Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(18): eadj8042, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38691608

ABSTRACT

Overactivation of the transforming growth factor-ß (TGFß) signaling in Duchenne muscular dystrophy (DMD) is a major hallmark of disease progression, leading to fibrosis and muscle dysfunction. Here, we investigated the role of SETDB1 (SET domain, bifurcated 1), a histone lysine methyltransferase involved in muscle differentiation. Our data show that, following TGFß induction, SETDB1 accumulates in the nuclei of healthy myotubes while being already present in the nuclei of DMD myotubes where TGFß signaling is constitutively activated. Transcriptomics revealed that depletion of SETDB1 in DMD myotubes leads to down-regulation of TGFß target genes coding for secreted factors involved in extracellular matrix remodeling and inflammation. Consequently, SETDB1 silencing in DMD myotubes abrogates the deleterious effect of their secretome on myoblast differentiation by impairing myoblast pro-fibrotic response. Our findings indicate that SETDB1 potentiates the TGFß-driven fibrotic response in DMD muscles, providing an additional axis for therapeutic intervention.


Subject(s)
Histone-Lysine N-Methyltransferase , Muscle Fibers, Skeletal , Muscular Dystrophy, Duchenne , Signal Transduction , Transforming Growth Factor beta , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Transforming Growth Factor beta/metabolism , Humans , Animals , Cell Differentiation , Mice , Myoblasts/metabolism , Fibrosis , Gene Expression Regulation
2.
Nat Struct Mol Biol ; 27(11): 1057-1068, 2020 11.
Article in English | MEDLINE | ID: mdl-32895554

ABSTRACT

Nucleosomes represent a challenge in regard to transcription. Histone eviction enables RNA polymerase II (RNAPII) progression through DNA, but compromises chromatin integrity. Here, we used the SNAP-tag system to distinguish new and old histones and monitor chromatin reassembly coupled to transcription in human cells. We uncovered a transcription-dependent loss of old histone variants H3.1 and H3.3. At transcriptionally active domains, H3.3 enrichment reflected both old H3.3 retention and new deposition. Mechanistically, we found that the histone regulator A (HIRA) chaperone is critical to processing both new and old H3.3 via different pathways. De novo H3.3 deposition is totally dependent on HIRA trimerization as well as on its partner ubinuclein 1 (UBN1), while antisilencing function 1 (ASF1) interaction with HIRA can be bypassed. By contrast, recycling of H3.3 requires HIRA but proceeds independently of UBN1 or HIRA trimerization and shows absolute dependency on ASF1-HIRA interaction. We propose a model whereby HIRA coordinates these distinct pathways during transcription to fine-tune chromatin states.


Subject(s)
Cell Cycle Proteins/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic , HeLa Cells , Histones/genetics , Humans , Nuclear Proteins/metabolism , Protein Multimerization
3.
Nat Commun ; 11(1): 1256, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152320

ABSTRACT

Vertebrates exhibit specific requirements for replicative H3 and non-replicative H3.3 variants during development. To disentangle whether this involves distinct modes of deposition or unique functions once incorporated into chromatin, we combined studies in Xenopus early development with chromatin assays. Here we investigate the extent to which H3.3 mutated at residues that differ from H3.2 rescue developmental defects caused by H3.3 depletion. Regardless of the deposition pathway, only variants at residue 31-a serine that can become phosphorylated-failed to rescue endogenous H3.3 depletion. Although an alanine substitution fails to rescue H3.3 depletion, a phospho-mimic aspartate residue at position 31 rescues H3.3 function. To explore mechanisms involving H3.3 S31 phosphorylation, we identified factors attracted or repulsed by the presence of aspartate at position 31, along with modifications on neighboring residues. We propose that serine 31-phosphorylated H3.3 acts as a signaling module that stimulates the acetylation of K27, providing a chromatin state permissive to the embryonic development program.


Subject(s)
Gastrulation/physiology , Histones/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , Amino Acid Sequence , Animals , Chromatin , Histones/genetics , Phosphorylation , Sequence Analysis, Protein , Serine
4.
Cell Chem Biol ; 26(11): 1573-1585.e10, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31543461

ABSTRACT

Anti-silencing function 1 (ASF1) is a conserved H3-H4 histone chaperone involved in histone dynamics during replication, transcription, and DNA repair. Overexpressed in proliferating tissues including many tumors, ASF1 has emerged as a promising therapeutic target. Here, we combine structural, computational, and biochemical approaches to design peptides that inhibit the ASF1-histone interaction. Starting from the structure of the human ASF1-histone complex, we developed a rational design strategy combining epitope tethering and optimization of interface contacts to identify a potent peptide inhibitor with a dissociation constant of 3 nM. When introduced into cultured cells, the inhibitors impair cell proliferation, perturb cell-cycle progression, and reduce cell migration and invasion in a manner commensurate with their affinity for ASF1. Finally, we find that direct injection of the most potent ASF1 peptide inhibitor in mouse allografts reduces tumor growth. Our results open new avenues to use ASF1 inhibitors as promising leads for cancer therapy.


Subject(s)
Cell Cycle Proteins/metabolism , Drug Design , Molecular Chaperones/metabolism , Peptides/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epitopes/chemistry , Epitopes/metabolism , Female , Histones/chemistry , Histones/metabolism , Humans , Kinetics , Mice , Mice, Inbred BALB C , Molecular Chaperones/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , Peptides/metabolism , Peptides/pharmacology , Peptides/therapeutic use , Thermodynamics , Transplantation, Homologous
5.
Nat Commun ; 9(1): 3181, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30093638

ABSTRACT

DNA replication is a challenge for the faithful transmission of parental information to daughter cells, as both DNA and chromatin organization must be duplicated. Replication stress further complicates the safeguard of epigenome integrity. Here, we investigate the transmission of the histone variants H3.3 and H3.1 during replication. We follow their distribution relative to replication timing, first in the genome and, second, in 3D using super-resolution microscopy. We find that H3.3 and H3.1 mark early- and late-replicating chromatin, respectively. In the nucleus, H3.3 forms domains, which decrease in density throughout replication, while H3.1 domains increase in density. Hydroxyurea impairs local recycling of parental histones at replication sites. Similarly, depleting the histone chaperone ASF1 affects recycling, leading to an impaired histone variant landscape. We discuss how faithful transmission of histone variants involves ASF1 and can be impacted by replication stress, with ensuing consequences for cell fate and tumorigenesis.


Subject(s)
Cell Cycle Proteins/chemistry , Chromatin/chemistry , DNA Replication , Histones/chemistry , Cell Lineage , DNA/chemistry , Epigenesis, Genetic , Genome, Human , HeLa Cells , Humans , Hydroxyurea/chemistry , Microscopy , Microscopy, Fluorescence , Molecular Chaperones , Nucleosomes/chemistry , S Phase
6.
Nat Commun ; 9(1): 3103, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30082790

ABSTRACT

The HIRA histone chaperone complex deposits the histone variant H3.3 onto chromatin in a DNA synthesis-independent manner. It comprises three identified subunits, HIRA, UBN1 and CABIN1, however the functional oligomerization state of the complex has not been investigated. Here we use biochemical and crystallographic analysis to show that the HIRA subunit forms a stable homotrimer that binds two subunits of CABIN1 in vitro. A HIRA mutant that is defective in homotrimer formation interacts less efficiently with CABIN1, is not enriched at DNA damage sites upon UV irradiation and cannot rescue new H3.3 deposition in HIRA knockout cells. The structural homology with the homotrimeric replisome component Ctf4/AND-1 enables the drawing of parallels and discussion of the functional importance of the homotrimerization state of the HIRA subunit.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Cell Cycle Proteins/chemistry , DNA/chemistry , Histone Chaperones/chemistry , Histones/chemistry , Molecular Chaperones/chemistry , Nuclear Proteins/chemistry , Transcription Factors/chemistry , Binding Sites , Cell Line, Tumor , Chromatin/chemistry , Crystallography, X-Ray , DNA Damage , Databases, Protein , Green Fluorescent Proteins/chemistry , HeLa Cells , Humans , Plasmids , Protein Binding , Protein Conformation , Ultraviolet Rays
7.
J Vis Exp ; (111)2016 05 17.
Article in English | MEDLINE | ID: mdl-27286495

ABSTRACT

Skeletal muscle terminal differentiation starts with the commitment of pluripotent mesodermal precursor cells to myoblasts. These cells have still the ability to proliferate or they can differentiate and fuse into multinucleated myotubes, which maturate further to form myofibers. Skeletal muscle terminal differentiation is orchestrated by the coordinated action of various transcription factors, in particular the members of the Muscle Regulatory Factors or MRFs (MyoD, Myogenin, Myf5, and MRF4), also called the myogenic bHLH transcription factors family. These factors cooperate with chromatin-remodeling complexes within elaborate transcriptional regulatory network to achieve skeletal myogenesis. In this, MyoD is considered the master myogenic transcription factor in triggering muscle terminal differentiation. This notion is strengthened by the ability of MyoD to convert non-muscle cells into skeletal muscle cells. Here we describe an approach used to identify MyoD protein partners in an exhaustive manner in order to elucidate the different factors involved in skeletal muscle terminal differentiation. The long-term aim is to understand the epigenetic mechanisms involved in the regulation of skeletal muscle genes, i.e., MyoD targets. MyoD partners are identified by using Tandem Affinity Purification (TAP-Tag) from a heterologous system coupled to mass spectrometry (MS) characterization, followed by validation of the role of relevant partners during skeletal muscle terminal differentiation. Aberrant forms of myogenic factors, or their aberrant regulation, are associated with a number of muscle disorders: congenital myasthenia, myotonic dystrophy, rhabdomyosarcoma and defects in muscle regeneration. As such, myogenic factors provide a pool of potential therapeutic targets in muscle disorders, both with regard to mechanisms that cause disease itself and regenerative mechanisms that can improve disease treatment. Thus, the detailed understanding of the intermolecular interactions and the genetic programs controlled by the myogenic factors is essential for the rational design of efficient therapies.


Subject(s)
Mass Spectrometry , Muscle, Skeletal/metabolism , MyoD Protein/analysis , Cell Differentiation , Humans , Muscle, Skeletal/cytology , Myogenin , Transcription Factors
8.
J Mol Biol ; 428(11): 2329-2343, 2016 06 05.
Article in English | MEDLINE | ID: mdl-27056598

ABSTRACT

Lysine methyltransferases G9a and GLP (G9a-like protein) are highly homologous and form functional heterodimeric complexes that establish mono- and dimethylation on histone H3 lysine 9 (H3K9me1, H3K9me2) in euchromatin. Here, we describe unexpected distinct roles for G9a and GLP in skeletal muscle terminal differentiation. Indeed, gain- or loss-of-function assays in myoblasts showed, in agreement with previous reports, that G9a inhibits terminal differentiation. While GLP plays a more intricate role in muscle differentiation,in one hand, both GLP gain and loss of function inhibit late steps of differentiation; on the other hand, in contrast to G9a, GLP overexpression promotes abnormal precocious expression of muscle differentiation-specific genes already in proliferating myoblasts. In agreement, transcriptomic analysis indicates that G9a and GLP regulate different sets of genes. Thus, GLP, but not G9a, inhibits proteasome subunit-encoding genes expression, resulting in an inhibition of the proteasome activities. Subsequently, GLP, but not G9a, overexpression stabilizes MyoD that is likely to be responsible for muscle markers expression in proliferating myoblasts.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Myoblasts/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Gene Expression/physiology , Gene Expression Profiling/methods , Lysine/metabolism , Methylation , Mice , Muscle Development/physiology
9.
Nat Rev Mol Cell Biol ; 16(8): 499-513, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26204160

ABSTRACT

The methylation of histone Lys residues by Lys methyltransferases (KMTs) regulates chromatin organization and either activates or represses gene expression, depending on the residue that is targeted. KMTs are emerging as key components in several cellular processes, and their deregulation is often associated with pathogenesis. Here, we review the current knowledge on the main KMTs that are associated with gene silencing: namely, those responsible for methylating histone H3 Lys 9 (H3K9), H3K27 and H4K20. We discuss their biochemical properties and the various mechanisms by which they are targeted to the chromatin and regulate gene expression, as well as new data on the interplay between them and other chromatin modifiers.


Subject(s)
Gene Silencing , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Protein Processing, Post-Translational , Animals , Chromatin Assembly and Disassembly , Humans , Methylation
10.
J Cell Sci ; 127(Pt 15): 3347-59, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24906798

ABSTRACT

Correct chromosome segregation requires a unique chromatin environment at centromeres and in their vicinity. Here, we address how the deposition of canonical H2A and H2A.Z histone variants is controlled at pericentric heterochromatin (PHC). Whereas in euchromatin newly synthesized H2A and H2A.Z are deposited throughout the cell cycle, we reveal two discrete waves of deposition at PHC - during mid to late S phase in a replication-dependent manner for H2A and during G1 phase for H2A.Z. This G1 cell cycle restriction is lost when heterochromatin features are altered, leading to the accumulation of H2A.Z at the domain. Interestingly, compromising PHC integrity also impacts upon neighboring centric chromatin, increasing the amount of centromeric CENP-A without changing the timing of its deposition. We conclude that the higher-order chromatin structure at the pericentric domain influences dynamics at the nucleosomal level within centromeric chromatin. The two different modes of rearrangement of the PHC during the cell cycle provide distinct opportunities to replenish one or the other H2A variant, highlighting PHC integrity as a potential signal to regulate the deposition timing and stoichiometry of histone variants at the centromere.


Subject(s)
Cell Cycle , Histones/metabolism , 3T3 Cells , Animals , Autoantigens/genetics , Autoantigens/metabolism , Centromere/genetics , Centromere Protein A , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication , Heterochromatin , Histones/genetics , Mice , Multiprotein Complexes/metabolism
11.
Curr Opin Cell Biol ; 23(3): 266-76, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21470840

ABSTRACT

Classical heterochromatin chromosomal landmarks, such as centromeres and telomeres, are characterized by specific chromatin signatures. Among these, the incorporation of histone variants has recently emerged as an important feature. Using the centromere as a paradigm, we consider the role of histone variant dynamics in locus-specific chromatin organization. We describe the distinct location and dynamics of CenH3, H3.3, and H2AZ at the centromere during the cell cycle. This leads us to present the current view concerning modes of incorporation at this chromosomal landmark. Finally, we highlight the importance of histone variants in the crosstalk between centric and pericentric domains for maintaining centromere identity.


Subject(s)
Centromere/metabolism , Chromosomes/metabolism , Histones/metabolism , Animals , Cell Cycle , Centromere/chemistry , Chromosomes/chemistry , Heterochromatin , Histones/chemistry , Histones/genetics , Humans
12.
J Cell Biol ; 192(4): 569-82, 2011 Feb 21.
Article in English | MEDLINE | ID: mdl-21321101

ABSTRACT

Centromeric protein A (CENP-A) is the epigenetic mark of centromeres. CENP-A replenishment is necessary in each cell cycle to compensate for the dilution associated to DNA replication, but how this is achieved mechanistically is largely unknown. We have developed an assay using Xenopus egg extracts that can recapitulate the spatial and temporal specificity of CENP-A deposition observed in human cells, providing us with a robust in vitro system amenable to molecular dissection. Here we show that this deposition depends on Xenopus Holliday junction-recognizing protein (xHJURP), a member of the HJURP/Scm3 family recently identified in yeast and human cells, further supporting the essential role of these chaperones in CENP-A loading. Despite little sequence homology, human HJURP can substitute for xHJURP. We also report that condensin II, but not condensin I, is required for CENP-A assembly and contributes to retention of centromeric CENP-A nucleosomes both in mitosis and interphase. We propose that the chromatin structure imposed by condensin II at centromeres enables CENP-A incorporation initiated by xHJURP.


Subject(s)
Adenosine Triphosphatases/physiology , Autoantigens/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/physiology , Multiprotein Complexes/physiology , Xenopus Proteins/physiology , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Centromere/metabolism , Centromere/ultrastructure , Centromere Protein A , Chromatin/metabolism , Chromatin/ultrastructure , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Histone Chaperones/metabolism , Histone Chaperones/physiology , Humans , Interphase , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...