Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.667
Filtrer
1.
Nat Commun ; 15(1): 7152, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39169041

RÉSUMÉ

For accurate mitotic cell division, replicated chromatin must be assembled into chromosomes and faithfully segregated into daughter cells. While protein factors like condensin play key roles in this process, it is unclear how chromosome assembly proceeds as molecular events of nucleosomes in living cells and how condensins act on nucleosomes to organize chromosomes. To approach these questions, we investigate nucleosome behavior during mitosis of living human cells using single-nucleosome tracking, combined with rapid-protein depletion technology and computational modeling. Our results show that local nucleosome motion becomes increasingly constrained during mitotic chromosome assembly, which is functionally distinct from condensed apoptotic chromatin. Condensins act as molecular crosslinkers, locally constraining nucleosomes to organize chromosomes. Additionally, nucleosome-nucleosome interactions via histone tails constrain and compact whole chromosomes. Our findings elucidate the physical nature of the chromosome assembly process during mitosis.


Sujet(s)
Adenosine triphosphatases , Chromatine , Protéines de liaison à l'ADN , Mitose , Complexes multiprotéiques , Nucléosomes , Humains , Nucléosomes/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Adenosine triphosphatases/métabolisme , Adenosine triphosphatases/génétique , Complexes multiprotéiques/métabolisme , Chromatine/métabolisme , Histone/métabolisme , Cellules HeLa , Chromosomes humains/métabolisme , Chromosomes humains/génétique , Chromosomes/métabolisme
2.
Proc Natl Acad Sci U S A ; 121(35): e2320804121, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39172790

RÉSUMÉ

Breast Cancer Type 1 Susceptibility Protein (BRCA1) is a tumor-suppressor protein that regulates various cellular pathways, including those that are essential for preserving genome stability. One essential mechanism involves a BRCA1-A complex that is recruited to double-strand breaks (DSBs) by RAP80 before initiating DNA damage repair (DDR). How RAP80 itself is recruited to DNA damage sites, however, is unclear. Here, we demonstrate an intrinsic correlation between a methyltransferase DOT1L-mediated RAP80 methylation and BRCA1-A complex chromatin recruitment that occurs during cancer cell radiotherapy resistance. Mechanistically, DOT1L is quickly recruited onto chromatin and methylates RAP80 at multiple lysines in response to DNA damage. Methylated RAP80 is then indispensable for binding to ubiquitinated H2A and subsequently triggering BRCA1-A complex recruitment onto DSBs. Importantly, DOT1L-catalyzed RAP80 methylation and recruitment of BRCA1 have clinical relevance, as inhibition of DOT1L or RAP80 methylation seems to enhance the radiosensitivity of cancer cells both in vivo and in vitro. These data reveal a crucial role for DOT1L in DDR through initiating recruitment of RAP80 and BRCA1 onto chromatin and underscore a therapeutic strategy based on targeting DOT1L to overcome tumor radiotherapy resistance.


Sujet(s)
Protéine BRCA1 , Réparation de l'ADN , Chaperons d'histones , Histone-lysine N-methyltransferase , Humains , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Chaperons d'histones/métabolisme , Chaperons d'histones/génétique , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Méthylation , Lignée cellulaire tumorale , Cassures double-brin de l'ADN , Chromatine/métabolisme , Methyltransferases/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Animaux , Femelle , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Souris , Radiotolérance/génétique , Méthylation de l'ADN
3.
Elife ; 132024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39177021

RÉSUMÉ

MYC family oncoproteins regulate the expression of a large number of genes and broadly stimulate elongation by RNA polymerase II (RNAPII). While the factors that control the chromatin association of MYC proteins are well understood, much less is known about how interacting proteins mediate MYC's effects on transcription. Here, we show that TFIIIC, an architectural protein complex that controls the three-dimensional chromatin organisation at its target sites, binds directly to the amino-terminal transcriptional regulatory domain of MYCN. Surprisingly, TFIIIC has no discernible role in MYCN-dependent gene expression and transcription elongation. Instead, MYCN and TFIIIC preferentially bind to promoters with paused RNAPII and globally limit the accumulation of non-phosphorylated RNAPII at promoters. Consistent with its ubiquitous role in transcription, MYCN broadly participates in hubs of active promoters. Depletion of TFIIIC further increases MYCN localisation to these hubs. This increase correlates with a failure of the nuclear exosome and BRCA1, both of which are involved in nascent RNA degradation, to localise to active promoters. Our data suggest that MYCN and TFIIIC exert an censoring function in early transcription that limits promoter accumulation of inactive RNAPII and facilitates promoter-proximal degradation of nascent RNA.


Sujet(s)
Chromatine , Protéine du proto-oncogène N-Myc , Régions promotrices (génétique) , RNA polymerase II , RNA polymerase II/métabolisme , RNA polymerase II/génétique , Protéine du proto-oncogène N-Myc/métabolisme , Protéine du proto-oncogène N-Myc/génétique , Humains , Chromatine/métabolisme , Liaison aux protéines , Facteurs de transcription TFII/métabolisme , Facteurs de transcription TFII/génétique , Transcription génétique , Lignée cellulaire tumorale
4.
Mol Biol Rep ; 51(1): 912, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39153092

RÉSUMÉ

Glioblastoma is the most aggressive brain cancer with an unfavorable prognosis for patient survival. Glioma stem cells, a subpopulation of cancer cells, drive tumor initiation, self-renewal, and resistance to therapy and, together with the microenvironment, play a crucial role in glioblastoma maintenance and progression. Neurotransmitters such as noradrenaline, dopamine, and serotonin have contrasting effects on glioblastoma development, stimulating or inhibiting its progression depending on the cellular context and through their action on glioma stem cells, perhaps changing the epigenetic landscape. Recent studies have revealed that serotonin and dopamine induce chromatin modifications related to transcriptional plasticity in the mammalian brain and possibly in glioblastoma; however, this topic still needs to be explored because of its potential implications for glioblastoma treatment. Also, it is essential to consider that neurotransmitters' effects depend on the tumor's microenvironment since it can significantly influence the response and behavior of cancer cells. This review examines the possible role of neurotransmitters as regulators of glioblastoma development, focusing on their impact on the chromatin of glioma stem cells.


Sujet(s)
Tumeurs du cerveau , Chromatine , Glioblastome , Cellules souches tumorales , Agents neuromédiateurs , Microenvironnement tumoral , Humains , Glioblastome/métabolisme , Glioblastome/génétique , Glioblastome/anatomopathologie , Agents neuromédiateurs/métabolisme , Chromatine/métabolisme , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Microenvironnement tumoral/génétique , Épigenèse génétique , Dopamine/métabolisme , Animaux , Sérotonine/métabolisme , Régulation de l'expression des gènes tumoraux
5.
Nat Commun ; 15(1): 6891, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39134516

RÉSUMÉ

HMGA1 is an abundant non-histone chromatin protein that has been implicated in embryonic development, cancer, and cellular senescence, but its specific role remains elusive. Here, we combine functional genomics approaches with graph theory to investigate how HMGA1 genomic deposition controls high-order chromatin networks in an oncogene-induced senescence model. While the direct role of HMGA1 in gene activation has been described previously, we find little evidence to support this. Instead, we show that the heterogeneous linear distribution of HMGA1 drives a specific 3D chromatin organization. HMGA1-dense loci form highly interactive networks, similar to, but independent of, constitutive heterochromatic loci. This, coupled with the exclusion of HMGA1-poor chromatin regions, leads to coordinated gene regulation through the repositioning of genes. In the absence of HMGA1, the whole process is largely reversed, but many regulatory interactions also emerge, amplifying the inflammatory senescence-associated secretory phenotype. Such HMGA1-mediated fine-tuning of gene expression contributes to the heterogeneous nature of senescence at the single-cell level. A similar 'buffer' effect of HMGA1 on inflammatory signalling is also detected in lung cancer cells. Our study reveals a mechanism through which HMGA1 modulates chromatin compartmentalization and gene regulation in senescence and beyond.


Sujet(s)
Vieillissement de la cellule , Chromatine , Protéine HMGA1a , Humains , Lignée cellulaire tumorale , Chromatine/métabolisme , Chromatine/génétique , Régulation de l'expression des gènes , Réseaux de régulation génique , Protéine HMGA1a/métabolisme , Protéine HMGA1a/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie
6.
Clin Epigenetics ; 16(1): 109, 2024 Aug 18.
Article de Anglais | MEDLINE | ID: mdl-39155390

RÉSUMÉ

BACKGROUND: Histone deacetylases (HDACs) are crucial regulators of gene expression, DNA synthesis, and cellular processes, making them essential targets in cancer research. HDAC6, specifically, influences protein stability and chromatin dynamics. Despite HDAC6's potential therapeutic value, its exact role in gene regulation and chromatin remodeling needs further clarification. This study examines how HDAC6 inactivation influences lysine acetyltransferase P300 stabilization and subsequent effects on chromatin structure and function in cancer cells. METHODS AND RESULTS: We employed the HDAC6 inhibitor ITF3756, siRNA, or CRISPR/Cas9 gene editing to inactivate HDAC6 in different epigenomic backgrounds. Constantly, this inactivation led to significant changes in chromatin accessibility, particularly increased acetylation of histone H3 lysines 9, 14, and 27 (ATAC-seq and H3K27Ac ChIP-seq analysis). Transcriptomics, proteomics, and gene ontology analysis revealed gene changes in cell proliferation, adhesion, migration, and apoptosis. Significantly, HDAC6 inactivation altered P300 ubiquitination, stabilizing P300 and leading to downregulating genes critical for cancer cell survival. CONCLUSIONS: Our study highlights the substantial impact of HDAC6 inactivation on the chromatin landscape of cancer cells and suggests a role for P300 in contributing to the anticancer effects. The stabilization of P300 with HDAC6 inhibition proposes a potential shift in therapeutic focus from HDAC6 itself to its interaction with P300. This finding opens new avenues for developing targeted cancer therapies, improving our understanding of epigenetic mechanisms in cancer cells.


Sujet(s)
Chromatine , Histone deacetylase 6 , Inhibiteurs de désacétylase d'histone , Humains , Histone deacetylase 6/génétique , Histone deacetylase 6/antagonistes et inhibiteurs , Chromatine/génétique , Chromatine/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Inhibiteurs de désacétylase d'histone/pharmacologie , Acétylation/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Protéine p300-E1A/génétique , Protéine p300-E1A/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Histone/métabolisme , Ubiquitination/effets des médicaments et des substances chimiques
7.
Elife ; 132024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39158543

RÉSUMÉ

Although the role of G-quadruplex (G4) DNA structures has been suggested in chromosomal looping this was not tested directly. Here, to test causal function, an array of G4s, or control sequence that does not form G4s, were inserted within chromatin in cells. In vivo G4 formation of the inserted G4 sequence array, and not the control sequence, was confirmed using G4-selective antibody. Compared to the control insert, we observed a remarkable increase in the number of 3D chromatin looping interactions from the inserted G4 array. This was evident within the immediate topologically associated domain (TAD) and throughout the genome. Locally, recruitment of enhancer histone marks and the transcriptional coactivator p300/Acetylated-p300 increased in the G4-array, but not in the control insertion. Resulting promoter-enhancer interactions and gene activation were clear up to 5 Mb away from the insertion site. Together, these show the causal role of G4s in enhancer function and long-range chromatin interactions. Mechanisms of 3D topology are primarily based on DNA-bound architectural proteins that induce/stabilize long-range interactions. Involvement of the underlying intrinsic DNA sequence/structure in 3D looping shown here therefore throws new light on how long-range chromosomal interactions might be induced or maintained.


Sujet(s)
Chromatine , G-quadruplexes , Régions promotrices (génétique) , Chromatine/métabolisme , Chromatine/composition chimique , Chromatine/génétique , Humains , Histone/métabolisme , Histone/composition chimique , Histone/génétique , Éléments activateurs (génétique)
8.
Cell Mol Life Sci ; 81(1): 361, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39158745

RÉSUMÉ

Genome-wide studies have demonstrated regulatory roles for diverse non-coding elements, but their precise and interrelated functions have often remained enigmatic. Addressing the need for mechanistic insight, we studied their roles in expression of Lhb which encodes the pituitary gonadotropic hormone that controls reproduction. We identified a bi-directional enhancer in gonadotrope-specific open chromatin, whose functional eRNA (eRNA2) supports permissive chromatin at the Lhb locus. The central untranscribed region of the enhancer contains an iMotif (iM), and is bound by Hmgb2 which stabilizes the iM and directs transcription specifically towards the functional eRNA2. A distinct downstream lncRNA, associated with an inducible G-quadruplex (G4) and iM, also facilitates Lhb expression, following its splicing in situ. GnRH activates Lhb transcription and increased levels of all three RNAs, eRNA2 showing the highest response, while estradiol, which inhibits Lhb, repressed levels of eRNA2 and the lncRNA. The levels of these regulatory RNAs and Lhb mRNA correlate highly in female mice, though strikingly not in males, suggesting a female-specific function. Our findings, which shed new light on the workings of non-coding elements and non-canonical DNA structures, reveal novel mechanisms regulating transcription which have implications not only in the central control of reproduction but also for other inducible genes.


Sujet(s)
Éléments activateurs (génétique) , ARN long non codant , ARN long non codant/génétique , ARN long non codant/métabolisme , Animaux , Éléments activateurs (génétique)/génétique , Femelle , Mâle , Souris , Régulation de l'expression des gènes , Souris de lignée C57BL , Chromatine/métabolisme , Chromatine/génétique , Humains , G-quadruplexes
9.
Sci Rep ; 14(1): 17786, 2024 08 01.
Article de Anglais | MEDLINE | ID: mdl-39090226

RÉSUMÉ

A long-standing question concerns the role of Z-DNA in transcription. Here we use a deep learning approach DeepZ that predicts Z-flipons based on DNA sequence, structural properties of nucleotides and omics data. We examined Z-flipons that are conserved between human and mouse genomes after generating whole-genome Z-flipon maps and then validated them by orthogonal approaches based on high resolution chemical mapping of Z-DNA and the transformer algorithm Z-DNABERT. For human and mouse, we revealed similar pattern of transcription factors, chromatin remodelers, and histone marks associated with conserved Z-flipons. We found significant enrichment of Z-flipons in alternative and bidirectional promoters associated with neurogenesis genes. We show that conserved Z-flipons are associated with increased experimentally determined transcription reinitiation rates compared to promoters without Z-flipons, but without affecting elongation or pausing. Our findings support a model where Z-flipons engage Transcription Factor E and impact phenotype by enabling the reset of preinitiation complexes when active, and the suppression of gene expression when engaged by repressive chromatin complexes.


Sujet(s)
ADN , Régions promotrices (génétique) , Animaux , Humains , Souris , ADN/génétique , ADN/métabolisme , Transcription génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Assemblage et désassemblage de la chromatine , Initiation de la transcription , Chromatine/génétique , Chromatine/métabolisme , Apprentissage profond , Séquence conservée
10.
Nat Commun ; 15(1): 6829, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39122693

RÉSUMÉ

mRNA biogenesis in the eukaryotic nucleus is a highly complex process. The numerous RNA processing steps are tightly coordinated to ensure that only fully processed transcripts are released from chromatin for export from the nucleus. Here, we present the hypothesis that fission yeast Dbp2, a ribonucleoprotein complex (RNP) remodelling ATPase of the DEAD-box family, is the key enzyme in an RNP assembly checkpoint at the 3'-end of genes. We show that Dbp2 interacts with the cleavage and polyadenylation complex (CPAC) and localises to cleavage bodies, which are enriched for 3'-end processing factors and proteins involved in nuclear RNA surveillance. Upon loss of Dbp2, 3'-processed, polyadenylated RNAs accumulate on chromatin and in cleavage bodies, and CPAC components are depleted from the soluble pool. Under these conditions, cells display an increased likelihood to skip polyadenylation sites and a delayed transcription termination, suggesting that levels of free CPAC components are insufficient to maintain normal levels of 3'-end processing. Our data support a model in which Dbp2 is the active component of an mRNP remodelling checkpoint that licenses RNA export and is coupled to CPAC release.


Sujet(s)
DEAD-box RNA helicases , Ribonucléoprotéines , Protéines de Schizosaccharomyces pombe , Schizosaccharomyces , Schizosaccharomyces/génétique , Schizosaccharomyces/métabolisme , Ribonucléoprotéines/métabolisme , Ribonucléoprotéines/génétique , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , Protéines de Schizosaccharomyces pombe/métabolisme , Protéines de Schizosaccharomyces pombe/génétique , Polyadénylation , ARN messager/métabolisme , ARN messager/génétique , Facteurs de clivage et de polyadénylation de l'ARN messager/métabolisme , Facteurs de clivage et de polyadénylation de l'ARN messager/génétique , Chromatine/métabolisme , ARN fongique/métabolisme , ARN fongique/génétique , Noyau de la cellule/métabolisme
11.
Nat Commun ; 15(1): 6852, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39127768

RÉSUMÉ

Cis-regulatory elements (CREs) are pivotal in orchestrating gene expression throughout diverse biological systems. Accurate identification and in-depth characterization of functional CREs are crucial for decoding gene regulation networks during cellular processes. In this study, we develop Kethoxal-Assisted Single-stranded DNA Assay for Transposase-Accessible Chromatin with Sequencing (KAS-ATAC-seq) to quantitatively analyze the transcriptional activity of CREs. A main advantage of KAS-ATAC-seq lies in its precise measurement of ssDNA levels within both proximal and distal ATAC-seq peaks, enabling the identification of transcriptional regulatory sequences. This feature is particularly adept at defining Single-Stranded Transcribing Enhancers (SSTEs). SSTEs are highly enriched with nascent RNAs and specific transcription factors (TFs) binding sites that define cellular identity. Moreover, KAS-ATAC-seq provides a detailed characterization and functional implications of various SSTE subtypes. Our analysis of CREs during mouse neural differentiation demonstrates that KAS-ATAC-seq can effectively identify immediate-early activated CREs in response to retinoic acid (RA) treatment. Our findings indicate that KAS-ATAC-seq provides more precise annotation of functional CREs in transcription. Future applications of KAS-ATAC-seq would help elucidate the intricate dynamics of gene regulation in diverse biological processes.


Sujet(s)
Facteurs de transcription , Animaux , Souris , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Transcription génétique , Éléments activateurs (génétique)/génétique , Chromatine/métabolisme , Chromatine/génétique , Sites de fixation , Humains , ADN simple brin/génétique , ADN simple brin/métabolisme , Séquençage après immunoprécipitation de la chromatine/méthodes , Transposases/métabolisme , Transposases/génétique , Éléments de régulation transcriptionnelle , Trétinoïne/pharmacologie , Trétinoïne/métabolisme , Régulation de l'expression des gènes , Différenciation cellulaire/génétique , Analyse de séquence d'ADN/méthodes , Séquences d'acides nucléiques régulatrices/génétique
12.
Chromosome Res ; 32(3): 11, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39126507

RÉSUMÉ

Interphase chromosomes reside within distinct nuclear regions known as chromosome territories (CTs). Recent observations from Hi-C analyses, a method mapping chromosomal interactions, have revealed varied decay in contact probabilities among different chromosomes. Our study explores the relationship between this contact decay and the particular shapes of the chromosome territories they occupy. For this, we employed molecular dynamics (MD) simulations to examine how confined polymers, resembling chromosomes, behave within different confinement geometries similar to chromosome territory boundaries. Our simulations unveil so far unreported relationships between contact probabilities and end-to-end distances varying based on different confinement geometries. These findings highlight the crucial impact of chromosome territories on shaping the larger-scale properties of 3D genome organization. They emphasize the intrinsic connection between the shapes of these territories and the contact behaviors exhibited by chromosomes. Understanding these correlations is key to accurately interpret Hi-C and microscopy data, and offers vital insights into the foundational principles governing genomic organization.


Sujet(s)
Chromosomes , Simulation de dynamique moléculaire , Polymères/composition chimique , Humains , Chromatine/génétique , Interphase
13.
PLoS Genet ; 20(8): e1011366, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39102423

RÉSUMÉ

In Saccharomyces cerevisiae, the forkhead (Fkh) transcription factor Fkh1 (forkhead homolog) enhances the activity of many DNA replication origins that act in early S-phase (early origins). Current models posit that Fkh1 acts directly to promote these origins' activity by binding to origin-adjacent Fkh1 binding sites (FKH sites). However, the post-DNA binding functions that Fkh1 uses to promote early origin activity are poorly understood. Fkh1 contains a conserved FHA (forkhead associated) domain, a protein-binding module with specificity for phosphothreonine (pT)-containing partner proteins. At a small subset of yeast origins, the Fkh1-FHA domain enhances the ORC (origin recognition complex)-origin binding step, the G1-phase event that initiates the origin cycle. However, the importance of the Fkh1-FHA domain to either chromosomal replication or ORC-origin interactions at genome scale is unclear. Here, S-phase SortSeq experiments were used to compare genome replication in proliferating FKH1 and fkh1-R80A mutant cells. The Fkh1-FHA domain promoted the activity of ≈ 100 origins that act in early to mid- S-phase, including the majority of centromere-associated origins, while simultaneously inhibiting ≈ 100 late origins. Thus, in the absence of a functional Fkh1-FHA domain, the temporal landscape of the yeast genome was flattened. Origins are associated with a positioned nucleosome array that frames a nucleosome depleted region (NDR) over the origin, and ORC-origin binding is necessary but not sufficient for this chromatin organization. To ask whether the Fkh1-FHA domain had an impact on this chromatin architecture at origins, ORC ChIPSeq data generated from proliferating cells and MNaseSeq data generated from G1-arrested and proliferating cell populations were assessed. Origin groups that were differentially regulated by the Fkh1-FHA domain were characterized by distinct effects of this domain on ORC-origin binding and G1-phase chromatin. Thus, the Fkh1-FHA domain controlled the distinct chromatin architecture at early origins in G1-phase and regulated origin activity in S-phase.


Sujet(s)
Chromatine , Réplication de l'ADN , Phase G1 , Complexe ORC , Origine de réplication , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Origine de réplication/génétique , Protéines de Saccharomyces cerevisiae/génétique , Protéines de Saccharomyces cerevisiae/métabolisme , Réplication de l'ADN/génétique , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Chromatine/génétique , Chromatine/métabolisme , Complexe ORC/génétique , Complexe ORC/métabolisme , Phase G1/génétique , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Phase S/génétique , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Domaines protéiques/génétique , Sites de fixation , Liaison aux protéines , Chromosomes de champignon/génétique , Chromosomes de champignon/métabolisme , Nucléosomes/métabolisme , Nucléosomes/génétique
14.
Nat Commun ; 15(1): 6952, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138204

RÉSUMÉ

Biomolecular condensates play a significant role in chromatin activities, primarily by concentrating and compartmentalizing proteins and/or nucleic acids. However, their genomic landscapes and compositions remain largely unexplored due to a lack of dedicated computational tools for systematic identification in vivo. To address this, we develop CondSigDetector, a computational framework designed to detect condensate-like chromatin-associated protein co-occupancy signatures (CondSigs), to predict genomic loci and component proteins of distinct chromatin-associated biomolecular condensates. Applying this framework to mouse embryonic stem cells (mESC) and human K562 cells enable us to depict the high-resolution genomic landscape of chromatin-associated biomolecular condensates, and uncover both known and potentially unknown biomolecular condensates. Multi-omics analysis and experimental validation further verify the condensation properties of CondSigs. Additionally, our investigation sheds light on the impact of chromatin-associated biomolecular condensates on chromatin activities. Collectively, CondSigDetector provides an approach to decode the genomic landscape of chromatin-associated condensates, facilitating a deeper understanding of their biological functions and underlying mechanisms in cells.


Sujet(s)
Condensats biomoléculaires , Chromatine , Chromatine/métabolisme , Chromatine/génétique , Humains , Animaux , Souris , Cellules K562 , Condensats biomoléculaires/métabolisme , Génomique/méthodes , Cellules souches embryonnaires de souris/métabolisme , Biologie informatique/méthodes , Génome
15.
Nat Commun ; 15(1): 7015, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39147779

RÉSUMÉ

During meiosis, nucleoprotein filaments of the strand exchange proteins RAD51 and DMC1 are crucial for repairing SPO11-generated DNA double-strand breaks (DSBs) by homologous recombination (HR). A balanced activity of positive and negative RAD51/DMC1 regulators ensures proper recombination. Fidgetin-like 1 (FIGNL1) was previously shown to negatively regulate RAD51 in human cells. However, FIGNL1's role during meiotic recombination in mammals remains unknown. Here, we decipher the meiotic functions of FIGNL1 and FIGNL1 Interacting Regulator of Recombination and Mitosis (FIRRM) using male germline-specific conditional knock-out (cKO) mouse models. Both FIGNL1 and FIRRM are required for completing meiotic prophase in mouse spermatocytes. Despite efficient recruitment of DMC1 on ssDNA at meiotic DSB hotspots, the formation of late recombination intermediates is defective in Firrm cKO and Fignl1 cKO spermatocytes. Moreover, the FIGNL1-FIRRM complex limits RAD51 and DMC1 accumulation on intact chromatin, independently from the formation of SPO11-catalyzed DSBs. Purified human FIGNL1ΔN alters the RAD51/DMC1 nucleoprotein filament structure and inhibits strand invasion in vitro. Thus, this complex might regulate RAD51 and DMC1 association at sites of meiotic DSBs to promote proficient strand invasion and processing of recombination intermediates.


Sujet(s)
Protéines du cycle cellulaire , Cassures double-brin de l'ADN , Protéines de liaison à l'ADN , Méiose , Souris knockout , Rad51 Recombinase , Spermatocytes , Rad51 Recombinase/métabolisme , Rad51 Recombinase/génétique , Animaux , Mâle , Méiose/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Humains , Souris , Spermatocytes/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Recombinaison homologue , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Altération de l'ADN , Endodeoxyribonucleases/métabolisme , Endodeoxyribonucleases/génétique , Chromatine/métabolisme , Protéines de liaison aux phosphates/métabolisme , Protéines de liaison aux phosphates/génétique
16.
J Exp Med ; 221(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39150482

RÉSUMÉ

Coordination of cellular metabolism is essential for optimal T cell responses. Here, we identify cytosolic acetyl-CoA production as an essential metabolic node for CD8 T cell function in vivo. We show that CD8 T cell responses to infection depend on acetyl-CoA derived from citrate via the enzyme ATP citrate lyase (ACLY). However, ablation of ACLY triggers an alternative, acetate-dependent pathway for acetyl-CoA production mediated by acyl-CoA synthetase short-chain family member 2 (ACSS2). Mechanistically, acetate fuels both the TCA cycle and cytosolic acetyl-CoA production, impacting T cell effector responses, acetate-dependent histone acetylation, and chromatin accessibility at effector gene loci. When ACLY is functional, ACSS2 is not required, suggesting acetate is not an obligate metabolic substrate for CD8 T cell function. However, loss of ACLY renders CD8 T cells dependent on acetate (via ACSS2) to maintain acetyl-CoA production and effector function. Together, ACLY and ACSS2 coordinate cytosolic acetyl-CoA production in CD8 T cells to maintain chromatin accessibility and T cell effector function.


Sujet(s)
ATP citrate (pro-S)-lyase , Acétates , Acétyl coenzyme A , Lymphocytes T CD8+ , Chromatine , Souris de lignée C57BL , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Animaux , Chromatine/métabolisme , Acétyl coenzyme A/métabolisme , ATP citrate (pro-S)-lyase/métabolisme , ATP citrate (pro-S)-lyase/génétique , Souris , Acétates/métabolisme , Acetate coA-ligase/métabolisme , Acetate coA-ligase/génétique , Acétylation , Souris knockout , Cytosol/métabolisme , Histone/métabolisme
17.
PLoS Comput Biol ; 20(8): e1011854, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39093856

RÉSUMÉ

Single-cell ATAC-seq sequencing data (scATAC-seq) has been widely used to investigate chromatin accessibility on the single-cell level. One important application of scATAC-seq data analysis is differential chromatin accessibility (DA) analysis. However, the data characteristics of scATAC-seq such as excessive zeros and large variability of chromatin accessibility across cells impose a unique challenge for DA analysis. Existing statistical methods focus on detecting the mean difference of the chromatin accessible regions while overlooking the distribution difference. Motivated by real data exploration that distribution difference exists among cell types, we introduce a novel composite statistical test named "scaDA", which is based on zero-inflated negative binomial model (ZINB), for performing differential distribution analysis of chromatin accessibility by jointly testing the abundance, prevalence and dispersion simultaneously. Benefiting from both dispersion shrinkage and iterative refinement of mean and prevalence parameter estimates, scaDA demonstrates its superiority to both ZINB-based likelihood ratio tests and published methods by achieving the highest power and best FDR control in a comprehensive simulation study. In addition to demonstrating the highest power in three real sc-multiome data analyses, scaDA successfully identifies differentially accessible regions in microglia from sc-multiome data for an Alzheimer's disease (AD) study that are most enriched in GO terms related to neurogenesis and the clinical phenotype of AD, and AD-associated GWAS SNPs.


Sujet(s)
Chromatine , Analyse sur cellule unique , Chromatine/génétique , Chromatine/métabolisme , Chromatine/composition chimique , Analyse sur cellule unique/méthodes , Analyse sur cellule unique/statistiques et données numériques , Humains , Biologie informatique/méthodes , Maladie d'Alzheimer/génétique , Modèles statistiques , Séquençage après immunoprécipitation de la chromatine/méthodes , Simulation numérique , Animaux , Analyse de séquence d'ADN/méthodes , Algorithmes
18.
Nat Plants ; 10(8): 1184-1200, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-39103456

RÉSUMÉ

Scaffolding is crucial for constructing most chromosome-level genomes. The high-throughput chromatin conformation capture (Hi-C) technology has become the primary scaffolding strategy due to its convenience and cost-effectiveness. As sequencing technologies and assembly algorithms advance, constructing haplotype-resolved genomes is increasingly preferred because haplotypes can provide additional genetic information on allelic and non-allelic variations. ALLHiC is a widely used allele-aware scaffolding tool designed for this purpose. However, its dependence on chromosome-level reference genomes and a higher chromosome misassignment rate still impede the unravelling of haplotype-resolved genomes. Here we present HapHiC, a reference-independent allele-aware scaffolding tool with superior performance on chromosome assignment as well as contig ordering and orientation. In addition, we provide new insights into the challenges in allele-aware scaffolding by conducting comprehensive analyses on various adverse factors. Finally, with the help of HapHiC, we constructed the haplotype-resolved allotriploid genome for Miscanthus × giganteus, an important lignocellulosic bioenergy crop.


Sujet(s)
Chromosomes de plante , Génome végétal , Haplotypes , Chromosomes de plante/génétique , Chromatine/génétique , Poaceae/génétique , Séquençage nucléotidique à haut débit/méthodes , Allèles
19.
Proc Natl Acad Sci U S A ; 121(33): e2409167121, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39116133

RÉSUMÉ

Linker histones play an essential role in chromatin packaging by facilitating compaction of the 11-nm fiber of nucleosomal "beads on a string." The result is a heterogeneous condensed state with local properties that range from dynamic, irregular, and liquid-like to stable and regular structures (the 30-nm fiber), which in turn impact chromatin-dependent activities at a fundamental level. The properties of the condensed state depend on the type of linker histone, particularly on the highly disordered C-terminal tail, which is the most variable region of the protein, both between species, and within the various subtypes and cell-type specific variants of a given organism. We have developed an in vitro model system comprising linker histone tail and linker DNA, which although very minimal, displays surprisingly complex behavior, and is sufficient to model the known states of linker histone-condensed chromatin: disordered "fuzzy" complexes ("open" chromatin), dense liquid-like assemblies (dynamic condensates), and higher-order structures (organized 30-nm fibers). A crucial advantage of such a simple model is that it allows the study of the various condensed states by NMR, circular dichroism, and scattering methods. Moreover, it allows capture of the thermodynamics underpinning the transitions between states through calorimetry. We have leveraged this to rationalize the distinct condensing properties of linker histone subtypes and variants across species that are encoded by the amino acid content of their C-terminal tails. Three properties emerge as key to defining the condensed state: charge density, lysine/arginine ratio, and proline-free regions, and we evaluate each separately using a strategic mutagenesis approach.


Sujet(s)
ADN , Histone , Nucléosomes , Histone/composition chimique , Histone/métabolisme , Histone/génétique , ADN/composition chimique , ADN/métabolisme , Nucléosomes/métabolisme , Nucléosomes/composition chimique , Chromatine/composition chimique , Chromatine/métabolisme , Chromatine/génétique , Animaux , Humains
20.
Genome Biol ; 25(1): 225, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39152456

RÉSUMÉ

BACKGROUND: Single-cell chromatin accessibility assays, such as scATAC-seq, are increasingly employed in individual and joint multi-omic profiling of single cells. As the accumulation of scATAC-seq and multi-omics datasets continue, challenges in analyzing such sparse, noisy, and high-dimensional data become pressing. Specifically, one challenge relates to optimizing the processing of chromatin-level measurements and efficiently extracting information to discern cellular heterogeneity. This is of critical importance, since the identification of cell types is a fundamental step in current single-cell data analysis practices. RESULTS: We benchmark 8 feature engineering pipelines derived from 5 recent methods to assess their ability to discover and discriminate cell types. By using 10 metrics calculated at the cell embedding, shared nearest neighbor graph, or partition levels, we evaluate the performance of each method at different data processing stages. This comprehensive approach allows us to thoroughly understand the strengths and weaknesses of each method and the influence of parameter selection. CONCLUSIONS: Our analysis provides guidelines for choosing analysis methods for different datasets. Overall, feature aggregation, SnapATAC, and SnapATAC2 outperform latent semantic indexing-based methods. For datasets with complex cell-type structures, SnapATAC and SnapATAC2 are preferred. With large datasets, SnapATAC2 and ArchR are most scalable.


Sujet(s)
Référenciation , Chromatine , Analyse sur cellule unique , Analyse sur cellule unique/méthodes , Chromatine/génétique , Chromatine/métabolisme , Humains , Biologie informatique/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE