project resulted in the creation of valuable human cellular models of paediatric gliomagenesis, which will allow us to further our understanding of the disease and carry out cell based drug discovery projects.
- Published
- 2018
14. Chromatin mutations in pediatric high grade gliomas
- Author
-
Hsiao P. J. Voon and Lee H. Wong
- Subjects
histone H3.3 ,H3.3 K27M ,DMG = diffuse midline glioma ,H3.3 G34R/V ,ATRX ,pediatric gliomas ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Pediatric high grade gliomas (HGG) are lethal tumors which are currently untreatable. A number of recent studies have provided much needed insights into the mutations and mechanisms which drive oncogenesis in pediatric HGGs. It is now clear that mutations in chromatin proteins, particularly H3.3 and its associated chaperone complex (ATRX), are a hallmark feature of pediatric HGGs. We review the current literature on the normal roles of the ATRX/H3.3 complex and how these functions are disrupted by oncogenic mutations. We discuss the current clinical trials and pre-clinical models that target chromatin and DNA, and how these agents fit into the ATRX/H3.3 mutation model. As chromatin mutations are a relatively new discovery in pediatric HGGs, developing clear mechanistic insights are a key step to improving therapies for these tumors.
- Published
- 2023
- Full Text
- View/download PDF
15. The role of histone modifications and telomere alterations in the pathogenesis of diffuse gliomas in adults and children
- Author
-
Lee, Julieann, Solomon, David A, and Tihan, Tarik
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Brain Disorders ,Cancer ,Human Genome ,Pediatric ,Pediatric Cancer ,Neurosciences ,Brain Cancer ,Genetics ,Rare Diseases ,2.1 Biological and endogenous factors ,4.2 Evaluation of markers and technologies ,Adult ,Animals ,Brain Neoplasms ,Child ,DNA Methylation ,Glioma ,Histone Code ,Histones ,Humans ,Telomere ,X-linked Nuclear Protein ,Histone H3.3 ,H3F3A ,HIST1H3B ,ATRX ,DAXX ,TERT ,Telomerase ,Alternative lengthening of telomeres ,Diffuse glioma ,Oligodendroglioma ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
Genetic profiling is an increasingly useful tool for sub-classification of gliomas in adults and children. Specific gene mutations, structural rearrangements, DNA methylation patterns, and gene expression profiles are now recognized to define molecular subgroups of gliomas that arise in distinct anatomic locations and patient age groups, and also provide a better prediction of clinical outcomes for glioma patients compared to histologic assessment alone. Understanding the role of these distinctive genetic alterations in gliomagenesis is also important for the development of potential targeted therapeutic interventions. Mutations including K27M and G34R/V that affect critical amino acids within the N-terminal tail of the histone H3 variants, H3.3 and H3.1 (encoded by H3F3A and HIST1H3B genes), are prime examples of mutations in diffuse gliomas with characteristic clinical associations that can help diagnostic classification and guide effective patient management. These histone H3 mutations frequently co-occur with inactivating mutations in ATRX in association with alternative lengthening of telomeres. Telomere length can also be maintained through upregulation of telomerase reverse transcriptase (TERT) expression driven by mutation within the TERT gene promoter region, an alteration most commonly found in oligodendrogliomas and primary glioblastomas arising in adults. Interestingly, the genetic alterations perturbing histone and telomere function in pediatric gliomas tend to be different from those present in adult tumors. We present a review of these mutations affecting the histone code and telomere length, highlighting their importance in prognosis and as targets for novel therapeutics in the treatment of diffuse gliomas.
- Published
- 2017
16. Diffuse Midline Gliomas with Histone H3‐K27M Mutation: A Series of 47 Cases Assessing the Spectrum of Morphologic Variation and Associated Genetic Alterations
- Author
-
Solomon, David A, Wood, Matthew D, Tihan, Tarik, Bollen, Andrew W, Gupta, Nalin, Phillips, Joanna JJ, and Perry, Arie
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Neurosciences ,Brain Cancer ,Brain Disorders ,Pediatric ,Cancer ,Rare Diseases ,Pediatric Cancer ,Genetics ,2.1 Biological and endogenous factors ,Adolescent ,Adult ,Aged ,Brain Neoplasms ,Child ,Child ,Preschool ,ErbB Receptors ,Female ,Gene Expression Regulation ,Neoplastic ,Genetic Association Studies ,Glioma ,Glycine ,Histones ,Humans ,Isocitrate Dehydrogenase ,Male ,Methionine ,Middle Aged ,Mutation ,PTEN Phosphohydrolase ,Tumor Suppressor Protein p53 ,X-linked Nuclear Protein ,Young Adult ,astrocytoma ,diffuse midline glioma ,diffuse intrinsic pontine glioma ,DIPG ,glioblastoma ,histone H3 ,1 ,3 ,H3F3A ,HIST1H3B ,K27M mutation ,histone H3.1 ,histone H3.3 ,Neurology & Neurosurgery ,Clinical sciences - Abstract
Somatic mutations of the H3F3A and HIST1H3B genes encoding the histone H3 variants, H3.3 and H3.1, were recently identified in high-grade gliomas arising in the thalamus, pons and spinal cord of children and young adults. However, the complete range of patients and locations in which these tumors arise, as well as the morphologic spectrum and associated genetic alterations remain undefined. Here, we describe a series of 47 diffuse midline gliomas with histone H3-K27M mutation. The 25 male and 22 female patients ranged in age from 2 to 65 years (median = 14). Tumors were centered not only in the pons, thalamus, and spinal cord, but also in the third ventricle, hypothalamus, pineal region and cerebellum. Patients with pontine tumors were younger (median = 7 years) than those with thalamic (median = 24 years) or spinal (median = 25 years) tumors. A wide morphologic spectrum was encountered including gliomas with giant cells, epithelioid and rhabdoid cells, primitive neuroectodermal tumor (PNET)-like foci, neuropil-like islands, pilomyxoid features, ependymal-like areas, sarcomatous transformation, ganglionic differentiation and pleomorphic xanthoastrocytoma (PXA)-like areas. In this series, histone H3-K27M mutation was mutually exclusive with IDH1 mutation and EGFR amplification, rarely co-occurred with BRAF-V600E mutation, and was commonly associated with p53 overexpression, ATRX loss (except in pontine gliomas), and monosomy 10.
- Published
- 2016
17. The H3.3 chaperone Hira complex orchestrates oocyte developmental competence.
- Author
-
Smith, Rowena, Susor, Andrej, Hao Ming, Tait, Janet, Conti, Marco, Zongliang Jiang, and Chih-Jen Lin
- Subjects
- *
OVUM , *RNA analysis , *CELL physiology , *GENE expression , *EMBRYOLOGY , *HISTONES - Abstract
Successful reproduction requires an oocyte competent to sustain early embryo development. By the end of oogenesis, the oocyte has entered a transcriptionally silenced state, the mechanisms and significance of which remain poorly understood. Histone H3.3, a histone H3 variant, has unique cell cycle-independent functions in chromatin structure and gene expression. Here, we have characterised the H3.3 chaperone Hira/Cabin1/Ubn1 complex, showing that loss of function of any of these subunits causes early embryogenesis failure in mouse. Transcriptome and nascent RNA analyses revealed that transcription is aberrantly silenced in mutant oocytes. Histone marks, including H3K4me3 and H3K9me3, are reduced and chromatin accessibility is impaired in Hira/Cabin1 mutants. Misregulated genes in mutant oocytes include Zscan4d, a two-cell specific gene involved in zygote genome activation. Overexpression of Zscan4 in the oocyte partially recapitulates the phenotypes of Hira mutants and Zscan4 knockdown in Cabin1 mutant oocytes partially restored their developmental potential, illustrating that temporal and spatial expression of Zscan4 is finetuned at the oocyte-to-embryo transition. Thus, the H3.3 chaperone Hira complex has a maternal effect function in oocyte developmental competence and embryogenesis, through modulating chromatin condensation and transcriptional quiescence. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
18. HIRA complex deposition of histone H3.3 is driven by histone tetramerization and histone-DNA binding.
- Author
-
Vogt A, Szurgot M, Gardner L, Schultz DC, and Marmorstein R
- Subjects
- Humans, Protein Binding, Nuclear Proteins, Molecular Chaperones, Histones metabolism, Cell Cycle Proteins metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins chemistry, Transcription Factors metabolism, Transcription Factors genetics, Histone Chaperones metabolism, Histone Chaperones chemistry, Protein Multimerization, DNA metabolism, DNA chemistry
- Abstract
The HIRA histone chaperone complex is comprised of four protein subunits: HIRA, UBN1, CABIN1, and transiently associated ASF1a. All four subunits have been demonstrated to play a role in the deposition of the histone variant H3.3 onto areas of actively transcribed euchromatin in cells. The mechanism by which these subunits function together to drive histone deposition has remained poorly understood. Here we present biochemical and biophysical data supporting a model whereby ASF1a delivers histone H3.3/H4 dimers to the HIRA complex, H3.3/H4 tetramerization drives the association of two HIRA/UBN1 complexes, and the affinity of the histones for DNA drives release of ASF1a and subsequent histone deposition. These findings have implications for understanding how other histone chaperone complexes may mediate histone deposition., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this paper., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
19. Chromatin alterations during the epididymal maturation of mouse sperm refine the paternally inherited epigenome.
- Author
-
Bedi, Yudhishtar S., Roach, Alexis N., Thomas, Kara N., Mehta, Nicole A., and Golding, Michael C.
- Subjects
- *
HISTONES , *SPERMATOZOA , *CHROMATIN , *POST-translational modification , *GENE enhancers , *EMBRYOLOGY , *SHEEP - Abstract
Background: Paternal lifestyle choices and male exposure history have a critical influence on the health and fitness of the next generation. Accordingly, defining the processes of germline programming is essential to resolving how the epigenetic memory of paternal experiences transmits to their offspring. Established dogma holds that all facets of chromatin organization and histone posttranslational modification are complete before sperm exits the testes. However, recent clinical and animal studies suggest that patterns of DNA methylation change during epididymal maturation. In this study, we used complementary proteomic and deep-sequencing approaches to test the hypothesis that sperm posttranslational histone modifications change during epididymal transit. Results: Using proteomic analysis to contrast immature spermatozoa and mature sperm isolated from the mouse epididymis, we find progressive changes in multiple histone posttranslational modifications, including H3K4me1, H3K27ac, H3K79me2, H3K64ac, H3K122ac, H4K16ac, H3K9me2, and H4K20me3. Interestingly, some of these changes only occurred on histone variant H3.3, and most involve chromatin modifications associated with gene enhancer activity. In contrast, the bivalent chromatin modifications, H3K4me3, and H3K27me3 remained constant. Using chromatin immunoprecipitation coupled with deep sequencing, we find that changes in histone h3, lysine 27 acetylation (H3K27ac) involve sharpening broad diffuse regions into narrow peaks centered on the promoter regions of genes driving embryonic development. Significantly, many of these regions overlap with broad domains of H3K4me3 in oocytes and ATAC-seq signatures of open chromatin identified in MII oocytes and sperm. In contrast, histone h3, lysine 9 dimethylation (H3K9me2) becomes enriched within the promoters of genes driving meiosis and in the distal enhancer regions of tissue-specific genes sequestered at the nuclear lamina. Maturing sperm contain the histone deacetylase enzymes HDAC1 and HDAC3, suggesting the NuRD complex may drive some of these changes. Finally, using Western blotting, we detected changes in chromatin modifications between caput and caudal sperm isolated from rams (Ovis aries), inferring changes in histone modifications are a shared feature of mammalian epididymal maturation. Conclusions: These data extend our understanding of germline programming and reveal that, in addition to trafficking noncoding RNAs, changes in histone posttranslational modifications are a core feature of epididymal maturation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. ZMYND11 variants are a novel cause of centrotemporal and generalised epilepsies with neurodevelopmental disorder.
- Author
-
Oates, Stephanie, Absoud, Michael, Goyal, Sushma, Bayley, Sophie, Baulcomb, Jennifer, Sims, Annemarie, Riddett, Amy, Allis, Katrina, Brasch‐Andersen, Charlotte, Balasubramanian, Meena, Bai, Renkui, Callewaert, Bert, Hüffmeier, Ulrike, Le Duc, Diana, Radtke, Maximilian, Korff, Christian, Kennedy, Joanna, Low, Karen, Møller, Rikke S., and Nielsen, Jens Erik Klint
- Subjects
- *
PARTIAL epilepsy , *NEURAL development , *MEDICAL genetics , *EPILEPSY , *PROGNOSIS , *EXOMES , *CHILDREN with epilepsy - Abstract
ZMYND11 is the critical gene in chromosome 10p15.3 microdeletion syndrome, a syndromic cause of intellectual disability. The phenotype of ZMYND11 variants has recently been extended to autism and seizures. We expand on the epilepsy phenotype of 20 individuals with pathogenic variants in ZMYND11. We obtained clinical descriptions of 16 new and nine published individuals, plus detailed case history of two children. New individuals were identified through GeneMatcher, ClinVar and the European Network for Therapies in Rare Epilepsy (NETRE). Genetic evaluation was performed using gene panels or exome sequencing; variants were classified using American College of Medical Genetics (ACMG) criteria. Individuals with ZMYND11 associated epilepsy fell into three groups: (i) atypical benign partial epilepsy or idiopathic focal epilepsy (n = 8); (ii) generalised epilepsies/infantile epileptic encephalopathy (n = 4); (iii) unclassified (n = 8). Seizure prognosis ranged from spontaneous remission to drug resistant. Neurodevelopmental deficits were invariable. Dysmorphic features were variable. Variants were distributed across the gene and mostly de novo with no precise genotype–phenotype correlation. ZMYND11 is one of a small group of chromatin reader genes associated in the pathogenesis of epilepsy, and specifically ABPE. More detailed epilepsy descriptions of larger cohorts and functional studies might reveal genotype–phenotype correlation. The epileptogenic mechanism may be linked to interaction with histone H3.3. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
21. Modulation of H3.3 chromatin assembly by PML: A way to regulate epigenetic inheritance.
- Author
-
Delbarre, Erwan and Janicki, Susan M.
- Subjects
- *
HEREDITY , *CHROMATIN , *HISTONES , *HETEROCHROMATIN , *CELL cycle , *PHYSIOLOGY - Abstract
Although the promyelocytic leukemia (PML) protein is renowned for regulating a wide range of cellular processes and as an essential component of PML nuclear bodies (PML‐NBs), the mechanisms through which it exerts its broad physiological impact are far from fully elucidated. Here, we review recent studies supporting an emerging view that PML's pleiotropic effects derive, at least partially, from its role in regulating histone H3.3 chromatin assembly, a critical epigenetic mechanism. These studies suggest that PML maintains heterochromatin organization by restraining H3.3 incorporation. Examination of PML's contribution to H3.3 chromatin assembly in the context of the cell cycle and PML‐NB assembly suggests that PML represses heterochromatic H3.3 deposition during S phase and that transcription and SUMOylation regulate PML's recruitment to heterochromatin. Elucidating PML' s contributions to H3.3‐mediated epigenetic regulation will provide insight into PML's expansive influence on cellular physiology and open new avenues for studying oncogenesis linked to PML malfunction. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
22. Diffuse midline glioma, H3-K27M mutant: Awareness leads to identification
- Author
-
Sadhana Tiwari, Ishita Pant, Sujata Chaturvedi, and Gurbachan Singh
- Subjects
Diffuse intrinsic pontine glioma ,H3-K27M-mutant ,histone H3 gene sequencing ,histone H3.1 ,histone H3.3 ,isocitrate dehydrogenase wildtype ,midline glioma ,pediatric brain tumor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Midline astrocytic neoplasms have distinct molecular characteristics, quite different from astrocytic neoplasms with similar morphology but not located in the midline. It is imperative that neuropathologists should be aware of the existence of these tumors, so they can be correctly diagnosed. Here, we discuss the case of a 14-year-old boy who presented with acute onset of vomiting followed by loss of consciousness. Subsequent magnetic resonance imaging revealed an ill-defined exophytic lesion arising from the brainstem and extending into the left cerebellopontine angle, with areas of hemorrhage and patchy restricted diffusion. The tumor was resected. Microscopy revealed medium-sized tumor cells in diffuse sheets, having round nuclei, granular chromatin, and scant cytoplasm. Microvascular and endothelial cell proliferation in small necrotic areas were seen. Mitosis was 0–1 per high-power field. By routine histopathological analysis, all features were consistent with the diagnosis of glioblastoma. Tumor cells were immunopositive for glial fibrillary acidic protein and isocitrate dehydrogenase-1 mutation (R132H), immunonegative for p53, and retained alpha thalassemia/mental retardation syndrome X-linked. It also showed a strong immunopositivity for H3-K27M mutation. A diagnosis of a diffuse midline glioma with H3-K27M mutation corresponding to the World Health Organization Grade IV was made. This case highlights the importance of exploring signature mutations in well-defined tumor categories such as H3-K27M-mutant diffuse midline glioma.
- Published
- 2019
- Full Text
- View/download PDF
23. Histone H3 variants at the root of metastasis
- Author
-
Didem Ilter, John Blenis, and Ana P. Gomes
- Subjects
tumor progression ,metastasis ,epigenetics ,histone h3.3 ,histone chaperones ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Chromatin remodeling is at the root of any cell fate decision, laying the foundation for the necessary reprogramming to occur. Our work shows histone H3 variants as a new addition to the ever-growing body of epigenetic regulators, one that is essential for cell fate transitions in carcinoma cells to promote tumor progression and metastasis.
- Published
- 2020
- Full Text
- View/download PDF
24. Histone H3.3 mutation in giant cell tumor of bone: an update in pathology.
- Author
-
Yamamoto, Hidetaka, Ishihara, Shin, Toda, Yu, and Oda, Yoshinao
- Subjects
- *
GIANT cell tumors , *BONE cancer , *BONE cells , *MULTINUCLEATED giant cells , *MOLECULAR pathology , *BIOMARKERS , *PATHOLOGY , *DENOSUMAB - Abstract
Giant cell tumor of bone (GCTB) is a locally aggressive bone tumor that frequently shows local recurrence and occasionally shows malignant transformation to high-grade sarcoma. Histologically, conventional GCTB is composed mainly of three types of cells: mononuclear neoplastic cells with an osteoblastic precursor phenotype, mononuclear histiocytic cells, and osteoclast-like multinucleated giant cells. These cells interact with each other via the RANKL-RANK axis and other mechanisms for tumor formation. The vast majority of GCTBs were recently revealed to harbor H3F3A p.G34W mutation, and a minor subset have H3F3A p.G34L, p.G34M, p.G34R, or p.G34V mutation. H3.3 G34W mutant-specific immunohistochemistry is a highly sensitive and specific surrogate marker for H3F3A p.G34W mutation in GCTB and thus useful for differential diagnoses of histological mimics. H3.3 mutant-specific immunohistochemistry has also contributed to the understanding of the bone-forming ability of neoplastic cells of GCTB and the remarkable new bone formation after treatment with denosumab, an inhibitor of RANKL. In primary and secondary malignant GCTBs, the H3F3A gene allele can be preserved or lost with malignant transformation. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
25. Knockout tales: the versatile roles of histone H3.3 in development and disease.
- Author
-
Klein, Rachel, Klein, Rachel, Knoepfler, Paul, Klein, Rachel, Klein, Rachel, and Knoepfler, Paul
- Abstract
Histone variant H3.3 plays novel roles in development as compared to canonical H3 proteins and is the most commonly mutated histone protein of any kind in human disease. Here we discuss how gene targeting studies of the two H3.3-coding genes H3f3a and H3f3b have provided important insights into H3.3 functions including in gametes as well as brain and lung development. Knockouts have also provided insights into the important roles of H3.3 in maintaining genomic stability and chromatin organization, processes that are also affected when H3.3 is mutated in human diseases such as pediatric tumors and neurodevelopmental syndromes. Overall, H3.3 is a unique histone linking development and disease via epigenomic machinery.
- Published
- 2023
26. Pak2 kinase promotes cellular senescence and organismal aging.
- Author
-
Jong-Sun Lee, Yan Mo, Haiyun Gan, Burgess, Rebecca J., Baker, Darren J., van Deursen, Jan M., and Zhiguo Zhang
- Subjects
- *
CELLULAR aging , *OLD age , *GENETIC regulation , *PROTEIN kinases - Abstract
Cellular senescence defines an irreversible cell growth arrest state linked to loss of tissue function and aging in mammals. This transition from proliferation to senescence is typically characterized by increased expression of the cell-cycle inhibitor p16INK4a and formation of senescence-associated heterochromatin foci (SAHF). SAHF formation depends on HIRA-mediated nucleosome assembly of histone H3.3, which is regulated by the serine/threonine protein kinase Pak2. However, it is unknown if Pak2 contributes to cellular senescence. Here, we show that depletion of Pak2 delayed oncogene-induced senescence in IMR90 human fibroblasts and oxidative stress-induced senescence of mouse embryonic fibroblasts (MEFs), whereas overexpression of Pak2 accelerated senescence of IMR90 cells. Importantly, depletion of Pak2 in BubR1 progeroid mice attenuated the onset of aging-associated phenotypes and extended life span. Pak2 is required for expression of genes involved in cellular senescence and regulated the deposition of newly synthesized H3.3 onto chromatin in senescent cells. Together, our results demonstrate that Pak2 is an important regulator of cellular senescence and organismal aging, in part through the regulation of gene expression and H3.3 nucleosome assembly. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
27. PML is recruited to heterochromatin during S phase and represses DAXX-mediated histone H3.3 chromatin assembly.
- Author
-
Shastrula, Prashanth Krishna, Sierra, Isabel, Zhong Deng, Keeney, Frederick, Hayden, James E., Lieberman, Paul M., and Janicki, Susan M.
- Subjects
- *
HETEROCHROMATIN , *HISTONES , *DNA replication - Abstract
The incorporation of the histone H3 variant, H3.3, into chromatin by the H3.3-specific chaperone, DAXX, and the ATP-dependent chromatin remodeling factor, ATRX, is a critical mechanism for silencing repetitive DNA. DAXX and ATRX are also components of promyelocytic nuclear bodies (PML-NBs), which have been identified as sites of H3.3 chromatin assembly. Here, we use a transgene array that can be visualized in single living cells to investigate the mechanisms that recruit PML-NB proteins (i.e. PML, DAXX, ATRX, and SUMO1/2/3) to heterochromatin and their functions in H3.3 chromatin assembly. We show that DAXX and PML are recruited to the array through distinct SUMOylation-dependent mechanisms. Additionally, PML is recruited during S phase and its depletion increases H3.3 deposition. Since this effect is abrogated when PML and DAXX are co-depleted, it suggests that PML represses DAXX-mediated H3.3 chromatin assembly. Taken together, these results suggest that, at heterochromatin, PML-NBs coordinate H3.3 chromatin assembly with DNA replication, which has important implications for understanding how transcriptional silencing is established and maintained. [ABSTRACT FROM AUTHOR]
- Published
- 2019
28. A knockout-first model of H3f3a gene targeting leads to developmental lethality
- Author
-
Kelly Bush, Vanessa Cervantes, Jennifer Q. Yee, Rachel H. Klein, and Paul S. Knoepfler
- Subjects
mouse knockouts ,Knockout ,Inbred C57BL ,histone H3.3 ,Article ,Histones ,Paediatrics and Reproductive Medicine ,Mice ,Endocrinology ,Pregnancy ,neurospheres ,Genetics ,Animals ,2.1 Biological and endogenous factors ,Aetiology ,development ,Pediatric ,Mammalian ,Cell Biology ,Fibroblasts ,Embryo ,Gene Targeting ,Mutation ,Female ,Biochemistry and Cell Biology ,H3f3b ,H3f3a ,Developmental Biology - Abstract
Histone variant H3.3 is encoded by two genes, H3f3a and H3f3b, which can be expressed differentially depending on tissue type. Previous work in our lab has shown that knockout of H3f3b causes some neonatal lethality and infertility in mice, and chromosomal defects in mouse embryonic fibroblasts (MEFs). Studies of H3f3a and H3f3b null mice by others have produced generally similar phenotypes to what we found in our H3f3b nulls, but the relative impacts of the loss of either H3f3a or H3f3b have varied depending on the approach and genetic background. Here we used a knockout-first approach to target the H3f3a gene for inactivation in C57BL6 mice. Homozygous H3f3a targeting produced a lethal phenotype at or before birth. E13.5 null embryos had some potential morphological differences from WT littermates including smaller size and reduced head size. An E18.5 null embryowas smaller than its control littermates with several potential defects including small head and brain size as well as small lungs, which would be consistent with a late gestation lethal phenotype. Despite a reduction in H3.3 and total H3 protein levels, the only histone H3 post-translational modification in the small panel assessed that was significantly altered was the unique H3.3 mark phospho-Serine31, which was consistently increased in null neurospheres. H3f3a null neurospheres also exhibited consistent gene expression changes including in protocadherins. Overall, our findings are consistent with the model that there are differential, cell-type-specific contributions of H3f3a and H3f3b to H3.3 functions in epigenetic and developmental processes.
- Published
- 2023
29. Polyomavirus minichromosomes: interactions with components of innate imunity
- Author
-
Satratzemis, Christos, Forstová, Jitka, and Trejbalová, Kateřina
- Subjects
DAXX ,mouse polyomavirus ,innate immunity ,histon H3.3 ,myší polyomavirus ,histone H3.3 ,vrozená imunita ,cGAS ,PML ,ATRX - Abstract
The genome of polyomaviruses is a circular dsDNA (double-stranded DNA) which is associated with cellular histones within virions and during the entire viral replication cycle. Given the structural similarity to eukaryotic chromatin, the complex of polyomaviral DNA with histones is called minichromosome. The chromatin state of minichromosomes influences viral transcription and replication which could be exploited by the host innate immune response. One of the components of innate immunity, that affects viral chromatin, is the non-canonical histone H3.3, its chaperone DAXX- ATRX (death domain associated protein 6-alpha-thalassemia, mental retardation X-linked syndrome) and protein complexes called PML (promyelocytic leukemia protein). In order to trigger the innate immune response, foreign and/or stress molecules have to detected. During mouse polyomavirus (MPyV) infection, the innate immune response is initiated via the DNA sensor cGAS (cyclic GMP- AMP synthase). In this master's thesis, the distribution of histone H3.3, its chaperone DAXX-ATRX and the PML protein was analyzed during infection with MPyV. Using mass spectrometry, the histone was detected within viral chromatin. The data suggest that the chaperone complex and PML are involved in the regulation of H3.3 incorporation into the chromatin...
- Published
- 2023
30. Histone H3 variants at the root of metastasis.
- Author
-
Ilter, Didem, Blenis, John, and Gomes, Ana P.
- Subjects
METASTASIS ,HISTONES ,CARCINOMA ,CANCER invasiveness ,CHROMATIN - Abstract
Chromatin remodeling is at the root of any cell fate decision, laying the foundation for the necessary reprogramming to occur. Our work shows histone H3 variants as a new addition to the ever-growing body of epigenetic regulators, one that is essential for cell fate transitions in carcinoma cells to promote tumor progression and metastasis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
31. Interferon stimulation creates chromatin marks and establishes transcriptional memory.
- Author
-
Patel, Mira C., Ryota Ouda, Anup Dey, Keiko Ozato, Rui Kamada, Kazuyasu Sakaguchi, Wenjing Yang, Yubo Zhang, Yanqin Yang, Yoshiyuki Wakabayashi, Jun Zhu, Takashi Fujita, and Tomohiko Tamura
- Subjects
- *
INTERFERONS , *CHROMATIN , *TRANSCRIPTION factors , *NATURAL immunity , *HISTONES , *EPIGENETICS - Abstract
Epigenetic memory for signal-dependent transcription has remained elusive. So far, the concept of epigenetic memory has been largely limited to cell-autonomous, preprogrammed processes such as development and metabolism. Here we show that IFNβ stimulation creates transcriptional memory in fibroblasts, conferring faster and greater transcription upon restimulation. The memory was inherited through multiple cell divisions and led to improved antiviral protection. Of ~2,000 IFNβ-stimulated genes (ISGs), about half exhibited memory, which we define as memory ISGs. The rest, designated nonmemory ISGs, did not show memory. Surprisingly, mechanistic analysis showed that IFN memory was not due to enhanced IFN signaling or retention of transcription factors on the ISGs. We demonstrated that this memory was attributed to accelerated recruitment of RNA polymerase II and transcription/chromatin factors, which coincided with acquisition of the histone H3.3 and H3K36me3 chromatin marks on memory ISGs. Similar memory was observed in bone marrow macrophages after IFNγ stimulation, suggesting that IFN stimulation modifies the shape of the innate immune response. Together, external signals can establish epigenetic memory in mammalian cells that imparts lasting adaptive performance upon various somatic cells. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
32. Tumor suppressor functions of DAXX through histone H3.3/H3K9me3 pathway in pancreatic NETs.
- Author
-
Hiroki Ueda, Yoshimitsu Akiyama, Shu Shimada, Kaoru Mogushi, Misaki Serizawa, Satoshi Matsumura, Yusuke Mitsunori, Arihiro Aihara, Daisuke Ban, Takanori Ochiai, Atsushi Kudo, Minoru Tanabe, and Shinji Tanaka
- Subjects
- *
NEUROENDOCRINE tumors , *TUMOR suppressor genes , *PROTEIN expression , *HISTONES , *IMMUNOPRECIPITATION , *IMMUNOHISTOCHEMISTRY , *TUMOR treatment - Abstract
Pancreatic neuroendocrine tumors (PanNETs) have considerable malignant potential. Frequent somatic mutations and loss of DAXX protein expression have been found in PanNETs. DAXX is known as a transcriptional repressor; however, molecular functions underlying DAXX loss remain unclear in PanNETs. We evaluated DAXX expression by immunohistochemistry in 44 PanNETs. DAXX-knockdown (KD) and -knockout (KO) PanNET cells were analyzed for in vitro and vivo. The target genes were screened by microarray and chromatin immunoprecipitation (ChIP) assays for DAXX, histone H3.3 and H3K9me3 complex. In clinicopathological features, low DAXX expression was signifcantly correlated with nonfunctional tumors, higher Ki-67 index and WHO grade. Microarray and ChIP assays of DAXX-KD/KO identifed 12 genes as the direct targets of DAXX transcriptional repressor. Among them, expression of fve genes including STC2 was suppressed by DAXX/H3.3/H3K9me3 pathway. DAXX-KD/KO cells enhanced sphere forming activity, but its effect was suppressed by knockdown of STC2. In xenograft models, tumorigenicity and tumor vessel density were signifcantly increased in DAXX-KO cells with high expression of STC2. Clinically, higher recurrence rate was recognized in PanNETs with low expression of DAXX and high expression of STC2 than others (P = 0.018). Our data suggest that DAXX plays as a tumor suppressor and DAXX/H3.3 complex suppresses target genes by promoting H3K9me3 in PanNETs. Combination of DAXX loss and its target gene STC2 overexpression might be effective biomarkers and therapeutic candidates. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
33. Chromatin remodeling of histone H3 variants by DDM1 underlies epigenetic inheritance of DNA methylation.
- Author
-
Lee, Seung Cho, Adams, Dexter W., Ipsaro, Jonathan J., Cahn, Jonathan, Lynn, Jason, Kim, Hyun-Soo, Berube, Benjamin, Major, Viktoria, Calarco, Joseph P., LeBlanc, Chantal, Bhattacharjee, Sonali, Ramu, Umamaheswari, Grimanelli, Daniel, Jacob, Yannick, Voigt, Philipp, Joshua-Tor, Leemor, and Martienssen, Robert A.
- Subjects
- *
HEREDITY , *DNA methylation , *HISTONE methylation , *HISTONES , *CHROMATIN , *CELL cycle , *GENOMICS , *METHYLTRANSFERASES - Abstract
Nucleosomes block access to DNA methyltransferase, unless they are remodeled by DECREASE in DNA METHYLATION 1 (DDM1LSH/HELLS), a Snf2-like master regulator of epigenetic inheritance. We show that DDM1 promotes replacement of histone variant H3.3 by H3.1. In ddm1 mutants, DNA methylation is partly restored by loss of the H3.3 chaperone HIRA, while the H3.1 chaperone CAF-1 becomes essential. The single-particle cryo-EM structure at 3.2 Å of DDM1 with a variant nucleosome reveals engagement with histone H3.3 near residues required for assembly and with the unmodified H4 tail. An N-terminal autoinhibitory domain inhibits activity, while a disulfide bond in the helicase domain supports activity. DDM1 co-localizes with H3.1 and H3.3 during the cell cycle, and with the DNA methyltransferase MET1Dnmt1, but is blocked by H4K16 acetylation. The male germline H3.3 variant MGH3/HTR10 is resistant to remodeling by DDM1 and acts as a placeholder nucleosome in sperm cells for epigenetic inheritance. [Display omitted] • DDM1 remodeling promotes H3.1 H2A.W deposition and DNA methylation • Cryo-EM structure reveals contacts with variant H3 residues and deacetylated H4 tails • An autoinhibitory N terminus and helicase S–S bond regulate DDM1 activity • Male germline H3.3 placeholder variant contributes to epigenetic inheritance How DDM1 drives DNA methylation and epigenetic inheritance is unknown. Through high-resolution genomic and structural analyses, it is shown how DDM1 is recruited to nucleosomes and provides MET1 access to naked replicated DNA, ensuring epigenetic inheritance of DNA methylation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
34. The histone H3.3 chaperone HIRA restrains erythroid-biased differentiation of adult hematopoietic stem cells
- Author
-
Richard H. Chapple, Matthew C. Hill, James F. Martin, Angelique Lin, Nicolas L. Young, Matthew V. Holt, Ayumi Kitano, Xiangguo Shi, Tianyuan Hu, Daisuke Nakada, Yu-Jung Tseng, Jonathan F. Tiessen, Kevin A. Hoegenauer, and Rebecca Murdaugh
- Subjects
Cell division ,Cell Cycle Proteins ,Mice, Transgenic ,Biology ,Biochemistry ,histone H3.3 ,Article ,Histones ,Histone H3 ,Erythroid Cells ,Gene expression ,Genetics ,medicine ,Animals ,Histone Chaperones ,Epigenetics ,RNA-Seq ,Cell Self Renewal ,Mice, Knockout ,epigenetics ,Gene Expression Profiling ,Age Factors ,Hematopoietic stem cell ,Cell Differentiation ,Cell Biology ,differentiation ,Hematopoietic Stem Cells ,Hira ,hematopoiesis ,Chromatin ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,Adult Stem Cells ,medicine.anatomical_structure ,Gene Ontology ,Animals, Newborn ,hematopoietic stem cell ,Stem cell ,polycomb ,erythropoiesis ,Developmental Biology ,Transcription Factors - Abstract
Summary Histone variants contribute to the complexity of the chromatin landscape and play an integral role in defining DNA domains and regulating gene expression. The histone H3 variant H3.3 is incorporated into genic elements independent of DNA replication by its chaperone HIRA. Here we demonstrate that Hira is required for the self-renewal of adult hematopoietic stem cells (HSCs) and to restrain erythroid differentiation. Deletion of Hira led to rapid depletion of HSCs while differentiated hematopoietic cells remained largely unaffected. Depletion of HSCs after Hira deletion was accompanied by increased expression of bivalent and erythroid genes, which was exacerbated upon cell division and paralleled increased erythroid differentiation. Assessing H3.3 occupancy identified a subset of polycomb-repressed chromatin in HSCs that depends on HIRA to maintain the inaccessible, H3.3-occupied state for gene repression. HIRA-dependent H3.3 incorporation thus defines distinct repressive chromatin that represses erythroid differentiation of HSCs., Graphical abstract, Highlights • Deletion of Hira depletes adult HSCs but not fetal liver HSCs • HIRA represses hematopoietic development and erythropoiesis genes in HSCs • Cell division exacerbates gene derepression in Hira-deficient HSCs • H3.3 deposition and the closed state of polycomb-repressed chromatin depend on HIRA, Murdaugh et al. demonstrate that deletion of the histone H3.3 chaperone Hira depletes adult but not fetal liver HSCs by inducing differentiation. Hira regulates H3.3 deposition at a subset of polycomb-repressed chromatin that encodes genes involved in hematopoietic development, which becomes accessible and derepressed in the absence of Hira.
- Published
- 2021
35. A Molecular Prospective for HIRA Complex Assembly and H3.3-Specific Histone Chaperone Function.
- Author
-
Ricketts, M. Daniel and Marmorstein, Ronen
- Subjects
- *
MOLECULAR chaperones , *HISTONES , *POST-translational modification , *CHROMATIN , *CELL cycle regulation - Abstract
Incorporation of variant histone sequences, in addition to post-translational modification of histones, serves to modulate the chromatin environment. Different histone chaperone proteins mediate the storage and chromatin deposition of variant histones. Although the two non-centromeric histone H3 variants, H3.1 and H3.3, differ by only 5 aa, replacement of histone H3.1 with H3.3 can modulate the transcription for highly expressed and developmentally required genes, lead to the formation of repressive heterochromatin, or aid in DNA and chromatin repair. The human histone cell cycle regulator (HIRA) complex composed of HIRA, ubinuclein-1, CABIN1, and transiently anti-silencing function 1, forms one of the two complexes that bind and deposit H3.3/H4 into chromatin. A number of recent biochemical and structural studies have revealed important details underlying how these proteins assemble and function together as a multiprotein H3.3-specific histone chaperone complex. Here, we present a review of existing data and present a new model for the assembly of the HIRA complex and for the HIRA-mediated incorporation of H3.3/H4 into chromatin. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
36. Genetic mosaics and time-lapse imaging identify functions of histone H3.3 residues in mouse oocytes and embryos.
- Author
-
Zhou, Liquan, Baibakov, Boris, Canagarajah, Bertram, Dean, Jurrien, and Bo Xiong
- Subjects
- *
EMBRYOS , *HISTONES , *MOSAICISM - Abstract
During development from oocyte to embryo, genetic programs in mouse germ cells are reshaped by chromatin remodeling to orchestrate the onset of development. Epigenetic modifications of specific amino acid residues of core histones and their isoforms can dramatically alter activation and suppression of gene expression. H3.3 is a histone H3 variant that plays essential roles in mouse oocytes and early embryos, but the functional role of individual amino acid residues has been unclear because of technical hurdles. Here, we describe two strategies that successfully investigated the functions of three individual H3.3 residues in oogenesis, cleavagestage embryogenesis and early development. We first generated genetic mosaic ovaries and blastocysts with stochastic expression of wild-type or mutant H3.3 alleles and showed dominant negative effects of H3.3R26 and H3.3K27 in modulating oogenesis and partitioning cells to the inner cell mass of the early embryo. Timelapse imaging assays also revealed the essential roles of H3.3K56 in efficient H2B incorporation and paternal pronuclei formation. Application of these strategies can be extended to investigate roles of additional H3.3 residues and has implications for use in other developmental systems. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
37. Enhancer regions show high histone H3.3 turnover that changes during differentiation
- Author
-
Aimee M Deaton, Mariluz Gómez-Rodríguez, Jakub Mieczkowski, Michael Y Tolstorukov, Sharmistha Kundu, Ruslan I Sadreyev, Lars ET Jansen, and Robert E Kingston
- Subjects
chromatin ,histone H3.3 ,turnover ,differentiation ,stem cells ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The organization of DNA into chromatin is dynamic; nucleosomes are frequently displaced to facilitate the ability of regulatory proteins to access specific DNA elements. To gain insight into nucleosome dynamics, and to follow how dynamics change during differentiation, we used a technique called time-ChIP to quantitatively assess histone H3.3 turnover genome-wide during differentiation of mouse ESCs. We found that, without prior assumptions, high turnover could be used to identify regions involved in gene regulation. High turnover was seen at enhancers, as observed previously, with particularly high turnover at super-enhancers. In contrast, regions associated with the repressive Polycomb-Group showed low turnover in ESCs. Turnover correlated with DNA accessibility. Upon differentiation, numerous changes in H3.3 turnover rates were observed, the majority of which occurred at enhancers. Thus, time-ChIP measurement of histone turnover shows that active enhancers are unusually dynamic in ESCs and changes in highly dynamic nucleosomes predominate at enhancers during differentiation.
- Published
- 2016
- Full Text
- View/download PDF
38. Chromatin alterations during the epididymal maturation of mouse sperm refine the paternally inherited epigenome
- Author
-
Yudhishtar S. Bedi, Alexis N. Roach, Kara N. Thomas, Nicole A. Mehta, and Michael C. Golding
- Subjects
Epididymis ,Male ,Proteomics ,Epididymal maturation ,Research ,Histone h3.3 ,QH426-470 ,DNA Methylation ,H3K27ac ,Chromatin structure ,Spermatozoa ,Sperm ,H3K9me2 ,Chromatin ,Developmental programming ,Epigenome ,Mice ,Genetics ,Paternal Inheritance ,Animals ,Paternal epigenetic inheritance ,Molecular Biology ,Gene enhancers - Abstract
Background Paternal lifestyle choices and male exposure history have a critical influence on the health and fitness of the next generation. Accordingly, defining the processes of germline programming is essential to resolving how the epigenetic memory of paternal experiences transmits to their offspring. Established dogma holds that all facets of chromatin organization and histone posttranslational modification are complete before sperm exits the testes. However, recent clinical and animal studies suggest that patterns of DNA methylation change during epididymal maturation. In this study, we used complementary proteomic and deep-sequencing approaches to test the hypothesis that sperm posttranslational histone modifications change during epididymal transit. Results Using proteomic analysis to contrast immature spermatozoa and mature sperm isolated from the mouse epididymis, we find progressive changes in multiple histone posttranslational modifications, including H3K4me1, H3K27ac, H3K79me2, H3K64ac, H3K122ac, H4K16ac, H3K9me2, and H4K20me3. Interestingly, some of these changes only occurred on histone variant H3.3, and most involve chromatin modifications associated with gene enhancer activity. In contrast, the bivalent chromatin modifications, H3K4me3, and H3K27me3 remained constant. Using chromatin immunoprecipitation coupled with deep sequencing, we find that changes in histone h3, lysine 27 acetylation (H3K27ac) involve sharpening broad diffuse regions into narrow peaks centered on the promoter regions of genes driving embryonic development. Significantly, many of these regions overlap with broad domains of H3K4me3 in oocytes and ATAC-seq signatures of open chromatin identified in MII oocytes and sperm. In contrast, histone h3, lysine 9 dimethylation (H3K9me2) becomes enriched within the promoters of genes driving meiosis and in the distal enhancer regions of tissue-specific genes sequestered at the nuclear lamina. Maturing sperm contain the histone deacetylase enzymes HDAC1 and HDAC3, suggesting the NuRD complex may drive some of these changes. Finally, using Western blotting, we detected changes in chromatin modifications between caput and caudal sperm isolated from rams (Ovis aries), inferring changes in histone modifications are a shared feature of mammalian epididymal maturation. Conclusions These data extend our understanding of germline programming and reveal that, in addition to trafficking noncoding RNAs, changes in histone posttranslational modifications are a core feature of epididymal maturation.
- Published
- 2022
39. The H3.3K27M oncohistone affects replication stress outcome and provokes genomic instability in pediatric glioma
- Author
-
Jonas Seiler, Marin T Mondria, Irena Bockaj, Marcel A. T. M. van Vugt, Bjorn Bakker, Diana C.J. Spierings, Femke Ringnalda, Saskia L Meuleman, Mathilde J.C. Broekhuis, Ulrich Schüller, Floris Foijer, Hans Clevers, Colin Stok, Tosca. E. I. Martini, Eduardo Sabino de Camargo Magalhães, Hilda van den Bos, Yannick P Kok, Tiny G. J. Meeuwsen-de Boer, Inna Armandari, René Wardenaar, Petra L. Bakker, Sophia W.M. Bruggeman, Hubrecht Institute for Developmental Biology and Stem Cell Research, Stem Cell Aging Leukemia and Lymphoma (SALL), Damage and Repair in Cancer Development and Cancer Treatment (DARE), Guided Treatment in Optimal Selected Cancer Patients (GUTS), Restoring Organ Function by Means of Regenerative Medicine (REGENERATE), and Obstetrics and gynaecology
- Subjects
Genome instability ,CHROMATIN ,Cancer Research ,Mutant ,QH426-470 ,Biochemistry ,Pediatrics ,Histones ,Antibiotics ,Medicine and Health Sciences ,Cell Cycle and Cell Division ,Child ,Neurological Tumors ,Genetics (clinical) ,CHAPERONE ,Brain Neoplasms ,Chromosome Biology ,Antimicrobials ,HIGH-GRADE ,Drugs ,Glioma ,Chromatin ,Cell biology ,Gene Expression Regulation, Neoplastic ,Nucleic acids ,Histone ,Oncology ,Neurology ,Cell Processes ,Doxycycline ,Cellular Structures and Organelles ,HISTONE H3.3 ,CHROMOSOME SEGREGATION ,Research Article ,DNA Replication ,DNA damage ,INTRINSIC PONTINE GLIOMAS ,Mitosis ,Biology ,Microbiology ,Genomic Instability ,MCM PROTEINS ,COMMON FRAGILE SITES ,Antimalarials ,Malignant Tumors ,Microbial Control ,Nuclear Bodies ,DNA-binding proteins ,medicine ,Genetics ,Humans ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Pharmacology ,Cell Nucleus ,Biology and life sciences ,HUMAN-CELLS ,DNA replication ,Proteins ,Cancers and Neoplasms ,DNA ,Cell Biology ,medicine.disease ,DNA-DAMAGE ,biology.protein - Abstract
While comprehensive molecular profiling of histone H3.3 mutant pediatric high-grade glioma has revealed extensive dysregulation of the chromatin landscape, the exact mechanisms driving tumor formation remain poorly understood. Since H3.3 mutant gliomas also exhibit high levels of copy number alterations, we set out to address if the H3.3K27M oncohistone leads to destabilization of the genome. Hereto, we established a cell culture model allowing inducible H3.3K27M expression and observed an increase in mitotic abnormalities. We also found enhanced interaction of DNA replication factors with H3.3K27M during mitosis, indicating replication defects. Further functional analyses revealed increased genomic instability upon replication stress, as represented by mitotic bulky and ultrafine DNA bridges. This co-occurred with suboptimal 53BP1 nuclear body formation after mitosis in vitro, and in human glioma. Finally, we observed a decrease in ultrafine DNA bridges following deletion of the K27M mutant H3F3A allele in primary high-grade glioma cells. Together, our data uncover a role for H3.3 in DNA replication under stress conditions that is altered by the K27M mutation, promoting genomic instability and potentially glioma development., Author summary The childhood brain cancer high-grade glioma is a devastating disease that almost invariably ends in the death of the patient. The discovery that mutations in histone H3 variants are predominant in this type of cancer created great interest into the mechanism of transformation of these so-called oncohistones. Yet whereas most research is directed at understanding the role of mutant H3 in deregulating gene expression, we studied if and how it contributes to genomic instability–a research avenue that remains mostly unexplored. We discovered that the H3.3K27M oncohistone increases sensitivity to replication stress during S-phase, ultimately leading to mitotic aberrancies. This can explain why childhood high-grade glioma is among the most genomically unstable cancers.
- Published
- 2021
- Full Text
- View/download PDF
40. Significance of Histone H3.3 (G34W)-Mutant Protein in Pathological Diagnosis of Giant Cell Tumor of Bone.
- Author
-
Miskad UA, Syamsul F, Dahlan H, Sungowati NK, Achmad D, and Johan MP
- Subjects
- Humans, Histones genetics, Histones metabolism, Mutant Proteins metabolism, Cross-Sectional Studies, Giant Cell Tumor of Bone diagnosis, Giant Cell Tumor of Bone genetics, Giant Cell Tumor of Bone pathology, Bone Neoplasms diagnosis, Bone Neoplasms genetics, Bone Neoplasms metabolism
- Abstract
Objective: This study aimed to examine the expression of Histone H3.3 glycine 34 to tryptophan (G34W) mutant protein in Giant Cell Tumor of Bone (GCTB)., Methods: This analytic observation research used a cross-sectional study design on 71 bone tumors. The cases involved 54 tissue samples diagnosed as GCBT. It was divided into GCTB primer (n=37), recurrent GCTB (n=5), GCTB with metastasis (n=9), and malignant GCTB (n=3). There were 17 samples mimics of GCTB also tested, including chondroblastoma (n=1), giant cell reparative granuloma (n=2), giant cell of tendon sheath (n=7), chondromyxoid fibroma (n=2), aneurysmal bone cyst (n=2), and giant cell-rich osteosarcoma (n=3). The Immunohistochemistry was used to evaluate the expression of G34W-mutated protein in these bone tumors., Result: The representation H3.3 (G34W) was expressed in the nuclei of mononuclear stromal cells but not stained on osteoclast-like giant cells. This study was analyzed by the Chi-square test, Fisher's test, specificity test, and sensitivity test. We obtained p = 0.001 for Histone H3.3 (G34W) mutant expression in GCTB vs Non-GCTB. Statistically, there was no significant difference in the expression level of Histone H3.3 (G34W) in the GCTB and its variants p-value = 0.183. We also obtained that the specificity of Histone H3.3 expression on GCTB was 100% and the sensitivity of Histone H3.3 on GCTB was 77.8%., Conclusion: Histon H3.3 mutant as a mutated driver gene in an Indonesian GCTB can assist to diagnose GCTB and compare it from other bone tumors.
- Published
- 2023
- Full Text
- View/download PDF
41. Histone chaperone ASF1B promotes human β -cell proliferation via recruitment of histone H3.3.
- Author
-
Paul, Pradyut K., Rabaglia, Mary E., Wang, Chen-Yu, Stapleton, Donald S., Leng, Ning, Kendziorski, Christina, Lewis, Peter W., Keller, Mark P., and Attie, Alan D.
- Published
- 2016
- Full Text
- View/download PDF
42. Histone H3.3 promotes IgV gene diversification by enhancing formation of AID-accessible single-stranded DNA.
- Author
-
Romanello, Marina, Schiavone, Davide, Frey, Alexander, and Sale, Julian E
- Subjects
- *
IMMUNOGLOBULINS , *BIODIVERSITY , *CHROMATIN , *URACIL , *SINGLE-stranded DNA , *PROMOTERS (Genetics) , *BIOCHEMICAL substrates - Abstract
Immunoglobulin diversification is driven by activation-induced deaminase ( AID), which converts cytidine to uracil within the Ig variable (IgV) regions. Central to the recruitment of AID to the IgV genes are factors that regulate the generation of single-stranded DNA (ss DNA), the enzymatic substrate of AID. Here, we report that chicken DT40 cells lacking variant histone H3.3 exhibit reduced IgV sequence diversification. We show that this results from impairment of the ability of AID to access the IgV genes due to reduced formation of ss DNA during IgV transcription. Loss of H3.3 also diminishes IgV R-loop formation. However, reducing IgV R-loops by RNase HI overexpression in wild-type cells does not affect IgV diversification, showing that these structures are not necessary intermediates for AID access. Importantly, the reduction in the formation of AID-accessible ss DNA in cells lacking H3.3 is independent of any effect on the level of transcription or the kinetics of RNAPII elongation, suggesting the presence of H3.3 in the nucleosomes of the IgV genes increases the chances of the IgV DNA becoming single-stranded, thereby creating an effective AID substrate. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
43. O-linked N-acetylglucosamine transferase (OGT) interacts with the histone chaperone HIRA complex and regulates nucleosome assembly and cellular senescence.
- Author
-
Jong-Sun Lee and Zhiguo Zhang
- Subjects
- *
N-acetylglucosamine , *TRANSFERASES , *CHROMATIN , *CELL cycle , *CARRIER proteins , *GENE expression - Abstract
The histone chaperone HIRA complex, consisting of histone cell cycle regulator (HIRA), Ubinuclein1 (UBN1), and calcineurin binding protein 1 (CABIN1), deposits histone variant H3.3 to genic regions and regulates gene expression in various cellular processes, including cellular senescence. How HIRA-mediated nucleosome assembly of H3.3-H4 is regulated remains not well understood. Here, we show that O-linked N-acetylglucosamine (GlcNAc) transferase (OGT), an enzyme that catalyzes O-GlcNAcylation of serine or threonine residues, interacts with UBN1, modifies HIRA, and promotes nucleosome assembly of H3.3. Depletion of OGT or expression of the HIRA S231A O-GlcNAcylation-deficient mutant compromises formation of the HIRA-H3.3 complex and H3.3 nucleosome assembly. Importantly, OGT depletion or expression of the HIRA S231A mutant delays premature cellular senescence in primary human fibroblasts, whereas overexpression of OGT accelerates senescence. Taken together, these results support a model in which OGT modifies HIRA to regulate HIRA-H3.3 complex formation and H3.3 nucleosome assembly and reveal the mechanism by which OGT functions in cellular senescence. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
44. Atrx promotes heterochromatin formation at retrotransposons.
- Author
-
Sadic, Dennis, Schmidt, Katharina, Groh, Sophia, Kondofersky, Ivan, Ellwart, Joachim, Fuchs, Christiane, Theis, Fabian J, and Schotta, Gunnar
- Abstract
More than 50% of mammalian genomes consist of retrotransposon sequences. Silencing of retrotransposons by heterochromatin is essential to ensure genomic stability and transcriptional integrity. Here, we identified a short sequence element in intracisternal A particle ( IAP) retrotransposons that is sufficient to trigger heterochromatin formation. We used this sequence in a genome-wide sh RNA screen and identified the chromatin remodeler Atrx as a novel regulator of IAP silencing. Atrx binds to IAP elements and is necessary for efficient heterochromatin formation. In addition, Atrx facilitates a robust and largely inaccessible heterochromatin structure as Atrx knockout cells display increased chromatin accessibility at retrotransposons and non-repetitive heterochromatic loci. In summary, we demonstrate a direct role of Atrx in the establishment and robust maintenance of heterochromatin. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
45. Intellectual disability-associated dBRWD3 regulates gene expression through inhibition of HIRA/ YEM-mediated chromatin deposition of histone H3.3.
- Author
-
Chen, Wei‐Yu, Shih, Hsueh‐Tzu, Liu, Kuei‐Yan, Shih, Zong‐Siou, Chen, Li‐Kai, Tsai, Tsung‐Han, Chen, Mei‐Ju, Liu, Hsuan, Tan, Bertrand Chin‐Ming, Chen, Chien‐Yu, Lee, Hsiu‐Hsiang, Loppin, Benjamin, Aït‐Ahmed, Ounissa, and Wu, June‐Tai
- Abstract
Many causal mutations of intellectual disability have been found in genes involved in epigenetic regulations. Replication-independent deposition of the histone H3.3 variant by the HIRA complex is a prominent nucleosome replacement mechanism affecting gene transcription, especially in postmitotic neurons. However, how HIRA-mediated H3.3 deposition is regulated in these cells remains unclear. Here, we report that dBRWD3, the Drosophila ortholog of the intellectual disability gene BRWD3, regulates gene expression through H3.3, HIRA, and its associated chaperone Yemanuclein ( YEM), the fly ortholog of mammalian Ubinuclein1. In dBRWD3 mutants, increased H3.3 levels disrupt gene expression, dendritic morphogenesis, and sensory organ differentiation. Inactivation of yem or H3.3 remarkably suppresses the global transcriptome changes and various developmental defects caused by dBRWD3 mutations. Our work thus establishes a previously unknown negative regulation of H3.3 and advances our understanding of BRWD3-dependent intellectual disability. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
46. Histone variants H3.3 and H2A.Z/H3.3 facilitate excision of uracil from nucleosome core particles.
- Author
-
Li, Chuxuan, Rioux, Katelyn L., and Delaney, Sarah
- Subjects
- *
URACIL , *HISTONES , *BASE pairs , *DEOXYRIBOZYMES , *DNA repair , *DNA damage - Abstract
At the most fundamental level of chromatin organization, DNA is packaged as nucleosome core particles (NCPs) where DNA is wound around a core of histone proteins. This ubiquitous sequestration of DNA within NCPs presents a significant barrier to many biological processes, including DNA repair. We previously demonstrated that histone variants from the H2A family facilitate excision of uracil (U) lesions by DNA base excision repair (BER) glycosylases. Here, we consider how the histone variant H3.3 and double-variant H2A.Z/H3.3 modulate the BER enzymes uracil DNA glycosylase (UDG) and single-strand selective monofunctional uracil DNA glycosylase (SMUG1). Using an NCP model system with U:G base pairs at a wide variety of geometric positions we generate the global repair profile for both glycosylases. Enhanced excision of U by UDG and SMUG1 is observed with the H3.3 variant. We demonstrate that these H3.3-containing NCPs form two species: (1) octasomes, which contain the full complement of eight histone proteins and (2) hexasomes which are sub-nucleosomal particles that contain six histones. Both the octasome and hexasome species facilitate excision activity of UDG and SMUG1, with the largest impacts observed at sterically-occluded lesion sites and in terminal regions of DNA of the hexasome that do not closely interact with histones. For the double-variant H2A.Z/H3.3 NCPs, which exist as octasomes, the global repair profile reveals that UDG but not SMUG1 has increased U excision activity. The enhanced glycosylase activity reveals potential functions for these histone variants to facilitate BER in packaged DNA and contributes to our understanding of DNA repair in chromatin and its significance regarding mutagenesis and genomic integrity. [Display omitted] • Histone variants modulate BER activity in nucleosome core particles. • Enhanced excision of uracil by UDG and SMUG1 is observed with the H3.3 variant. • Enhanced excision of uracil by UDG is observed with the H2A.Z/H3.3 double variant. • Octasome and hexasome species of H3.3 NCPs facilitate excision by UDG and SMUG1. • Most enhancement at sterically-occluded sites and terminal regions of the hexasome. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
47. High histone variant H3.3 content in mouse prospermatogonia suggests a role in epigenetic reformatting.
- Author
-
Tang, Michelle C. W., Binos, Steve, Ong, Eng K., Wong, Lee H., and Mann, Jeffrey R.
- Published
- 2014
- Full Text
- View/download PDF
48. H3.3 replacement facilitates epigenetic reprogramming of donor nuclei in somatic cell nuclear transfer embryos.
- Author
-
Wen, Duancheng, Banaszynski, Laura A, Rosenwaks, Zev, Allis, C David, and Rafii, Shahin
- Subjects
- *
SOMATIC cell nuclear transfer , *SOMATIC cells , *EPIGENETICS , *EMBRYOS , *HUMAN chromatin , *OVUM , *HISTONES - Abstract
Transfer of a somatic nucleus into an enucleated oocyte is the most efficient approach for somatic cell reprogramming. While this process is known to involve extensive chromatin remodeling of the donor nucleus, the maternal factors responsible and the underlying chromatin-based mechanisms remain largely unknown. Here we discuss our recent findings demonstrating that the histone variant H3.3 plays an essential role in reprogramming and is required for reactivation of key pluripotency genes in somatic cell nuclear transfer (SCNT) embryos. Maternal-derived H3.3 replaces H3 in the donor nucleus shortly after oocyte activation, with the amount of replacement directly related to the differentiation status of the donor nucleus in SCNT embryos. We provide additional evidence to suggest that de novo synthesized H3.3 replaces histone H3 carrying repressive modifications in the donor nuclei of SCNT embryos, and hypothesize that replacement may occur at specific loci that must be reprogrammed for gene reactivation. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
49. Diffuse midline glioma, H3-K27M mutant: Awareness leads to identification
- Author
-
Sujata Chaturvedi, Ishita Pant, Sadhana Tiwari, and Gurbachan Singh
- Subjects
Diffuse intrinsic pontine glioma ,Pathology ,medicine.medical_specialty ,pediatric brain tumor ,histone H3.1 ,histone H3.3 ,lcsh:RC254-282 ,Lesion ,Glioma ,medicine ,Mitosis ,medicine.diagnostic_test ,Glial fibrillary acidic protein ,biology ,business.industry ,midline glioma ,Magnetic resonance imaging ,histone H3 gene sequencing ,Cerebellopontine angle ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,isocitrate dehydrogenase wildtype ,Cytoplasm ,biology.protein ,Brainstem ,medicine.symptom ,business ,H3-K27M-mutant - Abstract
Midline astrocytic neoplasms have distinct molecular characteristics, quite different from astrocytic neoplasms with similar morphology but not located in the midline. It is imperative that neuropathologists should be aware of the existence of these tumors, so they can be correctly diagnosed. Here, we discuss the case of a 14-year-old boy who presented with acute onset of vomiting followed by loss of consciousness. Subsequent magnetic resonance imaging revealed an ill-defined exophytic lesion arising from the brainstem and extending into the left cerebellopontine angle, with areas of hemorrhage and patchy restricted diffusion. The tumor was resected. Microscopy revealed medium-sized tumor cells in diffuse sheets, having round nuclei, granular chromatin, and scant cytoplasm. Microvascular and endothelial cell proliferation in small necrotic areas were seen. Mitosis was 0–1 per high-power field. By routine histopathological analysis, all features were consistent with the diagnosis of glioblastoma. Tumor cells were immunopositive for glial fibrillary acidic protein and isocitrate dehydrogenase-1 mutation (R132H), immunonegative for p53, and retained alpha thalassemia/mental retardation syndrome X-linked. It also showed a strong immunopositivity for H3-K27M mutation. A diagnosis of a diffuse midline glioma with H3-K27M mutation corresponding to the World Health Organization Grade IV was made. This case highlights the importance of exploring signature mutations in well-defined tumor categories such as H3-K27M-mutant diffuse midline glioma.
- Published
- 2019
50. The integrated genomic and epigenomic landscape of brainstem glioma
- Author
-
Chen, LH, Pan, C, Diplas, BH, Xu, C, Hansen, LJ, Wu, Y, Chen, X, Geng, Y, Sun, T, Sun, Y, Zhang, P, Wu, Z, Zhang, J, Li, D, Zhang, Y, Wu, W, Wang, Y, Li, G, Yang, J, Wang, X, Wang, S, Waitkus, MS, He, Y, McLendon, RE, Ashley, D, Yan, H, Zhang, L, Chen, LH, Pan, C, Diplas, BH, Xu, C, Hansen, LJ, Wu, Y, Chen, X, Geng, Y, Sun, T, Sun, Y, Zhang, P, Wu, Z, Zhang, J, Li, D, Zhang, Y, Wu, W, Wang, Y, Li, G, Yang, J, Wang, X, Wang, S, Waitkus, MS, He, Y, McLendon, RE, Ashley, D, Yan, H, and Zhang, L
- Abstract
Brainstem gliomas are a heterogeneous group of tumors that encompass both benign tumors cured with surgical resection and highly lethal cancers with no efficacious therapies. We perform a comprehensive study incorporating epigenetic and genomic analyses on a large cohort of brainstem gliomas, including Diffuse Intrinsic Pontine Gliomas. Here we report, from DNA methylation data, distinct clusters termed H3-Pons, H3-Medulla, IDH, and PA-like, each associated with unique genomic and clinical profiles. The majority of tumors within H3-Pons and-H3-Medulla harbors H3F3A mutations but shows distinct methylation patterns that correlate with anatomical localization within the pons or medulla, respectively. Clinical data show significantly different overall survival between these clusters, and pathway analysis demonstrates different oncogenic mechanisms in these samples. Our findings indicate that the integration of genetic and epigenetic data can facilitate better understanding of brainstem gliomagenesis and classification, and guide future studies for the development of novel treatments for this disease.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.