19 results on '"Garcia BA"'
Search Results
2. Histone H3.3 lysine 9 and 27 control repressive chromatin at cryptic enhancers and bivalent promoters.
- Author
-
Trovato M, Bunina D, Yildiz U, Fernandez-Novel Marx N, Uckelmann M, Levina V, Perez Y, Janeva A, Garcia BA, Davidovich C, Zaugg JB, and Noh KM
- Subjects
- Animals, Mice, Mutation, Histone Code, Transcription, Genetic, Endogenous Retroviruses genetics, Endogenous Retroviruses metabolism, Histones metabolism, Histones genetics, Promoter Regions, Genetic genetics, Chromatin metabolism, Mouse Embryonic Stem Cells metabolism, Enhancer Elements, Genetic genetics, Lysine metabolism
- Abstract
Histone modifications are associated with distinct transcriptional states, but it is unclear whether they instruct gene expression. To investigate this, we mutate histone H3.3 K9 and K27 residues in mouse embryonic stem cells (mESCs). Here, we find that H3.3K9 is essential for controlling specific distal intergenic regions and for proper H3K27me3 deposition at promoters. The H3.3K9A mutation resulted in decreased H3K9me3 at regions encompassing endogenous retroviruses and induced a gain of H3K27ac and nascent transcription. These changes in the chromatin environment unleash cryptic enhancers, resulting in the activation of distinctive transcriptional programs and culminating in protein expression normally restricted to specialized immune cell types. The H3.3K27A mutant disrupts the deposition and spreading of the repressive H3K27me3 mark, particularly impacting bivalent genes with higher basal levels of H3.3 at promoters. Therefore, H3.3K9 and K27 crucially orchestrate repressive chromatin states at cis-regulatory elements and bivalent promoters, respectively, and instruct proper transcription in mESCs., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
3. PHF6 cooperates with SWI/SNF complexes to facilitate transcriptional progression.
- Author
-
Mittal P, Myers JA, Carter RD, Radko-Juettner S, Malone HA, Rosikiewicz W, Robertson AN, Zhu Z, Narayanan IV, Hansen BS, Parrish M, Bhanu NV, Mobley RJ, Rehg JE, Xu B, Drosos Y, Pruett-Miller SM, Ljungman M, Garcia BA, Wu G, Partridge JF, and Roberts CWM
- Subjects
- Animals, Humans, Male, Mice, Abnormalities, Multiple, Cell Line, Tumor, Chromatin metabolism, Chromatin Assembly and Disassembly, Chromosomal Proteins, Non-Histone metabolism, Chromosomal Proteins, Non-Histone genetics, CRISPR-Cas Systems, Face abnormalities, Foot Deformities, Congenital genetics, Foot Deformities, Congenital metabolism, Hand Deformities, Congenital, Intellectual Disability genetics, Intellectual Disability metabolism, Mutation, Neck abnormalities, Transcription, Genetic, Micrognathism genetics, Micrognathism metabolism, Promoter Regions, Genetic genetics, Repressor Proteins metabolism, Repressor Proteins genetics, Rhabdoid Tumor genetics, Rhabdoid Tumor metabolism, Rhabdoid Tumor pathology, SMARCB1 Protein metabolism, SMARCB1 Protein genetics, Transcription Factors metabolism, Transcription Factors genetics
- Abstract
Genes encoding subunits of SWI/SNF (BAF) chromatin remodeling complexes are mutated in nearly 25% of cancers. To gain insight into the mechanisms by which SWI/SNF mutations drive cancer, we contributed ten rhabdoid tumor (RT) cell lines mutant for SWI/SNF subunit SMARCB1 to a genome-scale CRISPR-Cas9 depletion screen performed across 896 cell lines. We identify PHF6 as specifically essential for RT cell survival and demonstrate that dependency on Phf6 extends to Smarcb1-deficient cancers in vivo. As mutations in either SWI/SNF or PHF6 can cause the neurodevelopmental disorder Coffin-Siris syndrome, our findings of a dependency suggest a previously unrecognized functional link. We demonstrate that PHF6 co-localizes with SWI/SNF complexes at promoters, where it is essential for maintenance of an active chromatin state. We show that in the absence of SMARCB1, PHF6 loss disrupts the recruitment and stability of residual SWI/SNF complex members, collectively resulting in the loss of active chromatin at promoters and stalling of RNA Polymerase II progression. Our work establishes a mechanistic basis for the shared syndromic features of SWI/SNF and PHF6 mutations in CSS and the basis for selective dependency on PHF6 in SMARCB1-mutant cancers., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
4. Cancer-associated Histone H3 N-terminal arginine mutations disrupt PRC2 activity and impair differentiation.
- Author
-
Nacev BA, Dabas Y, Paul MR, Pacheco C, Mitchener M, Perez Y, Fang Y, Soshnev AA, Barrows D, Carroll T, Socci ND, St Jean SC, Tiwari S, Gruss MJ, Monette S, Tap WD, Garcia BA, Muir T, and Allis CD
- Subjects
- Animals, Humans, Mice, Chromatin metabolism, Epigenesis, Genetic, Mesenchymal Stem Cells metabolism, Cell Line, Tumor, Histones metabolism, Histones genetics, Cell Differentiation genetics, Arginine metabolism, Polycomb Repressive Complex 2 metabolism, Polycomb Repressive Complex 2 genetics, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Mutation
- Abstract
Dysregulated epigenetic states are a hallmark of cancer and often arise from genetic alterations in epigenetic regulators. This includes missense mutations in histones, which, together with associated DNA, form nucleosome core particles. However, the oncogenic mechanisms of most histone mutations are unknown. Here, we demonstrate that cancer-associated histone mutations at arginines in the histone H3 N-terminal tail disrupt repressive chromatin domains, alter gene regulation, and dysregulate differentiation. We find that histone H3R2C and R26C mutants reduce transcriptionally repressive H3K27me3. While H3K27me3 depletion in cells expressing these mutants is exclusively observed on the minor fraction of histone tails harboring the mutations, the same mutants recurrently disrupt broad H3K27me3 domains in the chromatin context, including near developmentally regulated promoters. H3K27me3 loss leads to de-repression of differentiation pathways, with concordant effects between H3R2 and H3R26 mutants despite different proximity to the PRC2 substrate, H3K27. Functionally, H3R26C-expressing mesenchymal progenitor cells and murine embryonic stem cell-derived teratomas demonstrate impaired differentiation. Collectively, these data show that cancer-associated H3 N-terminal arginine mutations reduce PRC2 activity and disrupt chromatin-dependent developmental functions, a cancer-relevant phenotype., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. Two DOT1 enzymes cooperatively mediate efficient ubiquitin-independent histone H3 lysine 76 tri-methylation in kinetoplastids.
- Author
-
Frisbie VS, Hashimoto H, Xie Y, De Luna Vitorino FN, Baeza J, Nguyen T, Yuan Z, Kiselar J, Garcia BA, and Debler EW
- Subjects
- Lysine metabolism, Histone-Lysine N-Methyltransferase metabolism, Methylation, Histones metabolism, Ubiquitin
- Abstract
In higher eukaryotes, a single DOT1 histone H3 lysine 79 (H3K79) methyltransferase processively produces H3K79me2/me3 through histone H2B mono-ubiquitin interaction, while the kinetoplastid Trypanosoma brucei di-methyltransferase DOT1A and tri-methyltransferase DOT1B efficiently methylate the homologous H3K76 without H2B mono-ubiquitination. Based on structural and biochemical analyses of DOT1A, we identify key residues in the methyltransferase motifs VI and X for efficient ubiquitin-independent H3K76 methylation in kinetoplastids. Substitution of a basic to an acidic residue within motif VI (Gx
6 K) is essential to stabilize the DOT1A enzyme-substrate complex, while substitution of the motif X sequence VYGE by CAKS renders a rigid active-site loop flexible, implying a distinct mechanism of substrate recognition. We further reveal distinct methylation kinetics and substrate preferences of DOT1A (H3K76me0) and DOT1B (DOT1A products H3K76me1/me2) in vitro, determined by a Ser and Ala residue within motif IV, respectively, enabling DOT1A and DOT1B to mediate efficient H3K76 tri-methylation non-processively but cooperatively, and suggesting why kinetoplastids have evolved two DOT1 enzymes., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
6. Chromatin profiling in human neurons reveals aberrant roles for histone acetylation and BET family proteins in schizophrenia.
- Author
-
Farrelly LA, Zheng S, Schrode N, Topol A, Bhanu NV, Bastle RM, Ramakrishnan A, Chan JC, Cetin B, Flaherty E, Shen L, Gleason K, Tamminga CA, Garcia BA, Li H, Brennand KJ, and Maze I
- Subjects
- Acetylation, Cell Cycle Proteins metabolism, Chromatin, Histones metabolism, Humans, Nerve Tissue Proteins metabolism, Neurons metabolism, Nuclear Proteins metabolism, Protein Processing, Post-Translational, Receptors, Cell Surface metabolism, Transcription Factors metabolism, Induced Pluripotent Stem Cells metabolism, Schizophrenia genetics
- Abstract
Schizophrenia (SZ) is a psychiatric disorder with complex genetic risk dictated by interactions between hundreds of risk variants. Epigenetic factors, such as histone posttranslational modifications (PTMs), have been shown to play critical roles in many neurodevelopmental processes, and when perturbed may also contribute to the precipitation of disease. Here, we apply an unbiased proteomics approach to evaluate combinatorial histone PTMs in human induced pluripotent stem cell (hiPSC)-derived forebrain neurons from individuals with SZ. We observe hyperacetylation of H2A.Z and H4 in neurons derived from SZ cases, results that were confirmed in postmortem human brain. We demonstrate that the bromodomain and extraterminal (BET) protein, BRD4, is a bona fide 'reader' of H2A.Z acetylation, and further provide evidence that BET family protein inhibition ameliorates transcriptional abnormalities in patient-derived neurons. Thus, treatments aimed at alleviating BET protein interactions with hyperacetylated histones may aid in the prevention or treatment of SZ., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
7. Biochemical and functional characterization of mutant KRAS epitopes validates this oncoprotein for immunological targeting.
- Author
-
Bear AS, Blanchard T, Cesare J, Ford MJ, Richman LP, Xu C, Baroja ML, McCuaig S, Costeas C, Gabunia K, Scholler J, Posey AD Jr, O'Hara MH, Smole A, Powell DJ Jr, Garcia BA, Vonderheide RH, Linette GP, and Carreno BM
- Subjects
- Adoptive Transfer, Alleles, Animals, Carcinogenesis genetics, Cell Line, Tumor, Histocompatibility Antigens Class I immunology, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mice, Mutation, Peptides genetics, Peptides immunology, Proteomics, Proto-Oncogene Proteins p21(ras) metabolism, Receptors, Antigen, T-Cell, alpha-beta genetics, Xenograft Model Antitumor Assays, CD8-Positive T-Lymphocytes immunology, Carcinogenesis immunology, Epitopes, T-Lymphocyte immunology, Lung Neoplasms immunology, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) immunology, Receptors, Antigen, T-Cell, alpha-beta immunology
- Abstract
Activating RAS missense mutations are among the most prevalent genomic alterations observed in human cancers and drive oncogenesis in the three most lethal tumor types. Emerging evidence suggests mutant KRAS (mKRAS) may be targeted immunologically, but mKRAS epitopes remain poorly defined. Here we employ a multi-omics approach to characterize HLA class I-restricted mKRAS epitopes. We provide proteomic evidence of mKRAS epitope processing and presentation by high prevalence HLA class I alleles. Select epitopes are immunogenic enabling mKRAS-specific TCRαβ isolation. TCR transfer to primary CD8
+ T cells confers cytotoxicity against mKRAS tumor cell lines independent of histologic origin, and the kinetics of lytic activity correlates with mKRAS peptide-HLA class I complex abundance. Adoptive transfer of mKRAS-TCR engineered CD8+ T cells leads to tumor eradication in a xenograft model of metastatic lung cancer. This study validates mKRAS peptides as bona fide epitopes facilitating the development of immune therapies targeting this oncoprotein., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
8. Structural insight on assembly-line catalysis in terpene biosynthesis.
- Author
-
Faylo JL, van Eeuwen T, Kim HJ, Gorbea Colón JJ, Garcia BA, Murakami K, and Christianson DW
- Subjects
- Alkyl and Aryl Transferases metabolism, Ascomycota chemistry, Ascomycota enzymology, Catalysis, Catalytic Domain, Cryoelectron Microscopy, Cyclization, Dimethylallyltranstransferase chemistry, Dimethylallyltranstransferase metabolism, Fungal Proteins metabolism, Glycosides biosynthesis, Lyases chemistry, Lyases metabolism, Multifunctional Enzymes, Polyisoprenyl Phosphates metabolism, Protein Conformation, Alkyl and Aryl Transferases chemistry, Diterpenes metabolism, Fungal Proteins chemistry
- Abstract
Fusicoccadiene synthase from Phomopsis amygdali (PaFS) is a unique bifunctional terpenoid synthase that catalyzes the first two steps in the biosynthesis of the diterpene glycoside Fusicoccin A, a mediator of 14-3-3 protein interactions. The prenyltransferase domain of PaFS generates geranylgeranyl diphosphate, which the cyclase domain then utilizes to generate fusicoccadiene, the tricyclic hydrocarbon skeleton of Fusicoccin A. Here, we use cryo-electron microscopy to show that the structure of full-length PaFS consists of a central octameric core of prenyltransferase domains, with the eight cyclase domains radiating outward via flexible linker segments in variable splayed-out positions. Cryo-electron microscopy and chemical crosslinking experiments additionally show that compact conformations can be achieved in which cyclase domains are more closely associated with the prenyltransferase core. This structural analysis provides a framework for understanding substrate channeling, since most of the geranylgeranyl diphosphate generated by the prenyltransferase domains remains on the enzyme for cyclization to form fusicoccadiene.
- Published
- 2021
- Full Text
- View/download PDF
9. Cryo-EM structure of TFIIH/Rad4-Rad23-Rad33 in damaged DNA opening in nucleotide excision repair.
- Author
-
van Eeuwen T, Shim Y, Kim HJ, Zhao T, Basu S, Garcia BA, Kaplan CD, Min JH, and Murakami K
- Subjects
- DNA Adducts metabolism, DNA Helicases chemistry, DNA Helicases genetics, DNA-Binding Proteins genetics, Saccharomyces cerevisiae Proteins genetics, Transcription Factor TFIIH genetics, Cryoelectron Microscopy, DNA chemistry, DNA Damage, DNA Repair, DNA-Binding Proteins chemistry, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins chemistry, Transcription Factor TFIIH chemistry
- Abstract
The versatile nucleotide excision repair (NER) pathway initiates as the XPC-RAD23B-CETN2 complex first recognizes DNA lesions from the genomic DNA and recruits the general transcription factor complex, TFIIH, for subsequent lesion verification. Here, we present a cryo-EM structure of an NER initiation complex containing Rad4-Rad23-Rad33 (yeast homologue of XPC-RAD23B-CETN2) and 7-subunit coreTFIIH assembled on a carcinogen-DNA adduct lesion at 3.9-9.2 Å resolution. A ~30-bp DNA duplex could be mapped as it straddles between Rad4 and the Ssl2 (XPB) subunit of TFIIH on the 3' and 5' side of the lesion, respectively. The simultaneous binding with Rad4 and TFIIH was permitted by an unwinding of DNA at the lesion. Translocation coupled with torque generation by Ssl2 and Rad4 would extend the DNA unwinding at the lesion and deliver the damaged strand to Rad3 (XPD) in an open form suitable for subsequent lesion scanning and verification.
- Published
- 2021
- Full Text
- View/download PDF
10. Cryo-EM structures of engineered active bc 1 -cbb 3 type CIII 2 CIV super-complexes and electronic communication between the complexes.
- Author
-
Steimle S, van Eeuwen T, Ozturk Y, Kim HJ, Braitbard M, Selamoglu N, Garcia BA, Schneidman-Duhovny D, Murakami K, and Daldal F
- Subjects
- Bacterial Proteins genetics, Catalytic Domain, Coenzymes chemistry, Coenzymes metabolism, Cryoelectron Microscopy, Electron Transport, Electron Transport Complex III genetics, Electron Transport Complex IV genetics, Genetic Engineering, Rhodobacter capsulatus chemistry, Rhodobacter capsulatus genetics, Rhodobacter capsulatus metabolism, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Electron Transport Complex III chemistry, Electron Transport Complex III metabolism, Electron Transport Complex IV chemistry, Electron Transport Complex IV metabolism, Rhodobacter capsulatus enzymology
- Abstract
Respiratory electron transport complexes are organized as individual entities or combined as large supercomplexes (SC). Gram-negative bacteria deploy a mitochondrial-like cytochrome (cyt) bc
1 (Complex III, CIII2 ), and may have specific cbb3 -type cyt c oxidases (Complex IV, CIV) instead of the canonical aa3 -type CIV. Electron transfer between these complexes is mediated by soluble (c2 ) and membrane-anchored (cy ) cyts. Here, we report the structure of an engineered bc1 -cbb3 type SC (CIII2 CIV, 5.2 Å resolution) and three conformers of native CIII2 (3.3 Å resolution). The SC is active in vivo and in vitro, contains all catalytic subunits and cofactors, and two extra transmembrane helices attributed to cyt cy and the assembly factor CcoH. The cyt cy is integral to SC, its cyt domain is mobile and it conveys electrons to CIV differently than cyt c2 . The successful production of a native-like functional SC and determination of its structure illustrate the characteristics of membrane-confined and membrane-external respiratory electron transport pathways in Gram-negative bacteria.- Published
- 2021
- Full Text
- View/download PDF
11. Disruption of ATRX-RNA interactions uncovers roles in ATRX localization and PRC2 function.
- Author
-
Ren W, Medeiros N, Warneford-Thomson R, Wulfridge P, Yan Q, Bian J, Sidoli S, Garcia BA, Skordalakes E, Joyce E, Bonasio R, and Sarma K
- Subjects
- Animals, Chromatin Assembly and Disassembly genetics, Female, Fibroblasts enzymology, Fibroblasts metabolism, Heterochromatin metabolism, Histones chemistry, Histones metabolism, Methylation, Mice, Protein Binding, Protein Domains genetics, X-linked Nuclear Protein metabolism, Chromatin metabolism, Polycomb Repressive Complex 2 metabolism, RNA metabolism, X-linked Nuclear Protein genetics
- Abstract
Heterochromatin in the eukaryotic genome is rigorously controlled by the concerted action of protein factors and RNAs. Here, we investigate the RNA binding function of ATRX, a chromatin remodeler with roles in silencing of repetitive regions of the genome and in recruitment of the polycomb repressive complex 2 (PRC2). We identify ATRX RNA binding regions (RBRs) and discover that the major ATRX RBR lies within the N-terminal region of the protein, distinct from its PHD and helicase domains. Deletion of this ATRX RBR (ATRXΔRBR) compromises ATRX interactions with RNAs in vitro and in vivo and alters its chromatin binding properties. Genome-wide studies reveal that loss of RNA interactions results in a redistribution of ATRX on chromatin. Finally, our studies identify a role for ATRX-RNA interactions in regulating PRC2 localization to a subset of polycomb target genes.
- Published
- 2020
- Full Text
- View/download PDF
12. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment.
- Author
-
Chan JC, Morgan CP, Adrian Leu N, Shetty A, Cisse YM, Nugent BM, Morrison KE, Jašarević E, Huang W, Kanyuch N, Rodgers AB, Bhanu NV, Berger DS, Garcia BA, Ament S, Kane M, Neill Epperson C, and Bale TL
- Subjects
- Adolescent, Animals, Cell Culture Techniques, Epididymis metabolism, Epigenesis, Genetic, Epigenomics, Female, Germ Cells, Histones, Humans, Male, Mice, Mice, Inbred C57BL, MicroRNAs metabolism, Nanoparticles, Sperm Maturation genetics, Sperm Maturation physiology, Spermatogenesis genetics, Spermatogenesis physiology, Spermatozoa metabolism, Stress, Physiological, Testis, Extracellular Vesicles metabolism, Nervous System growth & development, Proteomics, Reproduction physiology
- Abstract
Extracellular vesicles (EVs) are a unique mode of intercellular communication capable of incredible specificity in transmitting signals involved in cellular function, including germ cell maturation. Spermatogenesis occurs in the testes, behind a protective barrier to ensure safeguarding of germline DNA from environmental insults. Following DNA compaction, further sperm maturation occurs in the epididymis. Here, we report reproductive tract EVs transmit information regarding stress in the paternal environment to sperm, potentially altering fetal development. Using intracytoplasmic sperm injection, we found that sperm incubated with EVs collected from stress-treated epididymal epithelial cells produced offspring with altered neurodevelopment and adult stress reactivity. Proteomic and transcriptomic assessment of these EVs showed dramatic changes in protein and miRNA content long after stress treatment had ended, supporting a lasting programmatic change in response to chronic stress. Thus, EVs as a normal process in sperm maturation, can also perform roles in intergenerational transmission of paternal environmental experience.
- Published
- 2020
- Full Text
- View/download PDF
13. A computational platform for high-throughput analysis of RNA sequences and modifications by mass spectrometry.
- Author
-
Wein S, Andrews B, Sachsenberg T, Santos-Rosa H, Kohlbacher O, Kouzarides T, Garcia BA, and Weisser H
- Subjects
- Base Sequence genetics, Databases, Factual statistics & numerical data, Datasets as Topic, Humans, Oligonucleotides chemistry, Oligonucleotides genetics, Oligonucleotides metabolism, RNA, Transfer chemistry, RNA, Transfer genetics, RNA, Transfer metabolism, Reproducibility of Results, Epigenomics methods, High-Throughput Screening Assays methods, RNA Processing, Post-Transcriptional genetics, Search Engine, Tandem Mass Spectrometry methods
- Abstract
The field of epitranscriptomics continues to reveal how post-transcriptional modification of RNA affects a wide variety of biological phenomena. A pivotal challenge in this area is the identification of modified RNA residues within their sequence contexts. Mass spectrometry (MS) offers a comprehensive solution by using analogous approaches to shotgun proteomics. However, software support for the analysis of RNA MS data is inadequate at present and does not allow high-throughput processing. Existing software solutions lack the raw performance and statistical grounding to efficiently handle the numerous modifications found on RNA. We present a free and open-source database search engine for RNA MS data, called NucleicAcidSearchEngine (NASE), that addresses these shortcomings. We demonstrate the capability of NASE to reliably identify a wide range of modified RNA sequences in four original datasets of varying complexity. In human tRNA, we characterize over 20 different modification types simultaneously and find many cases of incomplete modification.
- Published
- 2020
- Full Text
- View/download PDF
14. Quantitative live cell imaging reveals influenza virus manipulation of Rab11A transport through reduced dynein association.
- Author
-
Bhagwat AR, Le Sage V, Nturibi E, Kulej K, Jones J, Guo M, Tae Kim E, Garcia BA, Weitzman MD, Shroff H, and Lakdawala SS
- Subjects
- Biological Transport, Dyneins genetics, Host-Pathogen Interactions, Humans, Influenza A virus genetics, Influenza, Human genetics, Influenza, Human virology, RNA, Viral genetics, RNA, Viral metabolism, rab GTP-Binding Proteins genetics, Dyneins metabolism, Influenza A virus physiology, Influenza, Human metabolism, rab GTP-Binding Proteins metabolism
- Abstract
Assembly of infectious influenza A viruses (IAV) is a complex process involving transport from the nucleus to the plasma membrane. Rab11A-containing recycling endosomes have been identified as a platform for intracellular transport of viral RNA (vRNA). Here, using high spatiotemporal resolution light-sheet microscopy (~1.4 volumes/second, 330 nm isotropic resolution), we quantify Rab11A and vRNA movement in live cells during IAV infection and report that IAV infection decreases speed and increases arrest of Rab11A. Unexpectedly, infection with respiratory syncytial virus alters Rab11A motion in a manner opposite to IAV, suggesting that Rab11A is a common host component that is differentially manipulated by respiratory RNA viruses. Using two-color imaging we demonstrate co-transport of Rab11A and IAV vRNA in infected cells and provide direct evidence that vRNA-associated Rab11A have altered transport. The mechanism of altered Rab11A movement is likely related to a decrease in dynein motors bound to Rab11A vesicles during IAV infection.
- Published
- 2020
- Full Text
- View/download PDF
15. Histone H3K23-specific acetylation by MORF is coupled to H3K14 acylation.
- Author
-
Klein BJ, Jang SM, Lachance C, Mi W, Lyu J, Sakuraba S, Krajewski K, Wang WW, Sidoli S, Liu J, Zhang Y, Wang X, Warfield BM, Kueh AJ, Voss AK, Thomas T, Garcia BA, Liu WR, Strahl BD, Kono H, Li W, Shi X, Côté J, and Kutateladze TG
- Subjects
- Acetylation, Acylation, Binding Sites genetics, Cell Line, Tumor, Crystallography, X-Ray, HEK293 Cells, Histone Acetyltransferases chemistry, Histone Acetyltransferases genetics, Histones chemistry, Humans, K562 Cells, Molecular Dynamics Simulation, Protein Binding, Protein Domains, Histone Acetyltransferases metabolism, Histones metabolism, Lysine metabolism, Protein Processing, Post-Translational
- Abstract
Acetylation of histone H3K23 has emerged as an essential posttranslational modification associated with cancer and learning and memory impairment, yet our understanding of this epigenetic mark remains insufficient. Here, we identify the native MORF complex as a histone H3K23-specific acetyltransferase and elucidate its mechanism of action. The acetyltransferase function of the catalytic MORF subunit is positively regulated by the DPF domain of MORF (MORF
DPF ). The crystal structure of MORFDPF in complex with crotonylated H3K14 peptide provides mechanistic insight into selectivity of this epigenetic reader and its ability to recognize both histone and DNA. ChIP data reveal the role of MORFDPF in MORF-dependent H3K23 acetylation of target genes. Mass spectrometry, biochemical and genomic analyses show co-existence of the H3K23ac and H3K14ac modifications in vitro and co-occupancy of the MORF complex, H3K23ac, and H3K14ac at specific loci in vivo. Our findings suggest a model in which interaction of MORFDPF with acylated H3K14 promotes acetylation of H3K23 by the native MORF complex to activate transcription.- Published
- 2019
- Full Text
- View/download PDF
16. PFA ependymoma-associated protein EZHIP inhibits PRC2 activity through a H3 K27M-like mechanism.
- Author
-
Jain SU, Do TJ, Lund PJ, Rashoff AQ, Diehl KL, Cieslik M, Bajic A, Juretic N, Deshmukh S, Venneti S, Muir TW, Garcia BA, Jabado N, and Lewis PW
- Subjects
- Animals, Brain Neoplasms pathology, Carcinogenesis genetics, Cell Line, Tumor, Chromatin metabolism, CpG Islands, Cranial Fossa, Posterior, Datasets as Topic, Embryo, Mammalian, Ependymoma pathology, Fibroblasts, Gene Expression Regulation, Neoplastic, Gene Silencing, Glioma pathology, HEK293 Cells, Histones, Humans, Mice, Oncogene Proteins genetics, Primary Cell Culture, Recombinant Proteins genetics, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, Brain Neoplasms genetics, Ependymoma genetics, Glioma genetics, Oncogene Proteins metabolism, Polycomb Repressive Complex 2 metabolism
- Abstract
Posterior fossa type A (PFA) ependymomas exhibit very low H3K27 methylation and express high levels of EZHIP (Enhancer of Zeste Homologs Inhibitory Protein, also termed CXORF67). Here we find that a conserved sequence in EZHIP is necessary and sufficient to inhibit PRC2 catalytic activity in vitro and in vivo. EZHIP directly contacts the active site of the EZH2 subunit in a mechanism similar to the H3 K27M oncohistone. Furthermore, expression of H3 K27M or EZHIP in cells promotes similar chromatin profiles: loss of broad H3K27me3 domains, but retention of H3K27me3 at CpG islands. We find that H3K27me3-mediated allosteric activation of PRC2 substantially increases the inhibition potential of EZHIP and H3 K27M, providing a mechanism to explain the observed loss of H3K27me3 spreading in tumors. Our data indicate that PFA ependymoma and DIPG are driven in part by the action of peptidyl PRC2 inhibitors, the K27M oncohistone and the EZHIP 'oncohistone-mimic', that dysregulate gene silencing to promote tumorigenesis.
- Published
- 2019
- Full Text
- View/download PDF
17. H3K27M induces defective chromatin spread of PRC2-mediated repressive H3K27me2/me3 and is essential for glioma tumorigenesis.
- Author
-
Harutyunyan AS, Krug B, Chen H, Papillon-Cavanagh S, Zeinieh M, De Jay N, Deshmukh S, Chen CCL, Belle J, Mikael LG, Marchione DM, Li R, Nikbakht H, Hu B, Cagnone G, Cheung WA, Mohammadnia A, Bechet D, Faury D, McConechy MK, Pathania M, Jain SU, Ellezam B, Weil AG, Montpetit A, Salomoni P, Pastinen T, Lu C, Lewis PW, Garcia BA, Kleinman CL, Jabado N, and Majewski J
- Subjects
- Adolescent, Aged, Animals, Brain Neoplasms pathology, CRISPR-Cas Systems, Carcinogenesis genetics, Cell Line, Tumor, Cell Proliferation genetics, Child, CpG Islands genetics, DNA Methylation genetics, Epigenesis, Genetic, Female, Gene Editing methods, Gene Expression Regulation, Neoplastic, Glioblastoma pathology, HEK293 Cells, Histone Code genetics, Histones metabolism, Humans, Lysine genetics, Male, Methionine genetics, Mice, Mice, Inbred NOD, Mice, SCID, Mutation, Neurogenesis genetics, Xenograft Model Antitumor Assays, Brain Neoplasms genetics, Chromatin metabolism, Glioblastoma genetics, Histones genetics, Polycomb Repressive Complex 2 metabolism
- Abstract
Lys-27-Met mutations in histone 3 genes (H3K27M) characterize a subgroup of deadly gliomas and decrease genome-wide H3K27 trimethylation. Here we use primary H3K27M tumor lines and isogenic CRISPR-edited controls to assess H3K27M effects in vitro and in vivo. We find that whereas H3K27me3 and H3K27me2 are normally deposited by PRC2 across broad regions, their deposition is severely reduced in H3.3K27M cells. H3K27me3 is unable to spread from large unmethylated CpG islands, while H3K27me2 can be deposited outside these PRC2 high-affinity sites but to levels corresponding to H3K27me3 deposition in wild-type cells. Our findings indicate that PRC2 recruitment and propagation on chromatin are seemingly unaffected by K27M, which mostly impairs spread of the repressive marks it catalyzes, especially H3K27me3. Genome-wide loss of H3K27me3 and me2 deposition has limited transcriptomic consequences, preferentially affecting lowly-expressed genes regulating neurogenesis. Removal of H3K27M restores H3K27me2/me3 spread, impairs cell proliferation, and completely abolishes their capacity to form tumors in mice.
- Published
- 2019
- Full Text
- View/download PDF
18. Characterization of histone acylations links chromatin modifications with metabolism.
- Author
-
Simithy J, Sidoli S, Yuan ZF, Coradin M, Bhanu NV, Marchione DM, Klein BJ, Bazilevsky GA, McCullough CE, Magin RS, Kutateladze TG, Snyder NW, Marmorstein R, and Garcia BA
- Abstract
Over the last decade, numerous histone acyl post-translational modifications (acyl-PTMs) have been discovered, of which the functional significance is still under intense study. Here, we use high-resolution mass spectrometry to accurately quantify eight acyl-PTMs in vivo and after in vitro enzymatic assays. We assess the ability of seven histone acetyltransferases (HATs) to catalyze acylations on histones in vitro using short-chain acyl-CoA donors, proving that they are less efficient towards larger acyl-CoAs. We also observe that acyl-CoAs can acylate histones through non-enzymatic mechanisms. Using integrated metabolomic and proteomic approaches, we achieve high correlation (R
2 > 0.99) between the abundance of acyl-CoAs and their corresponding acyl-PTMs. Moreover, we observe a dose-dependent increase in histone acyl-PTM abundances in response to acyl-CoA supplementation in in nucleo reactions. This study represents a comprehensive profiling of scarcely investigated low-abundance histone marks, revealing that concentrations of acyl-CoAs affect histone acyl-PTM abundances by both enzymatic and non-enzymatic mechanisms.- Published
- 2017
- Full Text
- View/download PDF
19. An HDAC3-PROX1 corepressor module acts on HNF4α to control hepatic triglycerides.
- Author
-
Armour SM, Remsberg JR, Damle M, Sidoli S, Ho WY, Li Z, Garcia BA, and Lazar MA
- Subjects
- Animals, Gene Expression Regulation, Hepatocyte Nuclear Factor 4 genetics, Histone Deacetylases genetics, Homeodomain Proteins genetics, Lipids genetics, Male, Mice, Knockout, Protein Interaction Mapping methods, Tumor Suppressor Proteins genetics, Prospero-Related Homeobox 1 Protein, Hepatocyte Nuclear Factor 4 metabolism, Histone Deacetylases metabolism, Homeodomain Proteins metabolism, Liver metabolism, Triglycerides metabolism, Tumor Suppressor Proteins metabolism
- Abstract
The histone deacetylase HDAC3 is a critical mediator of hepatic lipid metabolism, and liver-specific deletion of HDAC3 leads to fatty liver. To elucidate the underlying mechanism, here we report a method of cross-linking followed by mass spectrometry to define a high-confidence HDAC3 interactome in vivo that includes the canonical NCoR-HDAC3 complex as well as Prospero-related homeobox 1 protein (PROX1). HDAC3 and PROX1 co-localize extensively on the mouse liver genome, and are co-recruited by hepatocyte nuclear factor 4α (HNF4α). The HDAC3-PROX1 module controls the expression of a gene program regulating lipid homeostasis, and hepatic-specific ablation of either component increases triglyceride content in liver. These findings underscore the importance of specific combinations of transcription factors and coregulators in the fine tuning of organismal metabolism.HDAC3 is a critical mediator of hepatic lipid metabolism and its loss leads to fatty liver. Here, the authors characterize the liver HDAC3 interactome in vivo, provide evidence that HDAC3 interacts with PROX1, and show that HDAC3 and PROX1 control expression of genes regulating lipid homeostasis.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.