Franck Martin,Asgar Abbas Kazrani,Julie Lafouge,Dana Mariel Diaz-Jimenez,Stéphanie Siebert,Leonie Fabbro-Burtschell,Emma Maillard,Karine Lapouge,Haydyn David Thomas Mertens,Claude Sauter,Alexander Leitner,Françoise Ochsenbein,Alexandre Blais,Elisa Bergamin
{"title":"Structure of the nucleosome-bound human BCL7A.","authors":"Franck Martin,Asgar Abbas Kazrani,Julie Lafouge,Dana Mariel Diaz-Jimenez,Stéphanie Siebert,Leonie Fabbro-Burtschell,Emma Maillard,Karine Lapouge,Haydyn David Thomas Mertens,Claude Sauter,Alexander Leitner,Françoise Ochsenbein,Alexandre Blais,Elisa Bergamin","doi":"10.1093/nar/gkaf273","DOIUrl":"https://doi.org/10.1093/nar/gkaf273","url":null,"abstract":"Proteins of the BCL7 family (BCL7A, BCL7B, and BCL7C) are among the most recently identified subunits of the mammalian SWI/SNF chromatin remodeler complex and are absent from the unicellular version of this complex. Their function in the complex is unknown, and very limited structural information is available, despite the fact that they are mutated in several cancer types, most notably blood malignancies and hence medically relevant. Here, using cryo-electron microscopy in combination with biophysical and biochemical approaches, we show that BCL7A forms a stable, high-affinity complex with the nucleosome core particle (NCP) through binding of BCL7A with the acidic patch of the nucleosome via an arginine anchor motif. This interaction is impaired by BCL7A mutations found in cancer. Further, we determined that BCL7A contributes to the remodeling activity of the mSWI/SNF complex and we examined its function at the genomic level. Our findings reveal how BCL7 proteins interact with the NCP and help rationalize the impact of cancer-associated mutations. By providing structural information on the positioning of BCL7 on the NCP, our results broaden the understanding of the mechanism by which SWI/SNF recognizes the chromatin fiber.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"246 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143819155","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"High-throughput screen of 100 000 small molecules in C9ORF72 ALS neurons identifies spliceosome modulators that mobilize G4C2 repeat RNA into nuclear export and repeat associated non-canonical translation.","authors":"Maartje J Luteijn,Varun Bhaskar,Dominic Trojer,Melanie Schürz,Hicham Mahboubi,Cornelia Handl,Nicolas Pizzato,Martin Pfeifer,Ruxandra Dafinca,Hans Voshol,Elisa Giorgetti,Carole Manneville,Isabelle P M Garnier,Matthias Müller,Fanning Zeng,Kathrin Buntin,Roger Markwalder,Harald Schröder,Jan Weiler,Dora Khar,Tim Schuhmann,Paul J Groot-Kormelink,Caroline Gubser Keller,Pierre Farmer,Angela MacKay,Martin Beibel,Guglielmo Roma,Giovanni D'Ario,Claudia Merkl,Michael Schebesta,Marc Hild,Fiona Elwood,Björn F Vahsen,Nina Ripin,Antoine Clery,Frederic Allain,Mark Labow,Daniela Gabriel,Jeffrey A Chao,Kevin Talbot,Mark Nash,Jürg Hunziker,Nicole C Meisner-Kober","doi":"10.1093/nar/gkaf253","DOIUrl":"https://doi.org/10.1093/nar/gkaf253","url":null,"abstract":"An intronic G4C2 repeat expansion in the C9ORF72 gene is the major known cause for Amyotrophic Lateral Sclerosis (ALS), with current evidence for both, loss of function and pathological gain of function disease mechanisms. We screened 96 200 small molecules in C9ORF72 patient iPS neurons for modulation of nuclear G4C2 RNA foci and identified 82 validated hits, including the Brd4 inhibitor JQ1 as well as novel analogs of Spliceostatin-A, a known modulator of SF3B1, the branch point binding protein of the U2-snRNP. Spliceosome modulation by these SF3B1 targeted compounds recruits SRSF1 to nuclear G4C2 RNA, mobilizing it from RNA foci into nucleocytoplasmic export. This leads to increased repeat-associated non-canonical (RAN) translation and ultimately, enhanced cell toxicity. Our data (i) provide a new pharmacological entry point with novel as well as known, publicly available tool compounds for dissection of C9ORF72 pathobiology in C9ORF72 ALS models, (ii) allowing to differentially modulate RNA foci versus RAN translation, and (iii) suggest that therapeutic RNA foci elimination strategies warrant caution due to a potential storage function, counteracting translation into toxic dipeptide repeat polyproteins. Instead, our data support modulation of nuclear export via SRSF1 or SR protein kinases as possible targets for future pharmacological drug discovery.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"12 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143819355","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sevil Zencir,Daniel Dilg,Maria Jessica Bruzzone,Françoise Stutz,Julien Soudet,David Shore,Benjamin Albert
{"title":"A two-step regulatory mechanism dynamically controls histone H3 acetylation by SAGA complex at growth-related promoters.","authors":"Sevil Zencir,Daniel Dilg,Maria Jessica Bruzzone,Françoise Stutz,Julien Soudet,David Shore,Benjamin Albert","doi":"10.1093/nar/gkaf276","DOIUrl":"https://doi.org/10.1093/nar/gkaf276","url":null,"abstract":"Acetylation of histone H3 at residue K9 (H3K9ac) is a dynamically regulated mark associated with transcriptionally active promoters in eukaryotes. However, our understanding of the relationship between H3K9ac and gene expression remains mostly correlative. In this study, we identify a large suite of growth-related (GR) genes in yeast that undergo a particularly strong down-regulation of both transcription and promoter-associated H3K9ac upon stress, and delineate the roles of transcriptional activators (TAs), repressors, SAGA (Spt-Ada-Gcn5 acetyltransferase) histone acetyltransferase, and RNA-polymerase II in this response. We demonstrate that H3K9 acetylation states are orchestrated by a two-step mechanism driven by the dynamic binding of transcriptional repressors (TRs) and activators, that is independent of transcription. In response to stress, promoter release of TAs at GR genes is a prerequisite for rapid reduction of H3K9ac, whereas binding of TRs is required to establish a hypo-acetylated, strongly repressed state.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"25 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143819357","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Nucleotide-level characterization and improvement of l-arabinose- and l-rhamnose-inducible systems in E. coli using a high-throughput approach.","authors":"Nancy M Kim,Danqia Peng,Nicholas R Sandoval","doi":"10.1093/nar/gkaf224","DOIUrl":"https://doi.org/10.1093/nar/gkaf224","url":null,"abstract":"The commonly used arabinose- and rhamnose-inducible Escherichia coli promoters, PBAD and PRha, exhibit tight regulation through activation via their respective transcription factors, AraC and RhaS, alongside the cyclic AMP receptor protein. The mechanisms of these promoters have been characterized on a parts level, but nucleotide-level analysis has yet to be elucidated. Therefore, we describe here a massively parallel reporter assay that maps regulatory sites at the nucleotide level. The relative importance of nucleotides in each binding site is revealed, including loci not included in previous annotations. For PBAD, we confirm known sites and reveal novel binding sites involved in modulating gene expression. In PRha, we refine the length and sequence specificity of rhaI half-sites, updating previous annotations and providing nucleotide level insights into RhaS-mediated regulation. Mutations that lead to increased promoter strength, wider dynamic range, and altered basal expression are identified for both promoters. Engineered versions of PBAD and PRha promoters based on this data show improvements in dynamic range alongside a seven- and three-fold increase in promoter strength, respectively, with a slight increase in basal expression for the PBAD promoters and no significant increase for PRha. This work expands the genetic parts \"toolkit\" and increases the understanding of these important commonly used promoters.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"4 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143822782","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hsiang-Chi Huang,Michelle Fong,Iwona Nowak,Evgeniia Shcherbinina,Vivian Lobo,Danica F Besavilla,Hang T Huynh,Karin Schön,Jakub O Westholm,Carola Fernandez,Angana A H Patel,Clotilde Wiel,Volkan I Sayin,Dimitrios G Anastasakis,Davide Angeletti,Aishe A Sarshad
{"title":"Nuclear AGO2 supports influenza A virus replication through type-I interferon regulation.","authors":"Hsiang-Chi Huang,Michelle Fong,Iwona Nowak,Evgeniia Shcherbinina,Vivian Lobo,Danica F Besavilla,Hang T Huynh,Karin Schön,Jakub O Westholm,Carola Fernandez,Angana A H Patel,Clotilde Wiel,Volkan I Sayin,Dimitrios G Anastasakis,Davide Angeletti,Aishe A Sarshad","doi":"10.1093/nar/gkaf268","DOIUrl":"https://doi.org/10.1093/nar/gkaf268","url":null,"abstract":"The role of Argonaute (AGO) proteins and the RNA interference (RNAi) machinery in mammalian antiviral response has been debated. Therefore, we set out to investigate how mammalian RNAi impacts influenza A virus (IAV) infection. We reveal that IAV infection triggers nuclear accumulation of AGO2, which is directly facilitated by p53 activation. Mechanistically, we show that IAV induces nuclear AGO2 targeting of TRIM71and type-I interferon-pathway genes for silencing. Accordingly, Tp53-/- mice do not accumulate nuclear AGO2 and demonstrate decreased susceptibility to IAV infection. Hence, the RNAi machinery is highjacked by the virus to evade the immune system and support viral replication. Furthermore, the FDA-approved drug, arsenic trioxide, prevents p53 nuclear translocation, increases interferon response and decreases viral replication in vitro and in a mouse model in vivo. Our data indicate that targeting the AGO2:p53-mediated silencing of innate immunity may offer a promising strategy to mitigate viral infections.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"20 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143827128","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Selict-seq profiles genome-wide off-target effects in adenosine base editing","authors":"Kexin Yuan, Xin Xi, Shaoqing Han, Jingyu Han, Bin Zhao, Qi Wei, Xiang Zhou","doi":"10.1093/nar/gkaf281","DOIUrl":"https://doi.org/10.1093/nar/gkaf281","url":null,"abstract":"Adenosine base editors (ABEs) facilitate A·T to G·C base pair conversion with significant therapeutic potential for correcting pathogenic point mutations in human genetic diseases, such as sickle cell anemia and β-thalassemia. Unlike CRISPR–Cas9 systems that induce double-strand breaks, ABEs operate through precise deamination, avoiding chromosomal instability. However, the off-target editing effects of ABEs remain inadequately characterized. In this study, we present a biochemical method Selict-seq, designed to evaluate genome-wide off-target editing by ABEs. Selict-seq specifically captures deoxyinosine-containing single-stranded DNA and precisely identifies deoxyadenosine-to-deoxyinosine (dA-to-dI) mutation sites, elucidating the off-target effects induced by ABEs. Through investigations involving three single-guide RNAs, we identified numerous unexpected off-target edits both within and outside the protospacer regions. Notably, ABE8e(V106W) exhibited distinct off-target characteristics, including high editing rates (>10%) at previously unreported sites (e.g. RNF2 and EMX1) and out-of-protospacer mutations. These findings significantly advance our understanding of the off-target landscape associated with ABEs. In summary, our approach enables an unbiased analysis of the ABE editome and provides a widely applicable tool for specificity evaluation of various emerging genome editing technologies that produce intermediate products as deoxyinosine.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"217 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143813537","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Georgina S Lloyd, Elton R Stephens, Alessandro Di Maio, Christopher M Thomas
{"title":"Activation, incompatibility, and displacement of FIB replicons in E. coli","authors":"Georgina S Lloyd, Elton R Stephens, Alessandro Di Maio, Christopher M Thomas","doi":"10.1093/nar/gkaf275","DOIUrl":"https://doi.org/10.1093/nar/gkaf275","url":null,"abstract":"Multi-replicon sex-factor F is the archetype of the largest plasmid group in clinical Enterobacteriaceae. Such plasmids spread antimicrobial resistance (AMR) and virulence functions in commensal bacteria of humans and animals. Displacing (curing) these plasmids by blocking replication and neutralizing addiction is successful with the curing cassette on a high-copy-number vector but, with conjugative IncP-1 plasmid RK2 as vector for our “anti-F cassette”, displacement of F’prolac is inefficient unless curing-plasmid copy-number is raised 1.5- to 2-fold. Here we report that it is the anti-FIB segment, originating from FIB-FII plasmid pO157, which needs potentiation. We show that the FIB replicon in F (F-FIB) is defective due to a sub-optimal rep ribosome-binding-site (rbs) but can be activated by FIB-Rep protein expressed from our anti-FIB segment joined to RK2. Deleting FIB-rep from the anti-F cassette removed the need for potentiation. A pO157-FIB single-replicon plasmid was displaced efficiently by the complete anti-F cassette without potentiation, but an F-FIB plasmid, mutated to have a pO157-like rep rbs, was not, indicating that sequence divergence between F and pO157 FIB replicons has weakened their negative cross-reactivity. Thus, raising vector copy-number slightly may be sufficient to increase displacement of plasmids similar but not identical to the sequences in the curing cassette.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"18 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143813726","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Federica Fiorentino, Matthias Thoms, Klemens Wild, Timo Denk, Jingdong Cheng, Jakub Zeman, Irmgard Sinning, Ed Hurt, Roland Beckmann
{"title":"Highly conserved ribosome biogenesis pathways between human and yeast revealed by the MDN1-NLE1 interaction and NLE1 containing pre-60S subunits","authors":"Federica Fiorentino, Matthias Thoms, Klemens Wild, Timo Denk, Jingdong Cheng, Jakub Zeman, Irmgard Sinning, Ed Hurt, Roland Beckmann","doi":"10.1093/nar/gkaf255","DOIUrl":"https://doi.org/10.1093/nar/gkaf255","url":null,"abstract":"The assembly of ribosomal subunits, primarily occurring in the nucleolar and nuclear compartments, is a highly complex process crucial for cellular function. This study reveals the conservation of ribosome biogenesis between yeast and humans, illustrated by the structural similarities of ribosomal subunit intermediates. By using X-ray crystallography and cryo-EM, the interaction between the human AAA+ ATPase MDN1 and the 60S assembly factor NLE1 is compared with the yeast homologs Rea1 and Rsa4. The MDN1-MIDAS and NLE1-Ubl complex structure at 2.3 Å resolution mirrors the highly conserved interaction patterns observed in yeast. Moreover, human pre-60S intermediates bound to the dominant negative NLE1-E85A mutant revealed at 2.8 Å resolution an architecture that largely matched the equivalent yeast structures. Conformation of rRNA, assembly factors and their interaction networks are highly conserved. Additionally, novel human pre-60S intermediates with a non-rotated 5S RNP and processed ITS2/foot structure but incomplete intersubunit surface were identified to be similar to counterparts observed in yeast. These findings confirm that the MDN1-NLE1-driven transition phase of the 60S assembly is essentially identical, supporting the idea that ribosome biogenesis is a highly conserved process across eukaryotic cells, employing an evolutionary preservation of ribosomal assembly mechanisms.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"34 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143813767","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Michael E Østergaard,Michele Carrer,Brooke A Anderson,Megan Afetian,Mohsen A Bakooshli,Jinro A Santos,Stephanie K Klein,Juliana Capitanio,Graeme C Freestone,Michael Tanowitz,Rodrigo Galindo-Murillo,Hans J Gaus,Chrissa A Dwyer,Michaela Jackson,Paymaan Jafar-Nejad,Frank Rigo,Punit P Seth,Katherine U Gaynor,Steven J Stanway,Liudvikas Urbonas,Megan A St Denis,Simone Pellegrino,Gustavo A Bezerra,Michael Rigby,Ellen Gowans,Katerine Van Rietschoten,Paul Beswick,Liuhong Chen,Michael J Skynner,Eric E Swayze
{"title":"Conjugation to a transferrin receptor 1-binding Bicycle peptide enhances ASO and siRNA potency in skeletal and cardiac muscles.","authors":"Michael E Østergaard,Michele Carrer,Brooke A Anderson,Megan Afetian,Mohsen A Bakooshli,Jinro A Santos,Stephanie K Klein,Juliana Capitanio,Graeme C Freestone,Michael Tanowitz,Rodrigo Galindo-Murillo,Hans J Gaus,Chrissa A Dwyer,Michaela Jackson,Paymaan Jafar-Nejad,Frank Rigo,Punit P Seth,Katherine U Gaynor,Steven J Stanway,Liudvikas Urbonas,Megan A St Denis,Simone Pellegrino,Gustavo A Bezerra,Michael Rigby,Ellen Gowans,Katerine Van Rietschoten,Paul Beswick,Liuhong Chen,Michael J Skynner,Eric E Swayze","doi":"10.1093/nar/gkaf270","DOIUrl":"https://doi.org/10.1093/nar/gkaf270","url":null,"abstract":"Improving the delivery of antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs) to skeletal and cardiac muscles remains a pivotal task toward the broader application of oligonucleotide therapeutics. The targeting of myofibers and cardiomyocytes via conjugation of ASOs and siRNAs to ligands that bind the human transferrin receptor 1 (TfR1) has gathered significant interest in recent years. However, the selection of ligands with low molecular weight and optimal biophysical and binding properties is crucial to maximize the potential of the TfR1 ligand-conjugated antisense (LICA) technology. Here, through effective combination of phage display and peptide medicinal chemistry, we identified and characterized a bicyclic peptide (Bicycle® molecule BCY17901), with a molecular weight of ∼2 kDa, that binds human TfR1 with high affinity and specificity. Conjugation to BCY17901 improved ASO and siRNA potency in skeletal and cardiac muscles of human TfR1 knock-in mice, after either intravenous or subcutaneous administration. Furthermore, single-nucleus RNA sequencing showed that conjugation to BCY17901 enhanced ASO activity in myonuclei of different muscle fiber types. Importantly, we demonstrated good translatability of our TfR1-targeting platform in skeletal and cardiac muscles of nonhuman primates. Our results offer great promise toward potential future applications of low-molecular-weight Bicycle LICA therapeutics for the treatment of diseases affecting skeletal muscle and heart.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"40 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143819156","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mariella Cuomo,Davide Costabile,Rosa Della Monica,Michela Buonaiuto,Federica Trio,Giulia De Riso,Roberta Visconti,Lorenzo Chiariotti
{"title":"A specific pluripotency-associated eRNA controls Nanog locus by shaping the epigenetic landscape and stabilizing enhancer-promoter interaction.","authors":"Mariella Cuomo,Davide Costabile,Rosa Della Monica,Michela Buonaiuto,Federica Trio,Giulia De Riso,Roberta Visconti,Lorenzo Chiariotti","doi":"10.1093/nar/gkaf274","DOIUrl":"https://doi.org/10.1093/nar/gkaf274","url":null,"abstract":"Despite a plethora of studies exploring the transcriptional regulation of the Nanog gene, the role of the enhancer RNAs (eRNAs) derived from Nanog-interacting super-enhancers (SEs) remains under-investigated. In the present study, we examined the functional role of the eRNAs transcribed from the -5 kb Nanog SE in mouse embryonic stem cells (mESCs) and found that an eRNA, here defined as -5KNAR, was essential to maintain the Nanog locus in an epigenetically active configuration, thereby ensuring pluripotency. We found that the here identified -5KNAR functionally interacts with the RAD21 protein, suggesting a role in stabilizing a cohesin complex at the Nanog locus, ensuring the generation and maintenance of an enhancer-promoter loop. Silencing of -5KNAR caused a cascade of events, including the generation of a DNA methylation wave (likely spreading from a single methylated CpG site), substantial chromatin remodeling, and loss of the enhancer-promoter loop, inducing Nanog silencing and mESC differentiation. Under these conditions, exogenous re-expression of Nanog was unable to restore either the endogenous Nanog expression or the enhancer-promoter interaction, suggesting that, at hierarchical level, the expression of the -5KNAR plays a prominent role in maintaining the pluripotency in mESCs.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":"1 1","pages":""},"PeriodicalIF":14.9,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143827127","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}