Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 166(4): 950-962, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27518565

ABSTRACT

Posttranslational modifications (PTMs) of tubulin specify microtubules for specialized cellular functions and comprise what is termed a "tubulin code." PTMs of histones comprise an analogous "histone code," although the "readers, writers, and erasers" of the cytoskeleton and epigenome have heretofore been distinct. We show that methylation is a PTM of dynamic microtubules and that the histone methyltransferase SET-domain-containing 2 (SETD2), which is responsible for H3 lysine 36 trimethylation (H3K36me3) of histones, also methylates α-tubulin at lysine 40, the same lysine that is marked by acetylation on microtubules. Methylation of microtubules occurs during mitosis and cytokinesis and can be ablated by SETD2 deletion, which causes mitotic spindle and cytokinesis defects, micronuclei, and polyploidy. These data now identify SETD2 as a dual-function methyltransferase for both chromatin and the cytoskeleton and show a requirement for methylation in maintenance of genomic stability and the integrity of both the tubulin and histone codes.


Subject(s)
Chromatin Assembly and Disassembly , Cytoskeleton/metabolism , Histone Code , Histone-Lysine N-Methyltransferase/metabolism , Cell Line, Tumor , Cytokinesis , Genomic Instability , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Microtubules/metabolism , Mitosis , Protein Processing, Post-Translational , Tubulin/metabolism
2.
Genes Dev ; 36(7-8): 468-482, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35450884

ABSTRACT

The nucleosome remodeling and deacetylase (NuRD) complex is one of the central chromatin remodeling complexes that mediates gene repression. NuRD is essential for numerous developmental events, including heart development. Clinical and genetic studies have provided direct evidence for the role of chromodomain helicase DNA-binding protein 4 (CHD4), the catalytic component of NuRD, in congenital heart disease (CHD), including atrial and ventricular septal defects. Furthermore, it has been demonstrated that CHD4 is essential for mammalian cardiomyocyte formation and function. A key unresolved question is how CHD4/NuRD is localized to specific cardiac target genes, as neither CHD4 nor NuRD can directly bind DNA. Here, we coupled a bioinformatics-based approach with mass spectrometry analyses to demonstrate that CHD4 interacts with the core cardiac transcription factors GATA4, NKX2-5, and TBX5 during embryonic heart development. Using transcriptomics and genome-wide occupancy data, we characterized the genomic landscape of GATA4, NKX2-5, and TBX5 repression and defined the direct cardiac gene targets of the GATA4-CHD4, NKX2-5-CHD4, and TBX5-CHD4 complexes. These data were used to identify putative cis-regulatory elements controlled by these complexes. We genetically interrogated two of these silencers in vivo: Acta1 and Myh11 We show that deletion of these silencers leads to inappropriate skeletal and smooth muscle gene misexpression, respectively, in the embryonic heart. These results delineate how CHD4/NuRD is localized to specific cardiac loci and explicates how mutations in the broadly expressed CHD4 protein lead to cardiac-specific disease states.


Subject(s)
DNA Helicases , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Animals , DNA Helicases/metabolism , Genes, Homeobox , Mammals/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Myocytes, Cardiac/metabolism , Nucleosomes , Transcription Factors/genetics
3.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38619323

ABSTRACT

Regulation of chromatin states is essential for proper temporal and spatial gene expression. Chromatin states are modulated by remodeling complexes composed of components that have enzymatic activities. CHD4 is the catalytic core of the nucleosome remodeling and deacetylase (NuRD) complex, which represses gene transcription. However, it remains to be determined how CHD4, a ubiquitous enzyme that remodels chromatin structure, functions in cardiomyocytes to maintain heart development. In particular, whether other proteins besides the NuRD components interact with CHD4 in the heart is controversial. Using quantitative proteomics, we identified that CHD4 interacts with SMYD1, a striated muscle-restricted histone methyltransferase that is essential for cardiomyocyte differentiation and cardiac morphogenesis. Comprehensive transcriptomic and chromatin accessibility studies of Smyd1 and Chd4 null embryonic mouse hearts revealed that SMYD1 and CHD4 repress a group of common genes and pathways involved in glycolysis, response to hypoxia, and angiogenesis. Our study reveals a mechanism by which CHD4 functions during heart development, and a previously uncharacterized mechanism regarding how SMYD1 represses cardiac transcription in the developing heart.


Subject(s)
DNA Helicases , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Heart , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Myocytes, Cardiac , Transcription Factors , Animals , Humans , Mice , Cell Differentiation/genetics , Chromatin/metabolism , Glycolysis/genetics , Heart/embryology , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice, Knockout , Muscle Proteins/metabolism , Muscle Proteins/genetics , Myocytes, Cardiac/metabolism , Proteomics , Transcription, Genetic
4.
Mol Cell ; 70(6): 1054-1066.e4, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29932900

ABSTRACT

Spt6 is an essential histone chaperone that mediates nucleosome reassembly during gene transcription. Spt6 also associates with RNA polymerase II (RNAPII) via a tandem Src2 homology domain. However, the significance of Spt6-RNAPII interaction is not well understood. Here, we show that Spt6 recruitment to genes and the nucleosome reassembly functions of Spt6 can still occur in the absence of its association with RNAPII. Surprisingly, we found that Spt6-RNAPII association is required for efficient recruitment of the Ccr4-Not de-adenylation complex to transcribed genes for essential degradation of a range of mRNAs, including mRNAs required for cell-cycle progression. These findings reveal an unexpected control mechanism for mRNA turnover during transcription facilitated by a histone chaperone.


Subject(s)
Histone Chaperones/metabolism , RNA Polymerase II/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcriptional Elongation Factors/metabolism , Histone Chaperones/genetics , Histones/genetics , Histones/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , RNA Polymerase II/genetics , RNA Stability , RNA, Messenger/genetics , Regulatory Elements, Transcriptional , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics
5.
Genome Res ; 31(12): 2327-2339, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34815311

ABSTRACT

Chromatin accessibility states that influence gene expression and other nuclear processes can be altered in disease. The constellation of transcription factors and chromatin regulatory complexes in cells results in characteristic patterns of chromatin accessibility. The study of these patterns in tissues has been limited because existing chromatin accessibility assays are ineffective for archival formalin-fixed, paraffin-embedded (FFPE) tissues. We have developed a method to efficiently extract intact chromatin from archival tissue via enhanced cavitation with a nanodroplet reagent consisting of a lipid shell with a liquid perfluorocarbon core. Inclusion of nanodroplets during the extraction of chromatin from FFPE tissues enhances the recovery of intact accessible and nucleosome-bound chromatin. We show that the addition of nanodroplets to the chromatin accessibility assay formaldehyde-assisted isolation of regulatory elements (FAIRE), does not affect the accessible chromatin signal. Applying the technique to FFPE human tumor xenografts, we identified tumor-relevant regions of accessible chromatin shared with those identified in primary tumors. Further, we deconvoluted non-tumor signal to identify cellular components of the tumor microenvironment. Incorporation of this method of enhanced cavitation into FAIRE offers the potential for extending chromatin accessibility to clinical diagnosis and personalized medicine, while also enabling the exploration of gene regulatory mechanisms in archival samples.

6.
Genome Res ; 30(11): 1605-1617, 2020 11.
Article in English | MEDLINE | ID: mdl-33020206

ABSTRACT

Histone H3 lysine 36 methylation (H3K36me) is a conserved histone modification associated with transcription and DNA repair. Although the effects of H3K36 methylation have been studied, the genome-wide dynamics of H3K36me deposition and removal are not known. We established rapid and reversible optogenetic control for Set2, the sole H3K36 methyltransferase in yeast, by fusing the enzyme with the light-activated nuclear shuttle (LANS) domain. Light activation resulted in efficient Set2-LANS nuclear localization followed by H3K36me3 deposition in vivo, with total H3K36me3 levels correlating with RNA abundance. Although genes showed disparate levels of H3K36 methylation, relative rates of H3K36me3 accumulation were largely linear and consistent across genes, suggesting that H3K36me3 deposition occurs in a directed fashion on all transcribed genes regardless of their overall transcription frequency. Removal of H3K36me3 was highly dependent on the demethylase Rph1. However, the per-gene rate of H3K36me3 loss weakly correlated with RNA abundance and followed exponential decay, suggesting H3K36 demethylases act in a global, stochastic manner. Altogether, these data provide a detailed temporal view of H3K36 methylation and demethylation that suggests transcription-dependent and -independent mechanisms for H3K36me deposition and removal, respectively.


Subject(s)
Histones/metabolism , Methyltransferases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic , Genome, Fungal , Histone Code , Histone Demethylases/metabolism , Histones/chemistry , Lysine/metabolism , Methylation , Models, Statistical , Optogenetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
7.
Proc Natl Acad Sci U S A ; 115(26): 6727-6732, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29891665

ABSTRACT

Cardiac development relies on proper cardiomyocyte differentiation, including expression and assembly of cell-type-specific actomyosin subunits into a functional cardiac sarcomere. Control of this process involves not only promoting expression of cardiac sarcomere subunits but also repressing expression of noncardiac myofibril paralogs. This level of transcriptional control requires broadly expressed multiprotein machines that modify and remodel the chromatin landscape to restrict transcription machinery access. Prominent among these is the nucleosome remodeling and deacetylase (NuRD) complex, which includes the catalytic core subunit CHD4. Here, we demonstrate that direct CHD4-mediated repression of skeletal and smooth muscle myofibril isoforms is required for normal cardiac sarcomere formation, function, and embryonic survival early in gestation. Through transcriptomic and genome-wide analyses of CHD4 localization, we identified unique CHD4 binding sites in smooth muscle myosin heavy chain, fast skeletal α-actin, and the fast skeletal troponin complex genes. We further demonstrate that in the absence of CHD4, cardiomyocytes in the developing heart form a hybrid muscle cell that contains cardiac, skeletal, and smooth muscle myofibril components. These misexpressed paralogs intercalate into the nascent cardiac sarcomere to disrupt sarcomere formation and cause impaired cardiac function in utero. These results demonstrate the genomic and physiological requirements for CHD4 in mammalian cardiac development.


Subject(s)
DNA Helicases/physiology , Gene Expression Regulation, Developmental , Heart Defects, Congenital/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Myocytes, Cardiac/physiology , Sarcomeres/physiology , Animals , DNA Helicases/chemistry , DNA Helicases/deficiency , Female , Gene Knockdown Techniques , Genes, Lethal , Heart/diagnostic imaging , Heart/embryology , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/embryology , Heart Defects, Congenital/pathology , Male , Mice , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Myofibrils/metabolism , Myofibrils/pathology , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Sarcomeres/ultrastructure , Transcription, Genetic , Ultrasonography, Prenatal
8.
Cancer ; 126(22): 4975-4983, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32830315

ABSTRACT

BACKGROUND: Cellular senescence, measured by expression of the cell cycle kinase inhibitor p16INK4a , may contribute to accelerated aging in survivors of childhood, adolescent, and young adult cancer. The authors measured peripheral blood T-lymphocyte p16INK4a expression among pediatric and young adult cancer survivors, hypothesizing that p16INK4a expression is higher after chemotherapy and among frail survivors. METHODS: A cross-sectional cohort of young adult survivors and age-matched, cancer-free controls were assessed for p16INK4a expression and frailty. Newly diagnosed pediatric patients underwent prospective measurements of p16INK4a expression before and after cancer therapy. Frailty was measured with a modified Fried frailty phenotype evaluating sarcopenia, weakness, slowness, energy expenditure, and exhaustion. RESULTS: The cross-sectional cohort enrolled 60 survivors and 29 age-matched controls with a median age of 21 years (range, 17-29 years). The prospective cohort enrolled 9 newly diagnosed patients (age range, 1-18 years). Expression of p16INK4a was higher among survivors compared with controls (9.6 vs 8.9 log2 p16 units; 2-sided P = .005, representing a 25-year age acceleration in survivors) and increased among newly diagnosed patients from matched pretreatment to posttreatment samples (7.3-8.9 log2 p16 units; 2-sided P = .002). Nine survivors (16%) were frail and had higher p16INK4a expression compared with robust survivors (10.5 [frail] vs 9.5 [robust] log2 p16 units; 2-sided P = .055), representing a 35-year age acceleration among frail survivors. CONCLUSIONS: Chemotherapy is associated with increased cellular senescence and molecular age in pediatric and young adult cancer survivors. Frail survivors, compared with robust survivors, exhibit higher levels of p16INK4a , suggesting that cellular senescence may be associated with early aging in survivors.


Subject(s)
Aging/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Frailty/physiopathology , Adolescent , Adult , Cancer Survivors , Cross-Sectional Studies , Humans , Young Adult
9.
J Biol Chem ; 293(35): 13592-13603, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29986887

ABSTRACT

Chromatin remodelers use bromodomains (BDs) to recognize histones. Polybromo 1 (PBRM1 or BAF180) is hypothesized to function as the nucleosome-recognition subunit of the PBAF chromatin-remodeling complex and is frequently mutated in clear cell renal cell carcinoma (ccRCC). Previous studies have applied in vitro methods to explore the binding specificities of the six individual PBRM1 BDs. However, BD targeting to histones and the influence of neighboring BD on nucleosome recognition have not been well characterized. Here, using histone microarrays and intact nucleosomes to investigate the histone-binding characteristics of the six PBRM1 BDs individually and combined, we demonstrate that BD2 and BD4 of PBRM1 mediate binding to acetylated histone peptides and to modified recombinant and cellular nucleosomes. Moreover, we show that neighboring BDs variably modulate these chromatin interactions, with BD1 and BD5 enhancing nucleosome interactions of BD2 and BD4, respectively, whereas BD3 attenuated these interactions. We also found that binding pocket missense mutations in BD4 observed in ccRCC disrupt PBRM1-chromatin interactions and that these mutations in BD4, but not similar mutations in BD2, in the context of full-length PBRM1, accelerate ccRCC cell proliferation. Taken together, our biochemical and mutational analyses have identified BD4 as being critically important for maintaining proper PBRM1 function and demonstrate that BD4 mutations increase ccRCC cell growth. Because of the link between PBRM1 status and sensitivity to immune checkpoint inhibitor treatment, these data also suggest the relevance of BD4 as a potential clinical target.


Subject(s)
Carcinoma, Renal Cell/metabolism , Chromatin/metabolism , Histones/metabolism , Kidney Neoplasms/metabolism , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Carcinoma, Renal Cell/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins , Histones/chemistry , Humans , Kidney Neoplasms/genetics , Models, Molecular , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Binding , Protein Domains , Sequence Alignment , Transcription Factors/chemistry , Transcription Factors/genetics
10.
Proc Natl Acad Sci U S A ; 113(11): 3018-23, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929321

ABSTRACT

Mutations in chromatin-modifying proteins and transcription factors are commonly associated with a wide variety of cancers. Through gain- or loss-of-function, these mutations may result in characteristic alterations of accessible chromatin, indicative of shifts in the landscape of regulatory elements genome-wide. The identification of compounds that reverse a specific chromatin signature could lead to chemical probes or potential therapies. To explore whether chromatin accessibility could serve as a platform for small molecule screening, we adapted formaldehyde-assisted isolation of regulatory elements (FAIRE), a chemical method to enrich for nucleosome-depleted genomic regions, as a high-throughput, automated assay. After demonstrating the validity and robustness of this approach, we applied this method to screen an epigenetically targeted small molecule library by evaluating regions of aberrant nucleosome depletion mediated by EWSR1-FLI1, the chimeric transcription factor critical for the bone and soft tissue tumor Ewing sarcoma. As a class, histone deacetylase inhibitors were greatly overrepresented among active compounds. These compounds resulted in diminished accessibility at targeted sites by disrupting transcription of EWSR1-FLI1. Capitalizing on precise differences in chromatin accessibility for drug discovery efforts offers significant advantages because it does not depend on the a priori selection of a single molecular target and may detect novel biologically relevant pathways.


Subject(s)
Chromatin/drug effects , High-Throughput Screening Assays/methods , Oncogene Proteins, Fusion/antagonists & inhibitors , Transcription, Genetic/drug effects , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Bone Neoplasms/pathology , Cell Line, Tumor , Chromatin/ultrastructure , Drug Design , Drug Evaluation, Preclinical , Histone Deacetylase Inhibitors/isolation & purification , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Molecular Targeted Therapy , Nucleosomes/ultrastructure , Oncogene Proteins, Fusion/genetics , Panobinostat , Phenylbutyrates/pharmacology , Sarcoma, Ewing/pathology , Small Molecule Libraries , Vorinostat
11.
Biochemistry ; 57(19): 2756-2761, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29658277

ABSTRACT

One of the most sensitive, time-consuming, and variable steps of chromatin immunoprecipitation (ChIP) is chromatin sonication. Traditionally, this process can take hours to properly sonicate enough chromatin for multiple ChIP assays. Further, the length of sheared DNA is often inconsistent. In order to faithfully measure chemical and structural changes at the chromatin level, sonication needs to be reliable. Thus, chromatin fragmentation by sonication represents a significant bottleneck to downstream quantitative analysis. To improve the consistency and efficiency of chromatin sonication, we developed and tested a cavitation enhancing reagent based on sonically active nanodroplets. Here, we show that nanodroplets increase sonication efficiency by 16-fold and provide more consistent levels of chromatin fragmentation. Using the previously characterized chromatin in vivo assay (CiA) platform, we generated two distinct chromatin states in order to test nanodroplet-assisted sonication sensitivity in measuring post-translational chromatin marks. By comparing euchromatin to chemically induced heterochromatin at the same CiA:Oct4 locus, we quantitatively measure the capability of our new sonication technique to resolve differences in chromatin structure. We confirm that nanodroplet-assisted sonication results are indistinguishable from those of samples processed with traditional sonication in downstream applications. While the processing time for each sample was reduced from 38.4 to 2.3 min, DNA fragment distribution sizes were significantly more consistent with a coefficient of variation 2.7 times lower for samples sonicated in the presence of nanodroplets. In conclusion, sonication utilizing the nanodroplet cavitation enhancement reagent drastically reduces the amount of processing time and provides consistently fragmented chromatin of high quality for downstream applications.


Subject(s)
Chromatin Immunoprecipitation/methods , DNA Fragmentation/radiation effects , DNA/genetics , Sonication/methods , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Chromatin/radiation effects , DNA/chemistry , DNA/radiation effects , Euchromatin/radiation effects , Heterochromatin/radiation effects , Mice , Nanoparticles/chemistry , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/genetics
12.
J Biol Chem ; 291(40): 21283-21295, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27528607

ABSTRACT

The yeast Set2 histone methyltransferase is a critical enzyme that plays a number of key roles in gene transcription and DNA repair. Recently, the human homologue, SETD2, was found to be recurrently mutated in a significant percentage of renal cell carcinomas, raising the possibility that the activity of SETD2 is tumor-suppressive. Using budding yeast and human cell line model systems, we examined the functional significance of two evolutionarily conserved residues in SETD2 that are recurrently mutated in human cancers. Whereas one of these mutations (R2510H), located in the Set2 Rpb1 interaction domain, did not result in an observable defect in SETD2 enzymatic function, a second mutation in the catalytic domain of this enzyme (R1625C) resulted in a complete loss of histone H3 Lys-36 trimethylation (H3K36me3). This mutant showed unchanged thermal stability as compared with the wild type protein but diminished binding to the histone H3 tail. Surprisingly, mutation of the conserved residue in Set2 (R195C) similarly resulted in a complete loss of H3K36me3 but did not affect dimethylated histone H3 Lys-36 (H3K36me2) or functions associated with H3K36me2 in yeast. Collectively, these data imply a critical role for Arg-1625 in maintaining the protein interaction with H3 and specific H3K36me3 function of this enzyme, which is conserved from yeast to humans. They also may provide a refined biochemical explanation for how H3K36me3 loss leads to genomic instability and cancer.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Methyltransferases/metabolism , Mutation , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Enzyme Stability/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Methylation , Methyltransferases/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Structure-Activity Relationship
13.
Genome Res ; 24(2): 241-50, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24158655

ABSTRACT

Comprehensive sequencing of human cancers has identified recurrent mutations in genes encoding chromatin regulatory proteins. For clear cell renal cell carcinoma (ccRCC), three of the five commonly mutated genes encode the chromatin regulators PBRM1, SETD2, and BAP1. How these mutations alter the chromatin landscape and transcriptional program in ccRCC or other cancers is not understood. Here, we identified alterations in chromatin organization and transcript profiles associated with mutations in chromatin regulators in a large cohort of primary human kidney tumors. By associating variation in chromatin organization with mutations in SETD2, which encodes the enzyme responsible for H3K36 trimethylation, we found that changes in chromatin accessibility occurred primarily within actively transcribed genes. This increase in chromatin accessibility was linked with widespread alterations in RNA processing, including intron retention and aberrant splicing, affecting ∼25% of all expressed genes. Furthermore, decreased nucleosome occupancy proximal to misspliced exons was observed in tumors lacking H3K36me3. These results directly link mutations in SETD2 to chromatin accessibility changes and RNA processing defects in cancer. Detecting the functional consequences of specific mutations in chromatin regulatory proteins in primary human samples could ultimately inform the therapeutic application of an emerging class of chromatin-targeted compounds.


Subject(s)
Carcinoma, Renal Cell/genetics , Chromatin/genetics , Histone-Lysine N-Methyltransferase/genetics , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/pathology , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/metabolism , Humans , Kidney Neoplasms/pathology , Mutation , Nuclear Proteins/genetics , RNA Processing, Post-Transcriptional/genetics , RNA Splicing/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics
14.
J Biol Chem ; 290(43): 26088-102, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26338712

ABSTRACT

G9a and GLP lysine methyltransferases form a heterodimeric complex that is responsible for the majority of histone H3 lysine 9 mono- and di-methylation (H3K9me1/me2). Widely interspaced zinc finger (WIZ) associates with the G9a-GLP protein complex, but its role in mediating lysine methylation is poorly defined. Here, we show that WIZ regulates global H3K9me2 levels by facilitating the interaction of G9a with chromatin. Disrupting the association of G9a-GLP with chromatin by depleting WIZ resulted in altered gene expression and protein-protein interactions that were distinguishable from that of small molecule-based inhibition of G9a/GLP, supporting discrete functions of the G9a-GLP-WIZ chromatin complex in addition to H3K9me2 methylation.


Subject(s)
Chromatin/metabolism , Kruppel-Like Transcription Factors/physiology , Methyltransferases/metabolism , Chromatin Immunoprecipitation , DNA Methylation , Gene Expression/genetics , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Kruppel-Like Transcription Factors/genetics , Protein Binding
15.
J Clin Periodontol ; 43(7): 584-94, 2016 07.
Article in English | MEDLINE | ID: mdl-26990150

ABSTRACT

AIM: Inflammatory periodontal disease is widespread in dogs. This study evaluated site-specific changes in the canine gingival crevicular fluid (GCF) proteome during longitudinal progression from very mild gingivitis to mild periodontitis. Periodontitis diagnosis in dogs requires general anaesthesia with associated risks and costs; our ultimate aim was to develop a periodontitis diagnostic for application in conscious dogs. The objective of this work was to identify potential biomarkers of periodontal disease progression in dogs. MATERIAL AND METHODS: Gingival crevicular fluid was sampled from a total of 10 teeth in eight dogs at three different stages of health/disease and samples prepared for quantitative mass spectrometry (data available via ProteomeXchange; identifier PXD003337). A univariate mixed model analysis determined significantly altered proteins between health states and six were evaluated by ELISA. RESULTS: Four hundred and six proteins were identified with 84 present in all samples. The prevalence of 40 proteins was found to be significantly changed in periodontitis relative to gingivitis. ELISA measurements confirmed that haptoglobin was significantly increased. CONCLUSIONS: This study demonstrates for the first time that proteins detected by mass spectrometry have potential to identify novel biomarkers for canine periodontal disease. Further work is required to validate additional biomarkers for a periodontitis diagnostic.


Subject(s)
Gingivitis , Periodontitis , Animals , Disease Models, Animal , Dogs , Gingival Crevicular Fluid , Periodontal Attachment Loss , Proteome
16.
Nanomedicine ; 12(4): 1053-1062, 2016 May.
Article in English | MEDLINE | ID: mdl-26772430

ABSTRACT

The biological activity of nanoparticle-directed therapies critically depends on cellular targeting. We examined the subtumoral fate of Particle Replication in Non-Wetting Templates (PRINT) nanoparticles in a xenografted melanoma tumor model by multi-color flow cytometry and in vivo confocal tumor imaging. These approaches were compared with the typical method of whole-organ quantification by radiolabeling. In contrast to radioactivity based detection which demonstrated a linear dose-dependent accumulation in the organ, flow cytometry revealed that particle association with cancer cells became dose-independent with increased particle doses and that the majority of the nanoparticles in the tumor were associated with cancer cells despite a low fractional association. In vivo imaging demonstrated an inverse relationship between tumor cell association and other immune cells, likely macrophages. Finally, variation in particle size nonuniformly affected subtumoral association. This study demonstrates the importance of subtumoral targeting when assessing nanoparticle activity within tumors. FROM THE CLINICAL EDITOR: Particle Replication in Non-Wetting Templates (PRINT) technology allows the production of nanoparticles with uniform size. The authors in the study utilized PRINT-produced nanoparticles to investigate specific tumor uptake by multi-color flow cytometry and in vivo confocal tumor imaging. This approach allowed further in-depth correlation between nanoparticle properties and tumor cells and should improve future design.


Subject(s)
Flow Cytometry , Melanoma/diagnostic imaging , Nanomedicine , Nanoparticles/adverse effects , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Macrophages/drug effects , Macrophages/pathology , Melanoma/pathology , Nanoparticles/administration & dosage , Particle Size , Tissue Distribution , Xenograft Model Antitumor Assays
17.
Genome Res ; 22(2): 259-70, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086061

ABSTRACT

Chromosomal translocations involving transcription factor genes have been identified in an increasingly wide range of cancers. Some translocations can create a protein "chimera" that is composed of parts from different proteins. How such chimeras cause cancer, and why they cause cancer in some cell types but not others, is not understood. One such chimera is EWS-FLI, the most frequently occurring translocation in Ewing Sarcoma, a malignant bone and soft tissue tumor of children and young adults. Using EWS-FLI and its parental transcription factor, FLI1, we created a unique experimental system to address questions regarding the genomic mechanisms by which chimeric transcription factors cause cancer. We found that in tumor cells, EWS-FLI targets regions of the genome distinct from FLI1, despite identical DNA-binding domains. In primary endothelial cells, however, EWS-FLI and FLI1 demonstrate similar targeting. To understand this mistargeting, we examined chromatin organization. Regions targeted by EWS-FLI are normally repressed and nucleosomal in primary endothelial cells. In tumor cells, however, bound regions are nucleosome depleted and harbor the chromatin signature of enhancers. We next demonstrated that through chimerism, EWS-FLI acquired the ability to alter chromatin. Expression of EWS-FLI results in nucleosome depletion at targeted sites, whereas silencing of EWS-FLI in tumor cells restored nucleosome occupancy. Thus, the EWS-FLI chimera acquired chromatin-altering activity, leading to mistargeting, chromatin disruption, and ultimately, transcriptional dysregulation.


Subject(s)
Chromatin/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Transcription, Genetic , Binding Sites/genetics , Cell Line, Tumor , Chimerism , Chromatin/metabolism , Endothelial Cells/metabolism , Enhancer Elements, Genetic , Gene Silencing , Humans , Microfilament Proteins/metabolism , Microsatellite Repeats , Neoplasms/genetics , Neoplasms/metabolism , Nucleotide Motifs , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Trans-Activators , Translocation, Genetic
18.
J Virol ; 87(21): 11831-42, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23986576

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic gammaherpesvirus which establishes latent infection in endothelial and B cells, as well as in primary effusion lymphoma (PEL). During latency, the viral genome exists as a circular DNA minichromosome (episome) and is packaged into chromatin analogous to human chromosomes. Only a small subset of promoters, those which drive latent RNAs, are active in latent episomes. In general, nucleosome depletion ("open chromatin") is a hallmark of eukaryotic regulatory elements such as promoters and transcriptional enhancers or insulators. We applied formaldehyde-assisted isolation of regulatory elements (FAIRE) followed by next-generation sequencing to identify regulatory elements in the KSHV genome and integrated these data with previously identified locations of histone modifications, RNA polymerase II occupancy, and CTCF binding sites. We found that (i) regions of open chromatin were not restricted to the transcriptionally defined latent loci; (ii) open chromatin was adjacent to regions harboring activating histone modifications, even at transcriptionally inactive loci; and (iii) CTCF binding sites fell within regions of open chromatin with few exceptions, including the constitutive LANA promoter and the vIL6 promoter. FAIRE-identified nucleosome depletion was similar among B and endothelial cell lineages, suggesting a common viral genome architecture in all forms of latency.


Subject(s)
Chromatin/metabolism , DNA, Viral/metabolism , Herpesvirus 8, Human/physiology , Virus Latency , B-Lymphocytes/virology , Cell Line , Chromatin Immunoprecipitation , Endothelial Cells/virology , Gene Expression Profiling , Herpesvirus 8, Human/genetics , Histones/metabolism , Humans , Microarray Analysis , Protein Binding , Regulatory Elements, Transcriptional , Transcription, Genetic
19.
BMC Microbiol ; 14: 196, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25085267

ABSTRACT

BACKGROUND: The current inability to culture the entirety of observed bacteria is well known and with the advent of ever more powerful molecular tools, that can survey bacterial communities at previously unattainable depth, the gap in our capacity to culture and define all of these species increases exponentially. This gap has essentially become the rate limiting step in determining how the knowledge of which species are present in a sample can be applied to understand the role of these species in an ecosystem or disease process. A case in point is periodontal disease, which is the most widespread oral disease in dogs. If untreated the disease results in significant pain, eventual loss of the dentition and potentially an increased risk of systemic diseases. Previous molecular based studies have identified the bacterial species associated with periodontal disease in dogs; however without cultured strains from many of these species it has not been possible to study whether they play a role in the disease process. RESULTS: Using a quantitative polymerase chain reaction (qPCR) directed approach a range of microbiological media were screened and optimized to enrich for previously uncultivated target species. A systematic screening methodology was then employed to isolate the species of interest. In cases where the target species were not cultivable in isolation, helper strains grown underneath a nitrocellulose membrane were used to provide the necessary growth factors. This guided media optimization approach enabled the purification of 14 species, 8 of which we had previously been unable to cultivate in isolation. It is also applicable to the targeted isolation of isolates from species that have previously been cultured (for example to study intra-species variation) as demonstrated by the successful isolation of 6 targeted isolates of already cultured species. CONCLUSIONS: To our knowledge this is the first time this combination of qPCR guided media optimization, strategic screening and helper strain support has been used successfully to isolate previously uncultured bacteria. This approach can be applied to any uncultured bacterial species where knowledge of their nutritional requirements or low relative abundance impedes their isolation.


Subject(s)
Bacteria/classification , Bacteria/isolation & purification , Dental Plaque/veterinary , Dog Diseases/microbiology , Periodontal Diseases/veterinary , Animals , Bacteria/growth & development , Bacteriological Techniques/methods , Culture Media/chemistry , Dental Plaque/microbiology , Dogs , Periodontal Diseases/microbiology , Real-Time Polymerase Chain Reaction
20.
Cancer Cell ; 9(6): 473-84, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16766266

ABSTRACT

Clear cell sarcoma (CCS) harbors a pathognomonic chromosomal translocation fusing the Ewing's sarcoma gene (EWS) to the CREB family transcription factor ATF1 and exhibits melanocytic features. We show that EWS-ATF1 occupies the MITF promoter, mimicking melanocyte-stimulating hormone (MSH) signaling to induce expression of MITF, the melanocytic master transcription factor and an amplified oncogene in melanoma. Knockdown/rescue studies revealed that MITF mediates the requirement of EWS-ATF1 for CCS survival in vitro and in vivo as well as for melanocytic differentiation. Moreover, MITF and TFE3 reciprocally rescue one another in lines derived from CCS or pediatric renal carcinoma. Seemingly unrelated tumors thus employ distinct strategies to oncogenically dysregulate the MiT family, collectively broadening the definition of MiT-associated human cancers.


Subject(s)
Activating Transcription Factor 1/metabolism , DNA-Binding Proteins/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/physiology , RNA-Binding Protein EWS/genetics , Sarcoma, Clear Cell/metabolism , Activating Transcription Factor 1/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , High Mobility Group Proteins/biosynthesis , Humans , Melanocyte-Stimulating Hormones/physiology , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Microphthalmia-Associated Transcription Factor/genetics , Neoplasm Transplantation , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Promoter Regions, Genetic , Regulatory Factor X Transcription Factors , SOXE Transcription Factors , Sarcoma, Clear Cell/pathology , Signal Transduction , Transcription Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL