721 results on '"Histones metabolism"'
Search Results
2. The H3.3K36M oncohistone disrupts the establishment of epigenetic memory through loss of DNA methylation.
- Author
-
Sinha J, Nickels JF, Thurm AR, Ludwig CH, Archibald BN, Hinks MM, Wan J, Fang D, and Bintu L
- Subjects
- Humans, Acetylation, Promoter Regions, Genetic, Repressor Proteins metabolism, Repressor Proteins genetics, Gene Expression Regulation, Neoplastic, Cell Line, Tumor, Lysine metabolism, Epigenetic Memory, Histones metabolism, Histones genetics, DNA Methylation, Epigenesis, Genetic, Mutation
- Abstract
Histone H3.3 is frequently mutated in tumors, with the lysine 36 to methionine mutation (K36M) being a hallmark of chondroblastomas. While it is known that H3.3K36M changes the epigenetic landscape, its effects on gene expression dynamics remain unclear. Here, we use a synthetic reporter to measure the effects of H3.3K36M on silencing and epigenetic memory after recruitment of the ZNF10 Krüppel-associated box (KRAB) domain, part of the largest class of human repressors and associated with H3K9me3 deposition. We find that H3.3K36M, which decreases H3K36 methylation and increases histone acetylation, leads to a decrease in epigenetic memory and promoter methylation weeks after KRAB release. We propose a model for establishment and maintenance of epigenetic memory, where the H3K36 methylation pathway is necessary to maintain histone deacetylation and convert H3K9me3 domains into DNA methylation for stable epigenetic memory. Our quantitative model can inform oncogenic mechanisms and guide development of epigenetic editing tools., Competing Interests: Declaration of interests L.B. is a co-founder of Stylus Medicine and a member of its scientific advisory board., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Activation of automethylated PRC2 by dimerization on chromatin.
- Author
-
Sauer PV, Pavlenko E, Cookis T, Zirden LC, Renn J, Singhal A, Hunold P, Hoehne-Wiechmann MN, van Ray O, Kaschani F, Kaiser M, Hänsel-Hertsch R, Sanbonmatsu KY, Nogales E, and Poepsel S
- Subjects
- Humans, Methylation, Nucleosomes metabolism, Nucleosomes genetics, Allosteric Regulation, HEK293 Cells, Protein Binding, Polycomb Repressive Complex 2 metabolism, Polycomb Repressive Complex 2 genetics, Chromatin metabolism, Chromatin genetics, Histones metabolism, Histones genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Enhancer of Zeste Homolog 2 Protein genetics, Protein Multimerization
- Abstract
Polycomb repressive complex 2 (PRC2) is an epigenetic regulator that trimethylates lysine 27 of histone 3 (H3K27me3) and is essential for embryonic development and cellular differentiation. H3K27me3 is associated with transcriptionally repressed chromatin and is established when PRC2 is allosterically activated upon methyl-lysine binding by the regulatory subunit EED. Automethylation of the catalytic subunit enhancer of zeste homolog 2 (EZH2) stimulates its activity by an unknown mechanism. Here, we show that human PRC2 forms a dimer on chromatin in which an inactive, automethylated PRC2 protomer is the allosteric activator of a second PRC2 that is poised to methylate H3 of a substrate nucleosome. Functional assays support our model of allosteric trans-autoactivation via EED, suggesting a previously unknown mechanism mediating context-dependent activation of PRC2. Our work showcases the molecular mechanism of auto-modification-coupled dimerization in the regulation of chromatin-modifying complexes., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Stabilization of the hexasome intermediate during histone exchange by yeast SWR1 complex.
- Author
-
Jalal ASB, Girvan P, Chua EYD, Liu L, Wang S, McCormack EA, Skehan MT, Knight CL, Rueda DS, and Wigley DB
- Subjects
- Models, Molecular, Adenosine Triphosphatases metabolism, Adenosine Triphosphatases genetics, Adenosine Triphosphatases chemistry, Protein Binding, Chromosomal Proteins, Non-Histone metabolism, Chromosomal Proteins, Non-Histone chemistry, Chromosomal Proteins, Non-Histone genetics, Histone Chaperones, Histones metabolism, Histones genetics, Histones chemistry, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae genetics, Nucleosomes metabolism, Nucleosomes ultrastructure, Nucleosomes genetics, Cryoelectron Microscopy, Protein Multimerization
- Abstract
The yeast SWR1 complex catalyzes the exchange of histone H2A/H2B dimers in nucleosomes with Htz1/H2B dimers. We use cryoelectron microscopy to determine the structure of an enzyme-bound hexasome intermediate in the reaction pathway of histone exchange, in which an H2A/H2B dimer has been extracted from a nucleosome prior to the insertion of a dimer comprising Htz1/H2B. The structure reveals a key role for the Swc5 subunit in stabilizing the unwrapping of DNA from the histone core of the hexasome. By engineering a crosslink between an Htz1/H2B dimer and its chaperone protein Chz1, we show that this blocks histone exchange by SWR1 but allows the incoming chaperone-dimer complex to insert into the hexasome. We use this reagent to trap an SWR1/hexasome complex with an incoming Htz1/H2B dimer that shows how the reaction progresses to the next step. Taken together the structures reveal insights into the mechanism of histone exchange by SWR1 complex., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Finish the unfinished: Chd1 resolving hexasome-nucleosome complex with FACT.
- Author
-
Yin H and Liu Y
- Subjects
- Chromatin Assembly and Disassembly, Histones metabolism, Histones genetics, Humans, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins genetics, DNA Helicases metabolism, DNA Helicases genetics, Protein Binding, Transcription, Genetic, High Mobility Group Proteins metabolism, High Mobility Group Proteins genetics, Transcriptional Elongation Factors metabolism, Transcriptional Elongation Factors genetics, Transcriptional Elongation Factors chemistry, Nucleosomes metabolism, Nucleosomes genetics, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, Cryoelectron Microscopy
- Abstract
In this issue of Molecular Cell, Engeholm et al.
1 present cryo-EM structures of the chromatin remodeler Chd1 bound to a hexasome-nucleosome complex, an intermediate state during transcription either with or without FACT to restore the missing H2A-H2B dimer. These two binding modes reveal how Chd1 and FACT cooperate in nucleosome re-establishment during transcription., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
6. Resolution of transcription-induced hexasome-nucleosome complexes by Chd1 and FACT.
- Author
-
Engeholm M, Roske JJ, Oberbeckmann E, Dienemann C, Lidschreiber M, Cramer P, and Farnung L
- Subjects
- Protein Binding, Models, Molecular, Adenosine Triphosphatases metabolism, Adenosine Triphosphatases genetics, Nucleosomes metabolism, Nucleosomes genetics, Nucleosomes ultrastructure, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Cryoelectron Microscopy, Transcriptional Elongation Factors metabolism, Transcriptional Elongation Factors genetics, Transcriptional Elongation Factors chemistry, Chromatin Assembly and Disassembly, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, Transcription, Genetic, High Mobility Group Proteins metabolism, High Mobility Group Proteins genetics, RNA Polymerase II metabolism, RNA Polymerase II genetics, Histones metabolism, Histones genetics
- Abstract
To maintain the nucleosome organization of transcribed genes, ATP-dependent chromatin remodelers collaborate with histone chaperones. Here, we show that at the 5' ends of yeast genes, RNA polymerase II (RNAPII) generates hexasomes that occur directly adjacent to nucleosomes. The resulting hexasome-nucleosome complexes are then resolved by Chd1. We present two cryoelectron microscopy (cryo-EM) structures of Chd1 bound to a hexasome-nucleosome complex before and after restoration of the missing inner H2A/H2B dimer by FACT. Chd1 uniquely interacts with the complex, positioning its ATPase domain to shift the hexasome away from the nucleosome. In the absence of the inner H2A/H2B dimer, its DNA-binding domain (DBD) packs against the ATPase domain, suggesting an inhibited state. Restoration of the dimer by FACT triggers a rearrangement that displaces the DBD and stimulates Chd1 remodeling. Our results demonstrate how chromatin remodelers interact with a complex nucleosome assembly and suggest how Chd1 and FACT jointly support transcription by RNAPII., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. PICKLE-mediated nucleosome condensing drives H3K27me3 spreading for the inheritance of Polycomb memory during differentiation.
- Author
-
Liang Z, Zhu T, Yu Y, Wu C, Huang Y, Hao Y, Song X, Fu W, Yuan L, Cui Y, Huang S, and Li C
- Subjects
- Gene Expression Regulation, Plant, Chromatin metabolism, Chromatin genetics, Nucleosomes metabolism, Nucleosomes genetics, Arabidopsis genetics, Arabidopsis metabolism, Histones metabolism, Histones genetics, Arabidopsis Proteins metabolism, Arabidopsis Proteins genetics, Polycomb Repressive Complex 2 metabolism, Polycomb Repressive Complex 2 genetics, Chromatin Assembly and Disassembly, Cell Differentiation
- Abstract
Spreading of H3K27me3 is crucial for the maintenance of mitotically inheritable Polycomb-mediated chromatin silencing in animals and plants. However, how Polycomb repressive complex 2 (PRC2) accesses unmodified nucleosomes in spreading regions for spreading H3K27me3 remains unclear. Here, we show in Arabidopsis thaliana that the chromatin remodeler PICKLE (PKL) plays a specialized role in H3K27me3 spreading to safeguard cell identity during differentiation. PKL specifically localizes to H3K27me3 spreading regions but not to nucleation sites and physically associates with PRC2. Loss of PKL disrupts the occupancy of the PRC2 catalytic subunit CLF in spreading regions and leads to aberrant dedifferentiation. Nucleosome density increase endowed by the ATPase function of PKL ensures that unmodified nucleosomes are accessible to PRC2 catalytic activity for H3K27me3 spreading. Our findings demonstrate that PKL-dependent nucleosome compaction is critical for PRC2-mediated H3K27me3 read-and-write function in H3K27me3 spreading, thus revealing a mechanism by which repressive chromatin domains are established and propagated., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Navigating the complexity of Polycomb repression: Enzymatic cores and regulatory modules.
- Author
-
Tamburri S, Rustichelli S, Amato S, and Pasini D
- Subjects
- Humans, Animals, Methylation, Transcription, Genetic, Polycomb-Group Proteins metabolism, Polycomb-Group Proteins genetics, Histones metabolism, Histones genetics, Protein Processing, Post-Translational
- Abstract
Polycomb proteins are a fundamental repressive system that plays crucial developmental roles by orchestrating cell-type-specific transcription programs that govern cell identity. Direct alterations of Polycomb activity are indeed implicated in human pathologies, including developmental disorders and cancer. General Polycomb repression is coordinated by three distinct activities that regulate the deposition of two histone post-translational modifications: tri-methylation of histone H3 lysine 27 (H3K27me3) and histone H2A at lysine 119 (H2AK119ub1). These activities exist in large and heterogeneous multiprotein ensembles consisting of common enzymatic cores regulated by heterogeneous non-catalytic modules composed of a large number of accessory proteins with diverse biochemical properties. Here, we have analyzed the current molecular knowledge, focusing on the functional interaction between the core enzymatic activities and their regulation mediated by distinct accessory modules. This provides a comprehensive analysis of the molecular details that control the establishment and maintenance of Polycomb repression, examining their underlying coordination and highlighting missing information and emerging new features of Polycomb-mediated transcriptional control., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. Histone chaperone HIRA, promyelocytic leukemia protein, and p62/SQSTM1 coordinate to regulate inflammation during cell senescence.
- Author
-
Dasgupta N, Lei X, Shi CH, Arnold R, Teneche MG, Miller KN, Rajesh A, Davis A, Anschau V, Campos AR, Gilson R, Havas A, Yin S, Chua ZM, Liu T, Proulx J, Alcaraz M, Rather MI, Baeza J, Schultz DC, Yip KY, Berger SL, and Adams PD
- Subjects
- Humans, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing genetics, Autophagy, Chromatin metabolism, Chromatin genetics, HEK293 Cells, Histones metabolism, Histones genetics, Membrane Proteins metabolism, Membrane Proteins genetics, Nucleotidyltransferases, Tumor Suppressor Proteins metabolism, Tumor Suppressor Proteins genetics, Cell Cycle Proteins metabolism, Cell Cycle Proteins genetics, Cellular Senescence, Histone Chaperones metabolism, Histone Chaperones genetics, Inflammation metabolism, Inflammation pathology, Inflammation genetics, NF-kappa B metabolism, NF-kappa B genetics, Nuclear Proteins metabolism, Nuclear Proteins genetics, Promyelocytic Leukemia Protein metabolism, Promyelocytic Leukemia Protein genetics, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases genetics, Sequestosome-1 Protein metabolism, Sequestosome-1 Protein genetics, Signal Transduction, Transcription Factors metabolism, Transcription Factors genetics
- Abstract
Cellular senescence, a stress-induced stable proliferation arrest associated with an inflammatory senescence-associated secretory phenotype (SASP), is a cause of aging. In senescent cells, cytoplasmic chromatin fragments (CCFs) activate SASP via the anti-viral cGAS/STING pathway. Promyelocytic leukemia (PML) protein organizes PML nuclear bodies (NBs), which are also involved in senescence and anti-viral immunity. The HIRA histone H3.3 chaperone localizes to PML NBs in senescent cells. Here, we show that HIRA and PML are essential for SASP expression, tightly linked to HIRA's localization to PML NBs. Inactivation of HIRA does not directly block expression of nuclear factor κB (NF-κB) target genes. Instead, an H3.3-independent HIRA function activates SASP through a CCF-cGAS-STING-TBK1-NF-κB pathway. HIRA physically interacts with p62/SQSTM1, an autophagy regulator and negative SASP regulator. HIRA and p62 co-localize in PML NBs, linked to their antagonistic regulation of SASP, with PML NBs controlling their spatial configuration. These results outline a role for HIRA and PML in the regulation of SASP., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Mrc1 regulates parental histone segregation and heterochromatin inheritance.
- Author
-
Toda T, Fang Y, Shan CM, Hua X, Kim JK, Tang LC, Jovanovic M, Tong L, Qiao F, Zhang Z, and Jia S
- Subjects
- Cell Cycle Proteins metabolism, Cell Cycle Proteins genetics, DNA Polymerase I metabolism, DNA Polymerase I genetics, Heterochromatin metabolism, Heterochromatin genetics, Protein Binding, DNA Replication, Epigenesis, Genetic, Histones metabolism, Histones genetics, Schizosaccharomyces genetics, Schizosaccharomyces metabolism, Schizosaccharomyces pombe Proteins metabolism, Schizosaccharomyces pombe Proteins genetics
- Abstract
Chromatin-based epigenetic memory relies on the symmetric distribution of parental histones to newly synthesized daughter DNA strands, aided by histone chaperones within the DNA replication machinery. However, the mechanism of parental histone transfer remains elusive. Here, we reveal that in fission yeast, the replisome protein Mrc1 plays a crucial role in promoting the transfer of parental histone H3-H4 to the lagging strand, ensuring proper heterochromatin inheritance. In addition, Mrc1 facilitates the interaction between Mcm2 and DNA polymerase alpha, two histone-binding proteins critical for parental histone transfer. Furthermore, Mrc1's involvement in parental histone transfer and epigenetic inheritance is independent of its known functions in DNA replication checkpoint activation and replisome speed control. Instead, Mrc1 interacts with Mcm2 outside of its histone-binding region, creating a physical barrier to separate parental histone transfer pathways. These findings unveil Mrc1 as a key player within the replisome, coordinating parental histone segregation to regulate epigenetic inheritance., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Nucleosome remodeler exclusion by histone deacetylation enforces heterochromatic silencing and epigenetic inheritance.
- Author
-
Sahu RK, Dhakshnamoorthy J, Jain S, Folco HD, Wheeler D, and Grewal SIS
- Subjects
- Acetylation, Humans, Histone Acetyltransferases metabolism, Histone Acetyltransferases genetics, Animals, Heterochromatin metabolism, Heterochromatin genetics, Nucleosomes metabolism, Nucleosomes genetics, Histones metabolism, Histones genetics, Gene Silencing, Epigenesis, Genetic, Histone Deacetylases metabolism, Histone Deacetylases genetics, Chromatin Assembly and Disassembly
- Abstract
Heterochromatin enforces transcriptional gene silencing and can be epigenetically inherited, but the underlying mechanisms remain unclear. Here, we show that histone deacetylation, a conserved feature of heterochromatin domains, blocks SWI/SNF subfamily remodelers involved in chromatin unraveling, thereby stabilizing modified nucleosomes that preserve gene silencing. Histone hyperacetylation, resulting from either the loss of histone deacetylase (HDAC) activity or the direct targeting of a histone acetyltransferase to heterochromatin, permits remodeler access, leading to silencing defects. The requirement for HDAC in heterochromatin silencing can be bypassed by impeding SWI/SNF activity. Highlighting the crucial role of remodelers, merely targeting SWI/SNF to heterochromatin, even in cells with functional HDAC, increases nucleosome turnover, causing defective gene silencing and compromised epigenetic inheritance. This study elucidates a fundamental mechanism whereby histone hypoacetylation, maintained by high HDAC levels in heterochromatic regions, ensures stable gene silencing and epigenetic inheritance, providing insights into genome regulatory mechanisms relevant to human diseases., Competing Interests: Declaration of interests S.I.S.G. is a member of the advisory board of Molecular Cell., (Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
12. FACT mediates the depletion of macroH2A1.2 to expedite gene transcription.
- Author
-
Ji D, Xiao X, Luo A, Fan X, Ma J, Wang D, Xia M, Ma L, Wang PY, Li W, and Chen P
- Subjects
- Humans, High Mobility Group Proteins metabolism, High Mobility Group Proteins genetics, Animals, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, Macrophages metabolism, Mutation, Chromatin Assembly and Disassembly, Mice, Chromatin metabolism, Chromatin genetics, Gene Expression Regulation, RAW 264.7 Cells, Protein Binding, HEK293 Cells, Nucleosomes metabolism, Nucleosomes genetics, Histones metabolism, Histones genetics, Transcription, Genetic, Transcriptional Elongation Factors genetics, Transcriptional Elongation Factors metabolism
- Abstract
The histone variant macroH2A is generally linked to transcriptionally inactive chromatin, but how macroH2A regulates chromatin structure and functions in the transcriptional process remains elusive. This study reveals that while the integration of human macroH2A1.2 into nucleosomes does not affect their stability or folding dynamics, it notably hinders the maintenance of facilitates chromatin transcription's (FACT's) function. We show that FACT effectively diminishes the stability of macroH2A1.2-nucleosomes and expedites their depletion subsequent to the initial unfolding process. Furthermore, we identify the residue S139 in macroH2A1.2 as a critical switch to modulate FACT's function in nucleosome maintenance. Genome-wide analyses demonstrate that FACT-mediated depletion of macroH2A-nucleosomes allows the correct localization of macroH2A, while the S139 mutation reshapes macroH2A distribution and influences stimulation-induced transcription and cellular response in macrophages. Our findings provide mechanistic insights into the intricate interplay between macroH2A and FACT at the nucleosome level and elucidate their collective role in transcriptional regulation and immune response of macrophages., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Structural insights into the cooperative nucleosome recognition and chromatin opening by FOXA1 and GATA4.
- Author
-
Zhou BR, Feng H, Huang F, Zhu I, Portillo-Ledesma S, Shi D, Zaret KS, Schlick T, Landsman D, Wang Q, and Bai Y
- Subjects
- Animals, Mice, Chromatin metabolism, Chromatin genetics, Histones metabolism, Histones genetics, Histones chemistry, Binding Sites, DNA metabolism, DNA genetics, DNA chemistry, Chromatin Assembly and Disassembly, Humans, Hepatocyte Nuclear Factor 3-alpha metabolism, Hepatocyte Nuclear Factor 3-alpha genetics, Nucleosomes metabolism, Nucleosomes genetics, Nucleosomes ultrastructure, GATA4 Transcription Factor metabolism, GATA4 Transcription Factor genetics, GATA4 Transcription Factor chemistry, Cryoelectron Microscopy, Protein Binding
- Abstract
Mouse FOXA1 and GATA4 are prototypes of pioneer factors, initiating liver cell development by binding to the N1 nucleosome in the enhancer of the ALB1 gene. Using cryoelectron microscopy (cryo-EM), we determined the structures of the free N1 nucleosome and its complexes with FOXA1 and GATA4, both individually and in combination. We found that the DNA-binding domains of FOXA1 and GATA4 mainly recognize the linker DNA and an internal site in the nucleosome, respectively, whereas their intrinsically disordered regions interact with the acidic patch on histone H2A-H2B. FOXA1 efficiently enhances GATA4 binding by repositioning the N1 nucleosome. In vivo DNA editing and bioinformatics analyses suggest that the co-binding mode of FOXA1 and GATA4 plays important roles in regulating genes involved in liver cell functions. Our results reveal the mechanism whereby FOXA1 and GATA4 cooperatively bind to the nucleosome through nucleosome repositioning, opening chromatin by bending linker DNA and obstructing nucleosome packing., Competing Interests: Declaration of interests The authors declare no competing interests., (Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
14. A FACT about macroH2A removal in immune gene activation.
- Author
-
Meers O and Buschbeck M
- Subjects
- Humans, Immunity, Innate genetics, Animals, Transcriptional Activation, Histones metabolism, Histones genetics, Epigenesis, Genetic
- Abstract
Histone variants contribute to epigenetic regulation in development and disease but require the chaperone machinery for correct deposition. In this issue of Molecular Cell, Ji et al.
1 explain how the chaperone complex FACT removes the histone variant macroH2A1.2 and demonstrate its importance for gene activation in innate immune cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
15. Histone variant H2BE enhances chromatin accessibility in neurons to promote synaptic gene expression and long-term memory.
- Author
-
Feierman ER, Louzon S, Prescott NA, Biaco T, Gao Q, Qiu Q, Choi K, Palozola KC, Voss AJ, Mehta SD, Quaye CN, Lynch KT, Fuccillo MV, Wu H, David Y, and Korb E
- Subjects
- Animals, Mice, Promoter Regions, Genetic, Mice, Inbred C57BL, Gene Expression Regulation, Transcription, Genetic, Male, Humans, Histones metabolism, Histones genetics, Chromatin metabolism, Chromatin genetics, Memory, Long-Term physiology, Neurons metabolism, Synapses metabolism, Synapses genetics
- Abstract
Histone proteins affect gene expression through multiple mechanisms, including through exchange with histone variants. Recent findings link histone variants to neurological disorders, yet few are well studied in the brain. Most notably, widely expressed variants of H2B remain elusive. We applied recently developed antibodies, biochemical assays, and sequencing approaches to reveal broad expression of the H2B variant H2BE and defined its role in regulating chromatin structure, neuronal transcription, and mouse behavior. We find that H2BE is enriched at promoters, and a single unique amino acid allows it to dramatically enhance chromatin accessibility. Further, we show that H2BE is critical for synaptic gene expression and long-term memory. Together, these data reveal a mechanism linking histone variants to chromatin accessibility, transcriptional regulation, neuronal function, and memory. This work further identifies a widely expressed H2B variant and uncovers a single histone amino acid with profound effects on genomic structure., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Competition between two HUSH complexes orchestrates the immune response to retroelement invasion.
- Author
-
Danac JMC, Matthews RE, Gungi A, Qin C, Parsons H, Antrobus R, Timms RT, and Tchasovnikarova IA
- Subjects
- Humans, HEK293 Cells, Histones metabolism, Histones genetics, Retroelements genetics, Epigenesis, Genetic, Long Interspersed Nucleotide Elements genetics, Signal Transduction, Interferons metabolism, Interferons immunology, Interferons genetics, HeLa Cells, Gene Silencing
- Abstract
The human silencing hub (HUSH) preserves genome integrity through the epigenetic repression of invasive genetic elements. However, despite our understanding of HUSH as an obligate complex of three subunits, only loss of MPP8 or Periphilin, but not TASOR, triggers interferon signaling following derepression of endogenous retroelements. Here, we resolve this paradox by characterizing a second HUSH complex that shares MPP8 and Periphilin but assembles around TASOR2, an uncharacterized paralog of TASOR. Whereas HUSH represses LINE-1 retroelements marked by the repressive histone modification H3K9me3, HUSH2 is recruited by the transcription factor IRF2 to repress interferon-stimulated genes. Mechanistically, HUSH-mediated retroelement silencing sequesters the limited pool of the shared subunits MPP8 and Periphilin, preventing TASOR2 from forming HUSH2 complexes and hence relieving the HUSH2-mediated repression of interferon-stimulated genes. Thus, competition between two HUSH complexes intertwines retroelement silencing with the induction of an immune response, coupling epigenetic and immune aspects of genome defense., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
17. Structure of the Hir histone chaperone complex.
- Author
-
Kim HJ, Szurgot MR, van Eeuwen T, Ricketts MD, Basnet P, Zhang AL, Vogt A, Sharmin S, Kaplan CD, Garcia BA, Marmorstein R, and Murakami K
- Subjects
- Models, Molecular, Molecular Chaperones metabolism, Molecular Chaperones chemistry, Molecular Chaperones genetics, Protein Multimerization, Binding Sites, Transcription Factors metabolism, Transcription Factors chemistry, Transcription Factors genetics, Protein Interaction Domains and Motifs, Histones metabolism, Histones chemistry, Histones genetics, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins ultrastructure, Cryoelectron Microscopy, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Cell Cycle Proteins metabolism, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Histone Chaperones metabolism, Histone Chaperones chemistry, Histone Chaperones genetics, Protein Binding
- Abstract
The evolutionarily conserved HIRA/Hir histone chaperone complex and ASF1a/Asf1 co-chaperone cooperate to deposit histone (H3/H4)
2 tetramers on DNA for replication-independent chromatin assembly. The molecular architecture of the HIRA/Hir complex and its mode of histone deposition have remained unknown. Here, we report the cryo-EM structure of the S. cerevisiae Hir complex with Asf1/H3/H4 at 2.9-6.8 Å resolution. We find that the Hir complex forms an arc-shaped dimer with a Hir1/Hir2/Hir3/Hpc2 stoichiometry of 2/4/2/4. The core of the complex containing two Hir1/Hir2/Hir2 trimers and N-terminal segments of Hir3 forms a central cavity containing two copies of Hpc2, with one engaged by Asf1/H3/H4, in a suitable position to accommodate a histone (H3/H4)2 tetramer, while the C-terminal segments of Hir3 harbor nucleic acid binding activity to wrap DNA around the Hpc2-assisted histone tetramer. The structure suggests a model for how the Hir/Asf1 complex promotes the formation of histone tetramers for their subsequent deposition onto DNA., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
18. Proximal termination generates a transcriptional state that determines the rate of establishment of Polycomb silencing.
- Author
-
Menon G, Mateo-Bonmati E, Reeck S, Maple R, Wu Z, Ietswaart R, Dean C, and Howard M
- Subjects
- Transcription, Genetic, Polyadenylation, Histone Demethylases metabolism, Histone Demethylases genetics, Transcription Termination, Genetic, Chromatin metabolism, Chromatin genetics, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics, Arabidopsis genetics, Arabidopsis metabolism, Arabidopsis Proteins genetics, Arabidopsis Proteins metabolism, Gene Silencing, Gene Expression Regulation, Plant, Histones metabolism, Histones genetics, RNA Polymerase II metabolism, RNA Polymerase II genetics, Polycomb Repressive Complex 2 metabolism, Polycomb Repressive Complex 2 genetics, MADS Domain Proteins genetics, MADS Domain Proteins metabolism
- Abstract
The mechanisms and timescales controlling de novo establishment of chromatin-mediated transcriptional silencing by Polycomb repressive complex 2 (PRC2) are unclear. Here, we investigate PRC2 silencing at Arabidopsis FLOWERING LOCUS C (FLC), known to involve co-transcriptional RNA processing, histone demethylation activity, and PRC2 function, but so far not mechanistically connected. We develop and test a computational model describing proximal polyadenylation/termination mediated by the RNA-binding protein FCA that induces H3K4me1 removal by the histone demethylase FLD. H3K4me1 removal feeds back to reduce RNA polymerase II (RNA Pol II) processivity and thus enhance early termination, thereby repressing productive transcription. The model predicts that this transcription-coupled repression controls the level of transcriptional antagonism to PRC2 action. Thus, the effectiveness of this repression dictates the timescale for establishment of PRC2/H3K27me3 silencing. We experimentally validate these mechanistic model predictions, revealing that co-transcriptional processing sets the level of productive transcription at the locus, which then determines the rate of the ON-to-OFF switch to PRC2 silencing., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
19. A CPF-like phosphatase module links transcription termination to chromatin silencing.
- Author
-
Mateo-Bonmatí E, Montez M, Maple R, Fiedler M, Fang X, Saalbach G, Passmore LA, and Dean C
- Subjects
- Phosphoprotein Phosphatases genetics, Phosphoprotein Phosphatases metabolism, mRNA Cleavage and Polyadenylation Factors metabolism, mRNA Cleavage and Polyadenylation Factors genetics, Histones metabolism, Histones genetics, Histone Deacetylases, Arabidopsis Proteins genetics, Arabidopsis Proteins metabolism, Arabidopsis genetics, Arabidopsis metabolism, Arabidopsis enzymology, Chromatin metabolism, Chromatin genetics, Gene Expression Regulation, Plant, MADS Domain Proteins genetics, MADS Domain Proteins metabolism, Transcription Termination, Genetic, RNA Polymerase II metabolism, RNA Polymerase II genetics, Gene Silencing
- Abstract
The interconnections between co-transcriptional regulation, chromatin environment, and transcriptional output remain poorly understood. Here, we investigate the mechanism underlying RNA 3' processing-mediated Polycomb silencing of Arabidopsis FLOWERING LOCUS C (FLC). We show a requirement for ANTHESIS PROMOTING FACTOR 1 (APRF1), a homolog of yeast Swd2 and human WDR82, known to regulate RNA polymerase II (RNA Pol II) during transcription termination. APRF1 interacts with TYPE ONE SERINE/THREONINE PROTEIN PHOSPHATASE 4 (TOPP4) (yeast Glc7/human PP1) and LUMINIDEPENDENS (LD), the latter showing structural features found in Ref2/PNUTS, all components of the yeast and human phosphatase module of the CPF 3' end-processing machinery. LD has been shown to co-associate in vivo with the histone H3 K4 demethylase FLOWERING LOCUS D (FLD). This work shows how the APRF1/LD-mediated polyadenylation/termination process influences subsequent rounds of transcription by changing the local chromatin environment at FLC., Competing Interests: Declaration of interests L.A.P. is on the advisory board for Molecular Cell., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. Heterochromatic 3D genome organization is directed by HP1a- and H3K9-dependent and independent mechanisms.
- Author
-
Stutzman AV, Hill CA, Armstrong RL, Gohil R, Duronio RJ, Dowen JM, and McKay DJ
- Subjects
- Animals, Methylation, Euchromatin metabolism, Euchromatin genetics, Centromere metabolism, Centromere genetics, Protein Binding, Genome, Insect, Chromosome Segregation, Protein Processing, Post-Translational, Heterochromatin metabolism, Heterochromatin genetics, Histones metabolism, Histones genetics, Chromosomal Proteins, Non-Histone metabolism, Chromosomal Proteins, Non-Histone genetics, Chromobox Protein Homolog 5, Drosophila Proteins metabolism, Drosophila Proteins genetics, Drosophila melanogaster genetics, Drosophila melanogaster metabolism
- Abstract
Whether and how histone post-translational modifications and the proteins that bind them drive 3D genome organization remains unanswered. Here, we evaluate the contribution of H3K9-methylated constitutive heterochromatin to 3D genome organization in Drosophila tissues. We find that the predominant organizational feature of wild-type tissues is the segregation of euchromatic chromosome arms from heterochromatic pericentromeres. Reciprocal perturbation of HP1a⋅H3K9me binding, using a point mutation in the HP1a chromodomain or replacement of the replication-dependent histone H3 with H3
K9R mutant histones, revealed that HP1a binding to methylated H3K9 in constitutive heterochromatin is required to limit contact frequency between pericentromeres and chromosome arms and regulate the distance between arm and pericentromeric regions. Surprisingly, the self-association of pericentromeric regions is largely preserved despite the loss of H3K9 methylation and HP1a occupancy. Thus, the HP1a⋅H3K9 interaction contributes to but does not solely drive the segregation of euchromatin and heterochromatin inside the nucleus., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
21. Learning from chromatin reconstitution: Pioneer factors enabling nucleosome remodelers.
- Author
-
Zaret K
- Subjects
- Humans, Animals, Histones metabolism, Histones genetics, Nucleosomes metabolism, Nucleosomes genetics, Chromatin Assembly and Disassembly, Chromatin metabolism, Chromatin genetics
- Abstract
Competing Interests: Declaration of interests The author declares no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
22. G-quadruplex folding in Xist RNA antagonizes PRC2 activity for stepwise regulation of X chromosome inactivation.
- Author
-
Lee YW, Weissbein U, Blum R, and Lee JT
- Subjects
- Animals, Mice, Mouse Embryonic Stem Cells metabolism, Chromatin metabolism, Chromatin genetics, X Chromosome genetics, X Chromosome metabolism, Gene Silencing, RNA Folding, Protein Binding, X Chromosome Inactivation, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, Polycomb Repressive Complex 2 metabolism, Polycomb Repressive Complex 2 genetics, G-Quadruplexes, Histones metabolism, Histones genetics
- Abstract
How Polycomb repressive complex 2 (PRC2) is regulated by RNA remains an unsolved problem. Although PRC2 binds G-tracts with the potential to form RNA G-quadruplexes (rG4s), whether rG4s fold extensively in vivo and whether PRC2 binds folded or unfolded rG4 are unknown. Using the X-inactivation model in mouse embryonic stem cells, here we identify multiple folded rG4s in Xist RNA and demonstrate that PRC2 preferentially binds folded rG4s. High-affinity rG4 binding inhibits PRC2's histone methyltransferase activity, and stabilizing rG4 in vivo antagonizes H3 at lysine 27 (H3K27me3) enrichment on the inactive X chromosome. Surprisingly, mutagenizing the rG4 does not affect PRC2 recruitment but promotes its release and catalytic activation on chromatin. H3K27me3 marks are misplaced, however, and gene silencing is compromised. Xist-PRC2 complexes become entrapped in the S1 chromosome compartment, precluding the required translocation into the S2 compartment. Thus, Xist rG4 folding controls PRC2 activity, H3K27me3 enrichment, and the stepwise regulation of chromosome-wide gene silencing., Competing Interests: Declaration of interests J.T.L. is a cofounder of Fulcrum Therapeutics, a scientific adviser to Skyhawk Therapeutics, and a Non-Executive Director of GSK., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
23. Principles of chromosome organization for meiotic recombination.
- Author
-
Biot M, Toth A, Brun C, Guichard L, de Massy B, and Grey C
- Subjects
- Animals, Mice, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, Histones metabolism, Histones genetics, Cell Cycle Proteins metabolism, Cell Cycle Proteins genetics, Binding Sites, Chromosomes genetics, Chromosomes metabolism, Chromatin metabolism, Chromatin genetics, CCCTC-Binding Factor metabolism, CCCTC-Binding Factor genetics, Humans, Nuclear Proteins genetics, Nuclear Proteins metabolism, Recombination, Genetic, Male, Meiosis genetics, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, DNA Breaks, Double-Stranded
- Abstract
In meiotic cells, chromosomes are organized as chromatin loop arrays anchored to a protein axis. This organization is essential to regulate meiotic recombination, from DNA double-strand break (DSB) formation to their repair. In mammals, it is unknown how chromatin loops are organized along the genome and how proteins participating in DSB formation are tethered to the chromosome axes. Here, we identify three categories of axis-associated genomic sites: PRDM9 binding sites, where DSBs form; binding sites of the insulator protein CTCF; and H3K4me3-enriched sites. We demonstrate that PRDM9 promotes the recruitment of MEI4 and IHO1, two proteins essential for DSB formation. In turn, IHO1 anchors DSB sites to the axis components HORMAD1 and SYCP3. We discovered that IHO1, HORMAD1, and SYCP3 are associated at the DSB ends during DSB repair. Our results highlight how interactions of proteins with specific genomic elements shape the meiotic chromosome organization for recombination., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. H3K4me1 facilitates promoter-enhancer interactions and gene activation during embryonic stem cell differentiation.
- Author
-
Kubo N, Chen PB, Hu R, Ye Z, Sasaki H, and Ren B
- Subjects
- Animals, Mice, Transcriptional Activation, Methylation, Gene Expression Regulation, Developmental, Myeloid-Lymphoid Leukemia Protein metabolism, Myeloid-Lymphoid Leukemia Protein genetics, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, Promoter Regions, Genetic, Enhancer Elements, Genetic, Histones metabolism, Histones genetics, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Mouse Embryonic Stem Cells metabolism, Mouse Embryonic Stem Cells cytology, Cell Differentiation, Lysine analogs & derivatives
- Abstract
Histone H3 lysine 4 mono-methylation (H3K4me1) marks poised or active enhancers. KMT2C (MLL3) and KMT2D (MLL4) catalyze H3K4me1, but their histone methyltransferase activities are largely dispensable for transcription during early embryogenesis in mammals. To better understand the role of H3K4me1 in enhancer function, we analyze dynamic enhancer-promoter (E-P) interactions and gene expression during neural differentiation of the mouse embryonic stem cells. We found that KMT2C/D catalytic activities were only required for H3K4me1 and E-P contacts at a subset of candidate enhancers, induced upon neural differentiation. By contrast, a majority of enhancers retained H3K4me1 in KMT2C/D catalytic mutant cells. Surprisingly, H3K4me1 signals at these KMT2C/D-independent sites were reduced after acute depletion of KMT2B, resulting in aggravated transcriptional defects. Our observations therefore implicate KMT2B in the catalysis of H3K4me1 at enhancers and provide additional support for an active role of H3K4me1 in enhancer-promoter interactions and transcription in mammalian cells., Competing Interests: Declaration of interests B.R. is a co-founder of Arima Genomics, Inc. and Epigenome Technologies, Inc., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
25. An RNA-dependent and phase-separated active subnuclear compartment safeguards repressive chromatin domains.
- Author
-
Lerra L, Panatta M, Bär D, Zanini I, Tan JY, Pisano A, Mungo C, Baroux C, Panse VG, Marques AC, and Santoro R
- Subjects
- Humans, Cell Nucleus metabolism, Cell Nucleus genetics, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics, HeLa Cells, Transcription, Genetic, RNA metabolism, RNA genetics, Animals, Gene Expression Regulation, Chromatin metabolism, Chromatin genetics, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, Histones metabolism, Histones genetics
- Abstract
The nucleus is composed of functionally distinct membraneless compartments that undergo phase separation (PS). However, whether different subnuclear compartments are connected remains elusive. We identified a type of nuclear body with PS features composed of BAZ2A that associates with active chromatin. BAZ2A bodies depend on RNA transcription and BAZ2A non-disordered RNA-binding TAM domain. Although BAZ2A and H3K27me3 occupancies anticorrelate in the linear genome, in the nuclear space, BAZ2A bodies contact H3K27me3 bodies. BAZ2A-body disruption promotes BAZ2A invasion into H3K27me3 domains, causing H3K27me3-body loss and gene upregulation. Weak BAZ2A-RNA interactions, such as with nascent transcripts, promote BAZ2A bodies, whereas the strong binder long non-coding RNA (lncRNA) Malat1 impairs them while mediating BAZ2A association to chromatin at nuclear speckles. In addition to unraveling a direct connection between nuclear active and repressive compartments through PS mechanisms, the results also showed that the strength of RNA-protein interactions regulates this process, contributing to nuclear organization and the regulation of chromatin and gene expression., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
26. A di-acetyl-decorated chromatin signature couples liquid condensation to suppress DNA end synapsis.
- Author
-
Bao K, Ma Y, Li Y, Shen X, Zhao J, Tian S, Zhang C, Liang C, Zhao Z, Yang Y, Zhang K, Yang N, Meng FL, Hao J, Yang J, Liu T, Yao Z, Ai D, and Shi L
- Subjects
- Animals, Transcription Factors metabolism, DNA genetics, DNA End-Joining Repair, Histones genetics, Histones metabolism, Chromosome Pairing, Ku Autoantigen genetics, Ku Autoantigen metabolism, Mammals metabolism, Chromatin genetics, Nuclear Proteins metabolism
- Abstract
Appropriate DNA end synapsis, regulated by core components of the synaptic complex including KU70-KU80, LIG4, XRCC4, and XLF, is central to non-homologous end joining (NHEJ) repair of chromatinized DNA double-strand breaks (DSBs). However, it remains enigmatic whether chromatin modifications can influence the formation of NHEJ synaptic complex at DNA ends, and if so, how this is achieved. Here, we report that the mitotic deacetylase complex (MiDAC) serves as a key regulator of DNA end synapsis during NHEJ repair in mammalian cells. Mechanistically, MiDAC removes combinatorial acetyl marks on histone H2A (H2AK5acK9ac) around DSB-proximal chromatin, suppressing hyperaccumulation of bromodomain-containing protein BRD4 that would otherwise undergo liquid-liquid phase separation with KU80 and prevent the proper installation of LIG4-XRCC4-XLF onto DSB ends. This study provides mechanistic insight into the control of NHEJ synaptic complex assembly by a specific chromatin signature and highlights the critical role of H2A hypoacetylation in restraining unscheduled compartmentalization of DNA repair machinery., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
27. Alternative splicing decouples local from global PRC2 activity.
- Author
-
Arecco N, Mocavini I, Blanco E, Ballaré C, Libman E, Bonnal S, Irimia M, and Di Croce L
- Subjects
- Animals, Mice, Histones genetics, Histones metabolism, Chromatin genetics, Protein Isoforms genetics, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism, Alternative Splicing
- Abstract
The Polycomb repressive complex 2 (PRC2) mediates epigenetic maintenance of gene silencing in eukaryotes via methylation of histone H3 at lysine 27 (H3K27). Accessory factors define two distinct subtypes, PRC2.1 and PRC2.2, with different actions and chromatin-targeting mechanisms. The mechanisms orchestrating PRC2 assembly are not fully understood. Here, we report that alternative splicing (AS) of PRC2 core component SUZ12 generates an uncharacterized isoform SUZ12-S, which co-exists with the canonical SUZ12-L isoform in virtually all tissues and developmental stages. SUZ12-S drives PRC2.1 formation and favors PRC2 dimerization. While SUZ12-S is necessary and sufficient for the repression of target genes via promoter-proximal H3K27me3 deposition, SUZ12-L maintains global H3K27 methylation levels. Mouse embryonic stem cells (ESCs) lacking either isoform exit pluripotency more slowly and fail to acquire neuronal cell identity. Our findings reveal a physiological mechanism regulating PRC2 assembly and higher-order interactions in eutherians, with impacts on H3K27 methylation and gene repression., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
28. Acetyl-CoA production by Mediator-bound 2-ketoacid dehydrogenases boosts de novo histone acetylation and is regulated by nitric oxide.
- Author
-
Russo M, Gualdrini F, Vallelonga V, Prosperini E, Noberini R, Pedretti S, Borriero C, Di Chiaro P, Polletti S, Imperato G, Marenda M, Ghirardi C, Bedin F, Cuomo A, Rodighiero S, Bonaldi T, Mitro N, Ghisletti S, and Natoli G
- Subjects
- Acetyl Coenzyme A metabolism, Acetylation, Mediator Complex metabolism, Oxidoreductases metabolism, Histones genetics, Histones metabolism, Nitric Oxide metabolism
- Abstract
Histone-modifying enzymes depend on the availability of cofactors, with acetyl-coenzyme A (CoA) being required for histone acetyltransferase (HAT) activity. The discovery that mitochondrial acyl-CoA-producing enzymes translocate to the nucleus suggests that high concentrations of locally synthesized metabolites may impact acylation of histones and other nuclear substrates, thereby controlling gene expression. Here, we show that 2-ketoacid dehydrogenases are stably associated with the Mediator complex, thus providing a local supply of acetyl-CoA and increasing the generation of hyper-acetylated histone tails. Nitric oxide (NO), which is produced in large amounts in lipopolysaccharide-stimulated macrophages, inhibited the activity of Mediator-associated 2-ketoacid dehydrogenases. Elevation of NO levels and the disruption of Mediator complex integrity both affected de novo histone acetylation within a shared set of genomic regions. Our findings indicate that the local supply of acetyl-CoA generated by 2-ketoacid dehydrogenases bound to Mediator is required to maximize acetylation of histone tails at sites of elevated HAT activity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
29. Mechanisms of RNF168 nucleosome recognition and ubiquitylation.
- Author
-
Hu Q, Zhao D, Cui G, Bhandari J, Thompson JR, Botuyan MV, and Mer G
- Subjects
- Cryoelectron Microscopy, Ubiquitination, Histones metabolism, Chromatin genetics, DNA Repair, Ubiquitin metabolism, Tumor Suppressor p53-Binding Protein 1 genetics, DNA Damage, Nucleosomes genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
RNF168 plays a central role in the DNA damage response (DDR) by ubiquitylating histone H2A at K13 and K15. These modifications direct BRCA1-BARD1 and 53BP1 foci formation in chromatin, essential for cell-cycle-dependent DNA double-strand break (DSB) repair pathway selection. The mechanism by which RNF168 catalyzes the targeted accumulation of H2A ubiquitin conjugates to form repair foci around DSBs remains unclear. Here, using cryoelectron microscopy (cryo-EM), nuclear magnetic resonance (NMR) spectroscopy, and functional assays, we provide a molecular description of the reaction cycle and dynamics of RNF168 as it modifies the nucleosome and recognizes its ubiquitylation products. We demonstrate an interaction of a canonical ubiquitin-binding domain within full-length RNF168, which not only engages ubiquitin but also the nucleosome surface, clarifying how such site-specific ubiquitin recognition propels a signal amplification loop. Beyond offering mechanistic insights into a key DDR protein, our study aids in understanding site specificity in both generating and interpreting chromatin ubiquitylation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
30. Regulation of replicative histone RNA metabolism by the histone chaperone ASF1.
- Author
-
Mendiratta S, Ray-Gallet D, Lemaire S, Gatto A, Forest A, Kerlin MA, and Almouzni G
- Subjects
- Humans, Histone Chaperones genetics, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, DNA Replication, Molecular Chaperones genetics, Molecular Chaperones metabolism, RNA genetics, Histones genetics, Histones metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
In S phase, duplicating and assembling the whole genome into chromatin requires upregulation of replicative histone gene expression. Here, we explored how histone chaperones control histone production in human cells to ensure a proper link with chromatin assembly. Depletion of the ASF1 chaperone specifically decreases the pool of replicative histones both at the protein and RNA levels. The decrease in their overall expression, revealed by total RNA sequencing (RNA-seq), contrasted with the increase in nascent/newly synthesized RNAs observed by 4sU-labeled RNA-seq. Further inspection of replicative histone RNAs showed a 3' end processing defect with an increase of pre-mRNAs/unprocessed transcripts likely targeted to degradation. Collectively, these data argue for a production defect of replicative histone RNAs in ASF1-depleted cells. We discuss how this regulation of replicative histone RNA metabolism by ASF1 as a "chaperone checkpoint" fine-tunes the histone dosage to avoid unbalanced situations deleterious for cell survival., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
31. Poly(ADP-ribosyl)ation enhances nucleosome dynamics and organizes DNA damage repair components within biomolecular condensates.
- Author
-
Nosella ML, Kim TH, Huang SK, Harkness RW, Goncalves M, Pan A, Tereshchenko M, Vahidi S, Rubinstein JL, Lee HO, Forman-Kay JD, and Kay LE
- Subjects
- Poly(ADP-ribose) Polymerases metabolism, Cryoelectron Microscopy, Biomolecular Condensates, DNA Repair, Histones genetics, Histones metabolism, DNA genetics, DNA metabolism, DNA Damage, Poly (ADP-Ribose) Polymerase-1 metabolism, Nucleosomes genetics, Poly ADP Ribosylation genetics
- Abstract
Nucleosomes, the basic structural units of chromatin, hinder recruitment and activity of various DNA repair proteins, necessitating modifications that enhance DNA accessibility. Poly(ADP-ribosyl)ation (PARylation) of proteins near damage sites is an essential initiation step in several DNA-repair pathways; however, its effects on nucleosome structural dynamics and organization are unclear. Using NMR, cryoelectron microscopy (cryo-EM), and biochemical assays, we show that PARylation enhances motions of the histone H3 tail and DNA, leaving the configuration of the core intact while also stimulating nuclease digestion and ligation of nicked nucleosomal DNA by LIG3. PARylation disrupted interactions between nucleosomes, preventing self-association. Addition of LIG3 and XRCC1 to PARylated nucleosomes generated condensates that selectively partition DNA repair-associated proteins in a PAR- and phosphorylation-dependent manner in vitro. Our results establish that PARylation influences nucleosomes across different length scales, extending from the atom-level motions of histone tails to the mesoscale formation of condensates with selective compositions., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
32. p300 is an obligate integrator of combinatorial transcription factor inputs.
- Author
-
Ferrie JJ, Karr JP, Graham TGW, Dailey GM, Zhang G, Tjian R, and Darzacq X
- Subjects
- Gene Expression Regulation, Histones metabolism, Chromatin genetics, Transcription Factors genetics, Transcription Factors metabolism, Transcription, Genetic
- Abstract
Transcription coactivators are proteins or protein complexes that mediate transcription factor (TF) function. However, they lack DNA-binding capacity, prompting the question of how they engage target loci. Three non-exclusive hypotheses have been posited: coactivators are recruited by complexing with TFs, by binding histones through epigenetic reader domains, or by partitioning into condensates through their extensive intrinsically disordered regions. Using p300 as a prototypical coactivator, we systematically mutated its annotated domains and show by single-molecule tracking in live U2OS cells that coactivator-chromatin binding depends entirely on combinatorial binding of multiple TF-interaction domains. Furthermore, we demonstrate that acetyltransferase activity opposes p300-chromatin association and that the N-terminal TF-interaction domains regulate that activity. Single TF-interaction domains are insufficient for chromatin binding and regulation of catalytic activity, implying a principle that we speculate could broadly apply to eukaryotic gene regulation: a TF must act in coordination with other TFs to recruit coactivator activity., Competing Interests: Declaration of interests R.T. and X.D. are co-founders of Eikon Therapeutics., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
33. Human histone H1 variants impact splicing outcome by controlling RNA polymerase II elongation.
- Author
-
Pascal C, Zonszain J, Hameiri O, Gargi-Levi C, Lev-Maor G, Tammer L, Levy T, Tarabeih A, Roy VR, Ben-Salmon S, Elbaz L, Eid M, Hakim T, Abu Rabe'a S, Shalev N, Jordan A, Meshorer E, and Ast G
- Subjects
- Humans, RNA Splicing, Transcription, Genetic, Chromatin genetics, Alternative Splicing, Histones genetics, Histones metabolism, RNA Polymerase II genetics, RNA Polymerase II metabolism
- Abstract
Histones shape chromatin structure and the epigenetic landscape. H1, the most diverse histone in the human genome, has 11 variants. Due to the high structural similarity between the H1s, their unique functions in transferring information from the chromatin to mRNA-processing machineries have remained elusive. Here, we generated human cell lines lacking up to five H1 subtypes, allowing us to characterize the genomic binding profiles of six H1 variants. Most H1s bind to specific sites, and binding depends on multiple factors, including GC content. The highly expressed H1.2 has a high affinity for exons, whereas H1.3 binds intronic sequences. H1s are major splicing regulators, especially of exon skipping and intron retention events, through their effects on the elongation of RNA polymerase II (RNAPII). Thus, H1 variants determine splicing fate by modulating RNAPII elongation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
34. The cell-cycle choreography of H3 variants shapes the genome.
- Author
-
Delaney K, Weiss N, and Almouzni G
- Subjects
- Cell Cycle genetics, DNA, Histone Chaperones genetics, Histones genetics, Histones metabolism, Chromatin genetics
- Abstract
Histone variants provide versatility in the basic unit of chromatin, helping to define dynamic landscapes and cell fates. Maintaining genome integrity is paramount for the cell, and it is intimately linked with chromatin dynamics, assembly, and disassembly during DNA transactions such as replication, repair, recombination, and transcription. In this review, we focus on the family of H3 variants and their dynamics in space and time during the cell cycle. We review the distinct H3 variants' specific features along with their escort partners, the histone chaperones, compiled across different species to discuss their distinct importance considering evolution. We place H3 dynamics at different times during the cell cycle with the possible consequences for genome stability. Finally, we examine how their mutation and alteration impact disease. The emerging picture stresses key parameters in H3 dynamics to reflect on how when they are perturbed, they become a source of stress for genome integrity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
35. Crucial roles of the BRCA1-BARD1 E3 ubiquitin ligase activity in homology-directed DNA repair.
- Author
-
Wang M, Li W, Tomimatsu N, Yu CH, Ji JH, Alejo S, Witus SR, Alimbetov D, Fitzgerald O, Wu B, Wang Q, Huang Y, Gan Y, Dong F, Kwon Y, Sareddy GR, Curiel TJ, Habib AA, Hromas R, Dos Santos Passos C, Yao T, Ivanov DN, Brzovic PS, Burma S, Klevit RE, and Zhao W
- Subjects
- Humans, BRCA1 Protein metabolism, Ubiquitination, Histones genetics, Histones metabolism, Ubiquitin-Protein Ligases metabolism, Recombinational DNA Repair, DNA, DNA Repair, Tumor Suppressor Proteins metabolism, Neoplasms
- Abstract
The tumor-suppressor breast cancer 1 (BRCA1) in complex with BRCA1-associated really interesting new gene (RING) domain 1 (BARD1) is a RING-type ubiquitin E3 ligase that modifies nucleosomal histone and other substrates. The importance of BRCA1-BARD1 E3 activity in tumor suppression remains highly controversial, mainly stemming from studying mutant ligase-deficient BRCA1-BARD1 species that we show here still retain significant ligase activity. Using full-length BRCA1-BARD1, we establish robust BRCA1-BARD1-mediated ubiquitylation with specificity, uncover multiple modes of activity modulation, and construct a truly ligase-null variant and a variant specifically impaired in targeting nucleosomal histones. Cells expressing either of these BRCA1-BARD1 separation-of-function alleles are hypersensitive to DNA-damaging agents. Furthermore, we demonstrate that BRCA1-BARD1 ligase is not only required for DNA resection during homology-directed repair (HDR) but also contributes to later stages for HDR completion. Altogether, our findings reveal crucial, previously unrecognized roles of BRCA1-BARD1 ligase activity in genome repair via HDR, settle prior controversies regarding BRCA1-BARD1 ligase functions, and catalyze new efforts to uncover substrates related to tumor suppression., Competing Interests: Declaration of interests D.N.I. is a co-founder and a shareholder of E3 Bioscience LLC, a commercial entity that manufactures FRET-active E2∼Ub conjugates used in this study., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
36. Structural insight into H4K20 methylation on H2A.Z-nucleosome by SUV420H1.
- Author
-
Huang L, Wang Y, Long H, Zhu H, Wen Z, Zhang L, Zhang W, Guo Z, Wang L, Tang F, Hu J, Bao K, Zhu P, Li G, and Zhou Z
- Subjects
- Methylation, DNA metabolism, DNA Replication, Histones metabolism, Nucleosomes genetics
- Abstract
DNA replication ensures the accurate transmission of genetic information during the cell cycle. Histone variant H2A.Z is crucial for early replication origins licensing and activation in which SUV420H1 preferentially recognizes H2A.Z-nucleosome and deposits H4 lysine 20 dimethylation (H4K20me2) on replication origins. Here, we report the cryo-EM structures of SUV420H1 bound to H2A.Z-nucleosome or H2A-nucleosome and demonstrate that SUV420H1 directly interacts with H4 N-terminal tail, the DNA, and the acidic patch in the nucleosome. The H4 (1-24) forms a lasso-shaped structure that stabilizes the SUV420H1-nucleosome complex and precisely projects the H4K20 residue into the SUV420H1 catalytic center. In vitro and in vivo analyses reveal a crucial role of the SUV420H1 KR loop (residues 214-223), which lies close to the H2A.Z-specific residues D97/S98, in H2A.Z-nucleosome preferential recognition. Together, our findings elucidate how SUV420H1 recognizes nucleosomes to ensure site-specific H4K20me2 modification and provide insights into how SUV420H1 preferentially recognizes H2A.Z nucleosome., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
37. Distinct layers of BRD4-PTEFb reveal bromodomain-independent function in transcriptional regulation.
- Author
-
Zheng B, Gold S, Iwanaszko M, Howard BC, Wang L, and Shilatifard A
- Subjects
- Histones metabolism, Gene Expression Regulation, Chromatin genetics, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Nuclear Proteins metabolism, Transcription Factors metabolism
- Abstract
The BET family protein BRD4, which forms the CDK9-containing BRD4-PTEFb complex, is considered to be a master regulator of RNA polymerase II (Pol II) pause release. Because its tandem bromodomains interact with acetylated histone lysine residues, it has long been thought that BRD4 requires these bromodomains for its recruitment to chromatin and transcriptional regulatory function. Here, using rapid depletion and genetic complementation with domain deletion mutants, we demonstrate that BRD4 bromodomains are dispensable for Pol II pause release. A minimal, bromodomain-less C-terminal BRD4 fragment containing the PTEFb-interacting C-terminal motif (CTM) is instead both necessary and sufficient to mediate Pol II pause release in the absence of full-length BRD4. Although BRD4-PTEFb can associate with chromatin through acetyl recognition, our results indicate that a distinct, active BRD4-PTEFb population functions to regulate transcription independently of bromodomain-mediated chromatin association. These findings may enable more effective pharmaceutical modulation of BRD4-PTEFb activity., Competing Interests: Declaration of interests The authors declare no conflict of interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
38. Histone H3 lysine 27 crotonylation mediates gene transcriptional repression in chromatin.
- Author
-
Liu N, Konuma T, Sharma R, Wang D, Zhao N, Cao L, Ju Y, Liu D, Wang S, Bosch A, Sun Y, Zhang S, Ji D, Nagatoishi S, Suzuki N, Kikuchi M, Wakamori M, Zhao C, Ren C, Zhou TJ, Xu Y, Meslamani J, Fu S, Umehara T, Tsumoto K, Akashi S, Zeng L, Roeder RG, Walsh MJ, Zhang Q, and Zhou MM
- Subjects
- Mice, Animals, Lysine metabolism, Transcription Factors metabolism, Gene Expression Regulation, Acetylation, Chromatin genetics, Histones metabolism
- Abstract
Histone lysine acylation, including acetylation and crotonylation, plays a pivotal role in gene transcription in health and diseases. However, our understanding of histone lysine acylation has been limited to gene transcriptional activation. Here, we report that histone H3 lysine 27 crotonylation (H3K27cr) directs gene transcriptional repression rather than activation. Specifically, H3K27cr in chromatin is selectively recognized by the YEATS domain of GAS41 in complex with SIN3A-HDAC1 co-repressors. Proto-oncogenic transcription factor MYC recruits GAS41/SIN3A-HDAC1 complex to repress genes in chromatin, including cell-cycle inhibitor p21. GAS41 knockout or H3K27cr-binding depletion results in p21 de-repression, cell-cycle arrest, and tumor growth inhibition in mice, explaining a causal relationship between GAS41 and MYC gene amplification and p21 downregulation in colorectal cancer. Our study suggests that H3K27 crotonylation signifies a previously unrecognized, distinct chromatin state for gene transcriptional repression in contrast to H3K27 trimethylation for transcriptional silencing and H3K27 acetylation for transcriptional activation., Competing Interests: Declaration of interests M.-M.Z. is a scientific founder, director, and shareholder of Parkside Scientific Inc., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
39. LKB1 controls inflammatory potential through CRTC2-dependent histone acetylation.
- Author
-
Compton SE, Kitchen-Goosen SM, DeCamp LM, Lau KH, Mabvakure B, Vos M, Williams KS, Wong KK, Shi X, Rothbart SB, Krawczyk CM, and Jones RG
- Subjects
- Humans, Acetylation, Cytokines metabolism, Inflammation genetics, Transcription Factors genetics, Transcription Factors metabolism, Histones genetics, Histones metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism
- Abstract
Deregulated inflammation is a critical feature driving the progression of tumors harboring mutations in the liver kinase B1 (LKB1), yet the mechanisms linking LKB1 mutations to deregulated inflammation remain undefined. Here, we identify deregulated signaling by CREB-regulated transcription coactivator 2 (CRTC2) as an epigenetic driver of inflammatory potential downstream of LKB1 loss. We demonstrate that LKB1 mutations sensitize both transformed and non-transformed cells to diverse inflammatory stimuli, promoting heightened cytokine and chemokine production. LKB1 loss triggers elevated CRTC2-CREB signaling downstream of the salt-inducible kinases (SIKs), increasing inflammatory gene expression in LKB1-deficient cells. Mechanistically, CRTC2 cooperates with the histone acetyltransferases CBP/p300 to deposit histone acetylation marks associated with active transcription (i.e., H3K27ac) at inflammatory gene loci, promoting cytokine expression. Together, our data reveal a previously undefined anti-inflammatory program, regulated by LKB1 and reinforced through CRTC2-dependent histone modification signaling, that links metabolic and epigenetic states to cell-intrinsic inflammatory potential., Competing Interests: Declaration of interests R.G.J. is a scientific advisor for Agios Pharmaceuticals and Servier Pharmaceuticals and is a member of the Scientific Advisory Board of Immunomet Therapeutics., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
40. The molecular basis of heterochromatin assembly and epigenetic inheritance.
- Author
-
Grewal SIS
- Subjects
- Histones genetics, Histones metabolism, Heterochromatin genetics, Heterochromatin metabolism, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Epigenesis, Genetic, Schizosaccharomyces pombe Proteins genetics, Schizosaccharomyces genetics
- Abstract
Heterochromatin plays a fundamental role in gene regulation, genome integrity, and silencing of repetitive DNA elements. Histone modifications are essential for the establishment of heterochromatin domains, which is initiated by the recruitment of histone-modifying enzymes to nucleation sites. This leads to the deposition of histone H3 lysine-9 methylation (H3K9me), which provides the foundation for building high-concentration territories of heterochromatin proteins and the spread of heterochromatin across extended domains. Moreover, heterochromatin can be epigenetically inherited during cell division in a self-templating manner. This involves a "read-write" mechanism where pre-existing modified histones, such as tri-methylated H3K9 (H3K9me3), support chromatin association of the histone methyltransferase to promote further deposition of H3K9me. Recent studies suggest that a critical density of H3K9me3 and its associated factors is necessary for the propagation of heterochromatin domains across multiple generations. In this review, I discuss the key experiments that have highlighted the importance of modified histones for epigenetic inheritance., Competing Interests: Declaration of interests The author is a member of the Molecular Cell advisory board., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
41. Prometheus unshackled: Liver regeneration makes you young.
- Author
-
Pollina EA and Greer EL
- Subjects
- Liver metabolism, Protein Processing, Post-Translational, Histones genetics, Histones metabolism, Liver Regeneration genetics
- Abstract
In this issue of Molecular Cell, Yang and colleagues
1 discover age-dependent increases in broad regions of the repressive histone modification H3K27me3. They also demonstrate partial reversion to younger H3K27me3 patterns and gene expression upon resection of older livers., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
42. Modular antibodies reveal DNA damage-induced mono-ADP-ribosylation as a second wave of PARP1 signaling.
- Author
-
Longarini EJ, Dauben H, Locatelli C, Wondisford AR, Smith R, Muench C, Kolvenbach A, Lynskey ML, Pope A, Bonfiglio JJ, Jurado EP, Fajka-Boja R, Colby T, Schuller M, Ahel I, Timinszky G, O'Sullivan RJ, Huet S, and Matic I
- Subjects
- Poly (ADP-Ribose) Polymerase-1 genetics, Poly (ADP-Ribose) Polymerase-1 metabolism, Chromatin, DNA Damage, Antibodies genetics, Signal Transduction, Histones genetics, Histones metabolism, ADP-Ribosylation
- Abstract
PARP1, an established anti-cancer target that regulates many cellular pathways, including DNA repair signaling, has been intensely studied for decades as a poly(ADP-ribosyl)transferase. Although recent studies have revealed the prevalence of mono-ADP-ribosylation upon DNA damage, it was unknown whether this signal plays an active role in the cell or is just a byproduct of poly-ADP-ribosylation. By engineering SpyTag-based modular antibodies for sensitive and flexible detection of mono-ADP-ribosylation, including fluorescence-based sensors for live-cell imaging, we demonstrate that serine mono-ADP-ribosylation constitutes a second wave of PARP1 signaling shaped by the cellular HPF1/PARP1 ratio. Multilevel chromatin proteomics reveals histone mono-ADP-ribosylation readers, including RNF114, a ubiquitin ligase recruited to DNA lesions through a zinc-finger domain, modulating the DNA damage response and telomere maintenance. Our work provides a technological framework for illuminating ADP-ribosylation in a wide range of applications and biological contexts and establishes mono-ADP-ribosylation by HPF1/PARP1 as an important information carrier for cell signaling., Competing Interests: Declaration of interests E.J.L., H.D., J.J.B., T.C., and I.M. declare the following competing financial interests: Max-Planck-Innovation, the technology transfer center of the Max Planck Society, has licensed the antibodies AbD33204, AbD33205, AbD33644, AbD34251, AbD33641, and AbD43647 to Bio-Rad Laboratories., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
43. A hyper-quiescent chromatin state formed during aging is reversed by regeneration.
- Author
-
Yang N, Occean JR, Melters DP, Shi C, Wang L, Stransky S, Doyle ME, Cui CY, Delannoy M, Fan J, Slama E, Egan JM, De S, Cunningham SC, de Cabo R, Sidoli S, Dalal Y, and Sen P
- Subjects
- Mice, Animals, Epigenesis, Genetic, Aging genetics, Transcription Factors metabolism, Chromatin genetics, Histones genetics, Histones metabolism
- Abstract
Epigenetic alterations are a key hallmark of aging but have been limitedly explored in tissues. Here, using naturally aged murine liver as a model and extending to other quiescent tissues, we find that aging is driven by temporal chromatin alterations that promote a refractory cellular state and compromise cellular identity. Using an integrated multi-omics approach and the first direct visualization of aged chromatin, we find that globally, old cells show H3K27me3-driven broad heterochromatinization and transcriptional suppression. At the local level, site-specific loss of H3K27me3 over promoters of genes encoding developmental transcription factors leads to expression of otherwise non-hepatocyte markers. Interestingly, liver regeneration reverses H3K27me3 patterns and rejuvenates multiple molecular and physiological aspects of the aged liver., Competing Interests: Declaration of interests The authors declare no competing interests., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
44. PRC2.1- and PRC2.2-specific accessory proteins drive recruitment of different forms of canonical PRC1.
- Author
-
Glancy E, Wang C, Tuck E, Healy E, Amato S, Neikes HK, Mariani A, Mucha M, Vermeulen M, Pasini D, and Bracken AP
- Subjects
- Polycomb-Group Proteins genetics, Polycomb-Group Proteins metabolism, Polycomb Repressive Complex 1 genetics, Polycomb Repressive Complex 1 metabolism, Chromatin genetics, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism, Histones genetics, Histones metabolism
- Abstract
Polycomb repressive complex 2 (PRC2) mediates H3K27me3 deposition, which is thought to recruit canonical PRC1 (cPRC1) via chromodomain-containing CBX proteins to promote stable repression of developmental genes. PRC2 forms two major subcomplexes, PRC2.1 and PRC2.2, but their specific roles remain unclear. Through genetic knockout (KO) and replacement of PRC2 subcomplex-specific subunits in naïve and primed pluripotent cells, we uncover distinct roles for PRC2.1 and PRC2.2 in mediating the recruitment of different forms of cPRC1. PRC2.1 catalyzes the majority of H3K27me3 at Polycomb target genes and is sufficient to promote recruitment of CBX2/4-cPRC1 but not CBX7-cPRC1. Conversely, while PRC2.2 is poor at catalyzing H3K27me3, we find that its accessory protein JARID2 is essential for recruitment of CBX7-cPRC1 and the consequent 3D chromatin interactions at Polycomb target genes. We therefore define distinct contributions of PRC2.1- and PRC2.2-specific accessory proteins to Polycomb-mediated repression and uncover a new mechanism for cPRC1 recruitment., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
45. NSD1 deposits histone H3 lysine 36 dimethylation to pattern non-CG DNA methylation in neurons.
- Author
-
Hamagami N, Wu DY, Clemens AW, Nettles SA, Li A, and Gabel HW
- Subjects
- Animals, Mice, Lysine metabolism, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Neurons metabolism, Histones genetics, Histones metabolism, DNA Methylation
- Abstract
During postnatal development, the DNA methyltransferase DNMT3A deposits high levels of non-CG cytosine methylation in neurons. This methylation is critical for transcriptional regulation, and loss of this mark is implicated in DNMT3A-associated neurodevelopmental disorders (NDDs). Here, we show in mice that genome topology and gene expression converge to shape histone H3 lysine 36 dimethylation (H3K36me2) profiles, which in turn recruit DNMT3A and pattern neuronal non-CG methylation. We show that NSD1, an H3K36 methyltransferase mutated in NDD, is required for the patterning of megabase-scale H3K36me2 and non-CG methylation in neurons. We find that brain-specific deletion of NSD1 causes altered DNA methylation that overlaps with DNMT3A disorder models to drive convergent dysregulation of key neuronal genes that may underlie shared phenotypes in NSD1- and DNMT3A-associated NDDs. Our findings indicate that H3K36me2 deposited by NSD1 is important for neuronal non-CG DNA methylation and suggest that the H3K36me2-DNMT3A-non-CG-methylation pathway is likely disrupted in NSD1-associated NDDs., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
46. Basic helix-loop-helix pioneer factors interact with the histone octamer to invade nucleosomes and generate nucleosome-depleted regions.
- Author
-
Donovan BT, Chen H, Eek P, Meng Z, Jipa C, Tan S, Bai L, and Poirier MG
- Subjects
- Histones metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Chromatin metabolism, Transcription Factors genetics, Transcription Factors metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Nucleosomes genetics, Nucleosomes metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Nucleosomes drastically limit transcription factor (TF) occupancy, while pioneer transcription factors (PFs) somehow circumvent this nucleosome barrier. In this study, we compare nucleosome binding of two conserved S. cerevisiae basic helix-loop-helix (bHLH) TFs, Cbf1 and Pho4. A cryo-EM structure of Cbf1 in complex with the nucleosome reveals that the Cbf1 HLH region can electrostatically interact with exposed histone residues within a partially unwrapped nucleosome. Single-molecule fluorescence studies show that the Cbf1 HLH region facilitates efficient nucleosome invasion by slowing its dissociation rate relative to DNA through interactions with histones, whereas the Pho4 HLH region does not. In vivo studies show that this enhanced binding provided by the Cbf1 HLH region enables nucleosome invasion and ensuing repositioning. These structural, single-molecule, and in vivo studies reveal the mechanistic basis of dissociation rate compensation by PFs and how this translates to facilitating chromatin opening inside cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
47. NPAS4 juggles neuronal activity-dependent transcription and DSB repair with NuA4.
- Author
-
Delint-Ramirez I and Madabhushi R
- Subjects
- DNA Breaks, Double-Stranded, Transcription Factors genetics, Transcription Factors metabolism, DNA Repair, Histones metabolism, Basic Helix-Loop-Helix Transcription Factors, Histone Acetyltransferases
- Abstract
In a recent study, Pollina et al.
1 discover a new neuron-specific NuA4-TIP60 chromatin remodeling complex that facilitates the repair of activity-induced DNA double-strand breaks (DSBs) in neurons and protects against mutations that accumulate with age and early death., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
48. Histone phosphorylation integrates the hepatic glucagon-PKA-CREB gluconeogenesis program in response to fasting.
- Author
-
Zhao Y, Li S, Chen Y, Wang Y, Wei Y, Zhou T, Zhang Y, Yang Y, Chen L, Liu Y, Hu C, Zhou B, and Ding Q
- Subjects
- Animals, Mice, Histones metabolism, Phosphorylation, 14-3-3 Proteins metabolism, Liver metabolism, Fasting metabolism, Cyclic AMP Response Element-Binding Protein genetics, Cyclic AMP Response Element-Binding Protein metabolism, Gluconeogenesis genetics, Glucagon metabolism
- Abstract
The glucagon-PKA signal is generally believed to control hepatic gluconeogenesis via the CREB transcription factor. Here we uncovered a distinct function of this signal in directly stimulating histone phosphorylation for gluconeogenic gene regulation in mice. In the fasting state, CREB recruited activated PKA to regions near gluconeogenic genes, where PKA phosphorylated histone H3 serine 28 (H3S28ph). H3S28ph, recognized by 14-3-3ζ, promoted recruitment of RNA polymerase II and transcriptional stimulation of gluconeogenic genes. In contrast, in the fed state, more PP2A was found near gluconeogenic genes, which counteracted PKA by dephosphorylating H3S28ph and repressing transcription. Importantly, ectopic expression of phosphomimic H3S28 efficiently restored gluconeogenic gene expression when liver PKA or CREB was depleted. These results together highlight a different functional scheme in regulating gluconeogenesis by the glucagon-PKA-CREB-H3S28ph cascade, in which the hormone signal is transmitted to chromatin for rapid and efficient gluconeogenic gene activation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
49. Histone chaperones: A multinodal highway network inside the cell.
- Author
-
Li Z and Zhang Z
- Subjects
- Nucleosomes genetics, Molecular Chaperones genetics, DNA Replication, Histone Chaperones genetics, Histone Chaperones metabolism, Histones genetics, Histones metabolism
- Abstract
Histone chaperones participate in the biogenesis, transportation, and deposition of histones. They contribute to processes impacted by nucleosomes including DNA replication, transcription, and epigenetic inheritance. In this issue, Carraro et al.
1 reveal an interconnected chaperone network and a surprising function of histone chaperone DAXX in de novo deposition of H3.3K9me3., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
50. DAXX adds a de novo H3.3K9me3 deposition pathway to the histone chaperone network.
- Author
-
Carraro M, Hendriks IA, Hammond CM, Solis-Mezarino V, Völker-Albert M, Elsborg JD, Weisser MB, Spanos C, Montoya G, Rappsilber J, Imhof A, Nielsen ML, and Groth A
- Subjects
- Humans, Nucleosomes genetics, Cell Cycle Proteins metabolism, DNA, Molecular Chaperones genetics, Molecular Chaperones metabolism, Co-Repressor Proteins genetics, Co-Repressor Proteins metabolism, Histones metabolism, Histone Chaperones genetics, Histone Chaperones metabolism
- Abstract
A multitude of histone chaperones are required to support histones from their biosynthesis until DNA deposition. They cooperate through the formation of histone co-chaperone complexes, but the crosstalk between nucleosome assembly pathways remains enigmatic. Using exploratory interactomics, we define the interplay between human histone H3-H4 chaperones in the histone chaperone network. We identify previously uncharacterized histone-dependent complexes and predict the structure of the ASF1 and SPT2 co-chaperone complex, expanding the role of ASF1 in histone dynamics. We show that DAXX provides a unique functionality to the histone chaperone network, recruiting histone methyltransferases to promote H3K9me3 catalysis on new histone H3.3-H4 prior to deposition onto DNA. Hereby, DAXX provides a molecular mechanism for de novo H3K9me3 deposition and heterochromatin assembly. Collectively, our findings provide a framework for understanding how cells orchestrate histone supply and employ targeted deposition of modified histones to underpin specialized chromatin states., Competing Interests: Declaration of interests C.M.H. and A.G. are inventors on a patent covering the therapeutic targeting of TONSL for cancer therapy. A.G. is co-founder and chief scientific officer of Ankrin Therapeutics. A.G. is a member of Molecular Cell’s Scientific Advisory Board. A.I. and M.V.-A. are cofounders of EpiQMAx. G.M. is co-founder and member of the board of directors of Twelve Bio and is a member of the Scientific Advisory Board at Ensoma., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.