Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 14.023
Filtrar
1.
Cell Commun Signal ; 22(1): 440, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39261837

RESUMO

BACKGROUND: Bivalent regions of chromatin (BvCR) are characterized by trimethylated lysine 4 (H3K4me3) and lysine 27 on histone H3 (H3K27me3) deposition which aid gene expression control during cell differentiation. The role of BvCR in post-transcriptional DNA damage response remains unidentified. Oncoprotein survivin binds chromatin and mediates IFNγ effects in CD4+ cells. In this study, we explored the role of BvCR in DNA damage response of autoimmune CD4+ cells in rheumatoid arthritis (RA). METHODS: We performed deep sequencing of the chromatin bound to survivin, H3K4me3, H3K27me3, and H3K27ac, in human CD4+ cells and identified BvCR, which possessed all three histone H3 modifications. Protein partners of survivin on chromatin were predicted by integration of motif enrichment analysis, computational machine-learning, and structural modeling, and validated experimentally by mass spectrometry and peptide binding array. Survivin-dependent change in BvCR and transcription of genes controlled by the BvCR was studied in CD4+ cells treated with survivin inhibitor, which revealed survivin-dependent biological processes. Finally, the survivin-dependent processes were mapped to the transcriptome of CD4+ cells in blood and in synovial tissue of RA patients and the effect of modern immunomodulating drugs on these processes was explored. RESULTS: We identified that BvCR dominated by H3K4me3 (H3K4me3-BvCR) accommodated survivin within cis-regulatory elements of the genes controlling DNA damage. Inhibition of survivin or JAK-STAT signaling enhanced H3K4me3-BvCR dominance, which improved DNA damage recognition and arrested cell cycle progression in cultured CD4+ cells. Specifically, BvCR accommodating survivin aided sequence-specific anchoring of the BRG1/SWI chromatin-remodeling complex coordinating DNA damage response. Mapping survivin interactome to BRG1/SWI complex demonstrated interaction of survivin with the subunits anchoring the complex to chromatin. Co-expression of BRG1, survivin and IFNγ in CD4+ cells rendered complete deregulation of DNA damage response in RA. Such cells possessed strong ability of homing to RA joints. Immunomodulating drugs inhibited the anchoring subunits of BRG1/SWI complex, which affected arthritogenic profile of CD4+ cells. CONCLUSIONS: BvCR execute DNA damage control to maintain genome fidelity in IFN-activated CD4+ cells. Survivin anchors the BRG1/SWI complex to BvCR to repress DNA damage response. These results offer a platform for therapeutic interventions targeting survivin and BRG1/SWI complex in autoimmunity.


Assuntos
Linfócitos T CD4-Positivos , Cromatina , Dano ao DNA , DNA Helicases , Proteínas Nucleares , Survivina , Fatores de Transcrição , Humanos , Survivina/metabolismo , Survivina/genética , Linfócitos T CD4-Positivos/metabolismo , Cromatina/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , DNA Helicases/metabolismo , DNA Helicases/genética , Histonas/metabolismo , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Artrite Reumatoide/genética
2.
Nat Commun ; 15(1): 8102, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39284827

RESUMO

Mammalian DNA replication relies on various DNA helicase and nuclease activities to ensure accurate genetic duplication, but how different helicase and nuclease activities are properly directed remains unclear. Here, we identify the ubiquitin-specific protease, USP50, as a chromatin-associated protein required to promote ongoing replication, fork restart, telomere maintenance, cellular survival following hydroxyurea or pyridostatin treatment, and suppression of DNA breaks near GC-rich sequences. We find that USP50 supports proper WRN-FEN1 localisation at or near stalled replication forks. Nascent DNA in cells lacking USP50 shows increased association of the DNA2 nuclease and RECQL4 and RECQL5 helicases and replication defects in cells lacking USP50, or FEN1 are driven by these proteins. Consequently, suppression of DNA2 or RECQL4/5 improves USP50-depleted cell resistance to agents inducing replicative stress and restores telomere stability. These data define an unexpected regulatory protein that promotes the balance of helicase and nuclease use at ongoing and stalled replication forks.


Assuntos
DNA Helicases , Replicação do DNA , RecQ Helicases , Helicase da Síndrome de Werner , RecQ Helicases/metabolismo , RecQ Helicases/genética , Replicação do DNA/efeitos dos fármacos , Humanos , Helicase da Síndrome de Werner/metabolismo , Helicase da Síndrome de Werner/genética , DNA Helicases/metabolismo , DNA Helicases/genética , Telômero/metabolismo , Telômero/genética , Endonucleases Flap/metabolismo , Endonucleases Flap/genética , Proteases Específicas de Ubiquitina/metabolismo , Proteases Específicas de Ubiquitina/genética , Células HeLa , Células HEK293 , Homeostase do Telômero/efeitos dos fármacos , Cromatina/metabolismo
3.
Cells ; 13(17)2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39273031

RESUMO

DNA double strand breaks (DSBs) are critical for the efficacy of radiotherapy as they lead to cell death if not repaired. DSBs caused by ionizing radiation (IR) initiate histone modifications and accumulate DNA repair proteins, including 53BP1, which forms distinct foci at damage sites and serves as a marker for DSBs. DSB repair primarily occurs through Non-Homologous End Joining (NHEJ) and Homologous Recombination (HR). NHEJ directly ligates DNA ends, employing proteins such as DNA-PKcs, while HR, involving proteins such as Rad54, uses a sister chromatid template for accurate repair and functions in the S and G2 phases of the cell cycle. Both pathways are crucial, as illustrated by the IR sensitivity in cells lacking DNA-PKcs or Rad54. We generated mouse embryonic stem (mES) cells which are knockout (KO) for DNA-PKcs and Rad54 to explore the combined role of HR and NHEJ in DSB repair. We found that cells lacking both DNA-PKcs and Rad54 are hypersensitive to X-ray radiation, coinciding with impaired 53BP1 focus resolution and a more persistent G2 phase cell cycle block. Additionally, mES cells deficient in DNA-PKcs or both DNA-PKcs and Rad54 exhibit an increased nuclear size approximately 18-24 h post-irradiation. To further explore the role of Rad54 in the absence of DNA-PKcs, we generated DNA-PKcs KO mES cells expressing GFP-tagged wild-type (WT) or ATPase-defective Rad54 to track the Rad54 foci over time post-irradiation. Cells lacking DNA-PKcs and expressing ATPase-defective Rad54 exhibited a similar phenotypic response to IR as those lacking both DNA-PKcs and Rad54. Despite a strong G2 phase arrest, live-cell imaging showed these cells eventually progress through mitosis, forming micronuclei. Additionally, mES cells lacking DNA-PKcs showed increased Rad54 foci over time post-irradiation, indicating an enhanced reliance on HR for DSB repair without DNA-PKcs. Our findings underscore the essential roles of HR and NHEJ in maintaining genomic stability post-IR in mES cells. The interplay between these pathways is crucial for effective DSB repair and cell cycle progression, highlighting potential targets for enhancing radiotherapy outcomes.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Recombinação Homóloga , Células-Tronco Embrionárias Murinas , Radiação Ionizante , Animais , Camundongos , Reparo do DNA por Junção de Extremidades/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/efeitos da radiação , Células-Tronco Embrionárias Murinas/citologia , Recombinação Homóloga/efeitos da radiação , Proteína Quinase Ativada por DNA/metabolismo , Proteína Quinase Ativada por DNA/genética , DNA Helicases/metabolismo , DNA Helicases/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteínas Nucleares
4.
PLoS One ; 19(9): e0309713, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39240887

RESUMO

The Regulator of Telomere Helicase 1 (RTEL1) gene encodes a critical DNA helicase intricately involved in the maintenance of telomeric structures and the preservation of genomic stability. Germline mutations in the RTEL1 gene have been clinically associated with Hoyeraal-Hreidarsson syndrome, a more severe version of Dyskeratosis Congenita. Although various research has sought to link RTEL1 mutations to specific disorders, no comprehensive investigation has yet been conducted on missense mutations. In this study, we attempted to investigate the functionally and structurally deleterious coding and non-coding SNPs of the RTEL1 gene using an in silico approach. Initially, out of 1392 nsSNPs, 43 nsSNPs were filtered out through ten web-based bioinformatics tools. With subsequent analysis using nine in silico tools, these 43 nsSNPs were further shortened to 11 most deleterious nsSNPs. Furthermore, analyses of mutated protein structures, evolutionary conservancy, surface accessibility, domains & PTM sites, cancer susceptibility, and interatomic interaction revealed the detrimental effect of these 11 nsSNPs on RTEL1 protein. An in-depth investigation through molecular docking with the DNA binding sequence demonstrated a striking change in the interaction pattern for F15L, M25V, and G706R mutant proteins, suggesting the more severe consequences of these mutations on protein structure and functionality. Among the non-coding variants, two had the highest likelihood of being regulatory variants, whereas one variant was predicted to affect the target region of a miRNA. Thus, this study lays the groundwork for extensive analysis of RTEL1 gene variants in the future, along with the advancement of precision medicine and other treatment modalities.


Assuntos
Simulação por Computador , DNA Helicases , Polimorfismo de Nucleotídeo Único , Humanos , DNA Helicases/genética , DNA Helicases/química , Simulação de Acoplamento Molecular , Mutação de Sentido Incorreto , Biologia Computacional/métodos , Disceratose Congênita/genética , Predisposição Genética para Doença
5.
Nat Commun ; 15(1): 7646, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39223123

RESUMO

Despite their prevalent cancer implications, the in vivo dynamics of SWI/SNF chromatin remodelers and how misregulation of such dynamics underpins cancer remain poorly understood. Using live-cell single-molecule tracking, we quantify the intranuclear diffusion and chromatin-binding of three key subunits common to all major human SWI/SNF remodeler complexes (BAF57, BAF155 and BRG1), and resolve two temporally distinct stable binding modes for the fully assembled complex. Super-resolved density mapping reveals heterogeneous, nanoscale remodeler binding "hotspots" across the nucleoplasm where multiple binding events (especially longer-lived ones) preferentially cluster. Importantly, we uncover distinct roles of the bromodomain in modulating chromatin binding/targeting in a DNA-accessibility-dependent manner, pointing to a model where successive longer-lived binding within "hotspots" leads to sustained productive remodeling. Finally, systematic comparison of six common BRG1 mutants implicated in various cancers unveils alterations in chromatin-binding dynamics unique to each mutant, shedding insight into a multi-modal landscape regulating the spatio-temporal organizational dynamics of SWI/SNF remodelers.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina , Proteínas Cromossômicas não Histona , DNA Helicases , Neoplasias , Proteínas Nucleares , Imagem Individual de Molécula , Fatores de Transcrição , Humanos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Imagem Individual de Molécula/métodos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , DNA Helicases/metabolismo , DNA Helicases/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Cromossômicas não Histona/genética , Cromatina/metabolismo , Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patologia , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Ligação Proteica , Mutação , Linhagem Celular Tumoral , Domínios Proteicos , Adenosina Trifosfatases
6.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(9): 1053-1058, 2024 Sep 10.
Artigo em Chinês | MEDLINE | ID: mdl-39217482

RESUMO

OBJECTIVE: To explore the clinical phenotype and genetic etiology for seven children with CHARGE syndrome (CS). METHODS: Clinical data of seven children with CS diagnosed between March 2020 and December 2022 at the Children's Hospital Affiliated to Zhengzhou University were analyzed. Genomic DNA was extracted from peripheral blood samples from the children and their parents, and subjected to whole exome sequencing. Candidate variants were verified by Sanger sequencing and pathogenicity analysis. RESULTS: The ages of the children had ranged from 1 day after birth to 12 years old, and all of them had shown growth retardation. The reasons for their admission had included postnatal breathing, swallowing and feeding difficulties in five cases. One child was found to have abnormal external genitalia in conjunct with hearing impairment, whilst another child had shown no secondary sexual characteristics during puberty. All of the children were found to harbor CHD7 gene variants, which included 3 nonsense variants, 2 frameshifting variants and 2 missense variants, i.e., c.6292C>T (p.R2098*), c.2754G>A (p.W918*), c.469C>T (p.R157*), c.3308T>A (p.V1103D), c.7111delC (p.Q2371Kfs), c.6023delA (p.D2008Vfs) and c.3565C>T (p.R1189C). All of the variants were de novo in origin. Based on the guidelines from the American College of Medical Genetics and Genomics (ACMG), the c.3308T>A (p.V1103D) and c.3565C>T (p.R1189C) variants were rated as likely pathogenic (PS2+PM2_Supporting+PP3), whilst the remainders were rated as pathogenic (PVS1+PS2+PM2_Supporting). CONCLUSION: There is strong clinical and genetic heterogeneity in CS. Early genetic testing may facilitate accurate diagnosis. The detection of novel variants has expanded the phenotypic spectrum of CS and the mutational spectrum of the CHD7 gene.


Assuntos
Síndrome CHARGE , DNA Helicases , Proteínas de Ligação a DNA , Sequenciamento do Exoma , Fenótipo , Humanos , Síndrome CHARGE/genética , Criança , Masculino , Pré-Escolar , Feminino , Lactente , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Recém-Nascido , Mutação
7.
Protein Sci ; 33(9): e5093, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39180489

RESUMO

RTEL1 is an essential DNA helicase which plays an important role in various aspects of genome stability, from telomere metabolism to DNA replication, repair and recombination. RTEL1 has been implicated in a number of genetic diseases and cancer development, including glioma, breast, lung and gastrointestinal tumors. RTEL1 is a FeS helicase but, in addition to the helicase core, it comprises a long C-terminal region which includes a number of folded domains connected by intrinsically disordered loops and mediates RTEL1 interaction with factors involved in pivotal cellular pathways. However, information on the architecture and the function of this region is still limited. We expressed and purified a variety of fragments encompassing the folded domains and the unstructured regions. We determined the crystal structure of the second repeat, confirming that it has a fold similar to the harmonin homology domains. SAXS data provide low-resolution information on all the fragments and suggest that the presence of the RING domain affects the overall architecture of the C-terminal region, making the structure significantly more compact. NMR data provide experimental information on the interaction between PCNA and the RTEL1 C-terminal region, revealing a putative low-affinity additional site of interaction. A biochemical analysis shows that the C-terminal region, in addition to a preference for telomeric RNA and DNA G-quadruplexes, has a high affinity for R-loops and D-loops, consistent with the role played by the RTEL1 helicase in homologous recombination, telomere maintenance and preventing replication-transcription conflicts. We further dissected the contribution of each domain in binding different substrates.


Assuntos
DNA Helicases , Humanos , DNA Helicases/química , DNA Helicases/metabolismo , DNA Helicases/genética , Cristalografia por Raios X , Modelos Moleculares , Antígeno Nuclear de Célula em Proliferação/química , Antígeno Nuclear de Célula em Proliferação/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Domínios Proteicos , Espalhamento a Baixo Ângulo
8.
Cell Death Dis ; 15(8): 621, 2024 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-39187513

RESUMO

Despite advancements in chemotherapy and the availability of novel therapies, the outcome of adult patients with B-cell acute lymphoblastic leukemia (B-ALL) remains unsatisfactory. Therefore, it is necessary to understand the molecular mechanisms underlying the progression of B-ALL. Brahma-related gene 1 (BRG1) is a poor prognostic factor for multiple cancers. Here, the expression of BRG1 was found to be higher in patients with B-ALL, irrespective of the molecular subtype, than in healthy individuals, and its overexpression was associated with a poor prognosis. Upregulation of BRG1 accelerated cell cycle progression into the S phase, resulting in increased cell proliferation, whereas its downregulation facilitated the apoptosis of B-ALL cells. Mechanistically, BRG1 occupies the transcriptional activation site of PPP2R1A, thereby inhibiting its expression and activating the PI3K/AKT signaling pathway to regulate the proto-oncogenes c-Myc and BCL-2. Consistently, silencing of BRG1 and administration of PFI-3 (a specific inhibitor targeting BRG1) significantly inhibited the progression of leukemia and effectively prolonged survival in cell-derived xenograft mouse models of B-ALL. Altogether, this study demonstrates that BRG1-induced overactivation of the PPP2R1A/PI3K/AKT signaling pathway plays an important role in promoting the progression of B-ALL. Therefore, targeting BRG1 represents a promising strategy for the treatment of B-ALL in adults.


Assuntos
DNA Helicases , Progressão da Doença , Proteínas Nucleares , Proteína Fosfatase 2 , Fatores de Transcrição , Animais , Feminino , Humanos , Masculino , Camundongos , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA Helicases/metabolismo , DNA Helicases/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Proteína Fosfatase 2/metabolismo , Proteína Fosfatase 2/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos
9.
DNA Repair (Amst) ; 141: 103731, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39089193

RESUMO

DNA replication is remarkably accurate with estimates of only a handful of mutations per human genome per cell division cycle. Replication stress caused by DNA lesions, transcription-replication conflicts, and other obstacles to the replication machinery must be efficiently overcome in ways that minimize errors and maximize completion of DNA synthesis. Replication fork reversal is one mechanism that helps cells tolerate replication stress. This process involves reannealing of parental template DNA strands and generation of a nascent-nascent DNA duplex. While fork reversal may be beneficial by facilitating DNA repair or template switching, it must be confined to the appropriate contexts to preserve genome stability. Many enzymes have been implicated in this process including ATP-dependent DNA translocases like SMARCAL1, ZRANB3, HLTF, and the helicase FBH1. In addition, the RAD51 recombinase is required. Many additional factors and regulatory activities also act to ensure reversal is beneficial instead of yielding undesirable outcomes. Finally, reversed forks must also be stabilized and often need to be restarted to complete DNA synthesis. Disruption or deregulation of fork reversal causes a variety of human diseases. In this review we will describe the latest models for reversal and key mechanisms of regulation.


Assuntos
Tolerância ao Dano no DNA , Animais , Humanos , DNA/metabolismo , DNA Helicases/metabolismo , DNA Helicases/genética , Reparo do DNA , Instabilidade Genômica
10.
Nat Commun ; 15(1): 7076, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39152113

RESUMO

During the repair of interstrand crosslinks (ICLs) a DNA double-strand break (DSB) is generated. The Fanconi anemia (FA) core complex, which is recruited to ICLs, promotes high-fidelity repair of this DSB by homologous recombination (HR). However, whether the FA core complex also promotes HR at ICL-independent DSBs, for example induced by ionizing irradiation or nucleases, remains controversial. Here, we identified the FA core complex members FANCL and Ube2T as HR-promoting factors in a CRISPR/Cas9-based screen. Using isogenic cell line models, we further demonstrated an HR-promoting function of FANCL and Ube2T, and of their ubiquitination substrate FANCD2. We show that FANCL and Ube2T localize at DSBs in a FANCM-dependent manner, and are required for the DSB accumulation of FANCD2. Mechanistically, we demonstrate that FANCL ubiquitin ligase activity is required for the accumulation of CtIP at DSBs, thereby promoting end resection and Rad51 loading. Together, these data demonstrate a dual genome maintenance function of the FA core complex and FANCD2 in promoting repair of both ICLs and DSBs.


Assuntos
Quebras de DNA de Cadeia Dupla , Proteína do Grupo de Complementação D2 da Anemia de Fanconi , Proteína do Grupo de Complementação L da Anemia de Fanconi , Recombinação Homóloga , Enzimas de Conjugação de Ubiquitina , Humanos , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteína do Grupo de Complementação L da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação L da Anemia de Fanconi/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/genética , Sistemas CRISPR-Cas , Ubiquitinação , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Endodesoxirribonucleases/metabolismo , Endodesoxirribonucleases/genética , Células HEK293 , Reparo de DNA por Recombinação , Reparo do DNA , Reparo do DNA por Junção de Extremidades , DNA Helicases
11.
J Clin Immunol ; 44(8): 180, 2024 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-39153074

RESUMO

Schimke immuno-osseous dysplasia is a rare multisystemic disorder caused by biallelic loss of function of the SMARCAL1 gene that plays a pivotal role in replication fork stabilization and thus DNA repair. Individuals affected from this disease suffer from disproportionate growth failure, steroid resistant nephrotic syndrome leading to renal failure and primary immunodeficiency mediated by T cell lymphopenia. With infectious complications being the leading cause of death in this disease, researching the nature of the immunodeficiency is crucial, particularly as the state is exacerbated by loss of antibodies due to nephrotic syndrome or immunosuppressive treatment. Building on previous findings that identified the loss of IL-7 receptor expression as a possible cause of the immunodeficiency and increased sensitivity to radiation-induced damage, we have employed spectral cytometry and multiplex RNA-sequencing to assess the phenotype and function of T cells ex-vivo and to study changes induced by in-vitro UV irradiation and reaction of cells to the presence of IL-7. Our findings highlight the mature phenotype of T cells with proinflammatory Th1 skew and signs of exhaustion and lack of response to IL-7. UV light irradiation caused a severe increase in the apoptosis of T cells, however the expression of the genes related to immune response and regulation remained surprisingly similar to healthy cells. Due to the disease's rarity, more studies will be necessary for complete understanding of this unique immunodeficiency.


Assuntos
Reparo do DNA , Osteocondrodisplasias , Doenças da Imunodeficiência Primária , Humanos , Doenças da Imunodeficiência Primária/genética , Doenças da Imunodeficiência Primária/diagnóstico , Doenças da Imunodeficiência Primária/imunologia , Osteocondrodisplasias/genética , Osteocondrodisplasias/imunologia , Reparo do DNA/genética , DNA Helicases/genética , Síndrome Nefrótica/etiologia , Síndrome Nefrótica/genética , Linfócitos T/imunologia , Arteriosclerose/genética , Arteriosclerose/etiologia , Arteriosclerose/imunologia , Masculino , Feminino , Embolia Pulmonar/genética , Embolia Pulmonar/etiologia , Doenças Ósseas Metabólicas/etiologia , Doenças Ósseas Metabólicas/genética , Transtornos do Crescimento/genética , Transtornos do Crescimento/etiologia , Raios Ultravioleta/efeitos adversos , Criança , Apoptose/genética , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia
13.
Sci Adv ; 10(34): eadl1150, 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39167653

RESUMO

An outbreak of mpox virus in May 2022 has spread over 110 nonpandemic regions in the world, posing a great threat to global health. Mpox virus E5, a helicase-primase, plays an essential role in DNA replication, but the molecular mechanisms are elusive. Here, we report seven structures of mpox virus E5 in a double hexamer (DH) and six in single hexamer in different conformations, indicating a rotation mechanism for helicase and a coupling action for primase. The DH is formed through the interface of zinc-binding domains, and the central channel density indicates potential double-stranded DNA (dsDNA), which helps to identify dsDNA binding residues Arg249, Lys286, Lys315, and Lys317. Our work is important not only for understanding poxviral DNA replication but also for the development of novel therapeutics for serious poxviral infections including smallpox virus and mpox virus.


Assuntos
DNA Helicases , DNA Primase , DNA Primase/metabolismo , DNA Primase/química , DNA Helicases/metabolismo , DNA Helicases/química , Modelos Moleculares , Proteínas Virais/metabolismo , Proteínas Virais/química , Proteínas Virais/genética , Multimerização Proteica , Replicação do DNA , Ligação Proteica , DNA Viral/metabolismo
15.
Virol J ; 21(1): 186, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39135075

RESUMO

BACKGROUND: The global outbreak of COVID-19 caused by the SARS-CoV-2 has led to millions of deaths. This unanticipated emergency has prompted virologists across the globe to delve deeper into the intricate dynamicity of the host-virus interface with an aim to identify antiviral targets and elucidate host and viral determinants of severe disease. AIM: The present study was undertaken to analyse the role of histone deacetylase 6 (HDAC6) in regulating SARS-CoV-2 infection. RESULTS: Gradual increase in HDAC6 expression was observed in different SARS-CoV-2-permissive cell lines following SARS-CoV-2 infection. The SARS-CoV-2 nucleocapsid protein (N protein) was identified as the primary viral factor responsible for upregulating HDAC6 expression. Downregulation of HDAC6 using shRNA or a specific inhibitor tubacin resulted in reduced viral replication suggesting proviral role of its deacetylase activity. Further investigations uncovered the interaction of HDAC6 with stress granule protein G3BP1 and N protein during infection. HDAC6-mediated deacetylation of SARS-CoV-2 N protein was found to be crucial for its association with G3BP1. CONCLUSION: This study provides valuable insights into the molecular mechanisms underlying the disruption of cytoplasmic stress granules during SARS-CoV-2 infection and highlights the significance of HDAC6 in the process.


Assuntos
COVID-19 , Proteínas do Nucleocapsídeo de Coronavírus , Desacetilase 6 de Histona , SARS-CoV-2 , Replicação Viral , Desacetilase 6 de Histona/metabolismo , Desacetilase 6 de Histona/genética , Humanos , SARS-CoV-2/fisiologia , Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , Proteínas do Nucleocapsídeo de Coronavírus/genética , COVID-19/virologia , COVID-19/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Acetilação , Linhagem Celular , Chlorocebus aethiops , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Células Vero , Animais , Interações Hospedeiro-Patógeno , Proteínas de Ligação a Poli-ADP-Ribose , DNA Helicases , RNA Helicases
16.
J Vis Exp ; (209)2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39141566

RESUMO

Eukaryotes have one replicative helicase known as CMG, which centrally organizes and drives the replisome, and leads the way at the front of replication forks. Obtaining a deep mechanistic understanding of the dynamics of CMG is critical to elucidating how cells achieve the enormous task of efficiently and accurately replicating their entire genome once per cell cycle. Single-molecule techniques are uniquely suited to quantify the dynamics of CMG due to their unparalleled temporal and spatial resolution. Nevertheless, single-molecule studies of CMG motion have thus far relied on pre-formed CMG purified from cells as a complex, which precludes the study of the steps leading up to its activation. Here, we describe a hybrid ensemble and single-molecule assay that allowed imaging at the single-molecule level of the motion of fluorescently labeled CMG after fully reconstituting its assembly and activation from 36 different purified S. cerevisiae polypeptides. This assay relies on the double functionalization of the ends of a linear DNA substrate with two orthogonal attachment moieties, and can be adapted to study similarly complex DNA-processing mechanisms at the single-molecule level.


Assuntos
Saccharomyces cerevisiae , Imagem Individual de Molécula , Saccharomyces cerevisiae/metabolismo , Imagem Individual de Molécula/métodos , DNA Helicases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Corantes Fluorescentes/química , Replicação do DNA , DNA Fúngico/genética
17.
Cell Mol Life Sci ; 81(1): 339, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39120648

RESUMO

Senataxin is an evolutionarily conserved DNA/RNA helicase, whose dysfunctions are linked to neurodegeneration and cancer. A main activity of this protein is the removal of R-loops, which are nucleic acid structures capable to promote DNA damage and replication stress. Here we found that Senataxin deficiency causes the release of damaged DNA into extranuclear bodies, called micronuclei, triggering the massive recruitment of cGAS, the apical sensor of the innate immunity pathway, and the downstream stimulation of interferon genes. Such cGAS-positive micronuclei are characterized by defective membrane envelope and are particularly abundant in cycling cells lacking Senataxin, but not after exposure to a DNA breaking agent or in absence of the tumor suppressor BRCA1 protein, a partner of Senataxin in R-loop removal. Micronuclei with a discontinuous membrane are normally cleared by autophagy, a process that we show is impaired in Senataxin-deficient cells. The formation of Senataxin-dependent inflamed micronuclei is promoted by the persistence of nuclear R-loops stimulated by the DSIF transcription elongation complex and the engagement of EXO1 nuclease activity on nuclear DNA. Coherently, high levels of EXO1 result in poor prognosis in a subset of tumors lacking Senataxin expression. Hence, R-loop homeostasis impairment, together with autophagy failure and unscheduled EXO1 activity, elicits innate immune response through micronuclei formation in cells lacking Senataxin.


Assuntos
Autofagia , Dano ao DNA , DNA Helicases , Inflamação , Enzimas Multifuncionais , Nucleotidiltransferases , Estruturas R-Loop , RNA Helicases , Humanos , Autofagia/genética , Proteína BRCA1/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/deficiência , DNA Helicases/metabolismo , DNA Helicases/genética , DNA Helicases/deficiência , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/deficiência , Exodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/genética , Imunidade Inata , Inflamação/patologia , Inflamação/metabolismo , Inflamação/genética , Enzimas Multifuncionais/metabolismo , Enzimas Multifuncionais/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Fosfoproteínas , RNA Helicases/metabolismo , RNA Helicases/genética
18.
Proc Natl Acad Sci U S A ; 121(33): e2318601121, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39116123

RESUMO

Serial capture affinity purification (SCAP) is a powerful method to isolate a specific protein complex. When combined with cross-linking mass spectrometry and computational approaches, one can build an integrated structural model of the isolated complex. Here, we applied SCAP to dissect a subpopulation of WDR76 in complex with SPIN1, a histone reader that recognizes trimethylated histone H3 lysine4 (H3K4me3). In contrast to a previous SCAP analysis of the SPIN1:SPINDOC complex, histones and the H3K4me3 mark were enriched with the WDR76:SPIN1 complex. Next, interaction network analysis of copurifying proteins and microscopy analysis revealed a potential role of the WDR76:SPIN1 complex in the DNA damage response. Since we detected 149 pairs of cross-links between WDR76, SPIN1, and histones, we then built an integrated structural model of the complex where SPIN1 recognized the H3K4me3 epigenetic mark while interacting with WDR76. Finally, we used the powerful Bayesian Integrative Modeling approach as implemented in the Integrative Modeling Platform to build a model of WDR76 and SPIN1 bound to the nucleosome.


Assuntos
Dano ao DNA , Histonas , Nucleossomos , Histonas/metabolismo , Histonas/química , Nucleossomos/metabolismo , Humanos , Ligação Proteica , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/química , Modelos Moleculares , ATPases Associadas a Diversas Atividades Celulares , DNA Helicases
19.
Nat Commun ; 15(1): 7092, 2024 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-39154037

RESUMO

Mammalian TIP60 is a multi-functional enzyme with histone acetylation and histone dimer exchange activities. It plays roles in diverse cellular processes including transcription, DNA repair, cell cycle control, and embryonic development. Here we report the cryo-electron microscopy structures of the human TIP60 complex with the core subcomplex and TRRAP module refined to 3.2-Å resolution. The structures show that EP400 acts as a backbone integrating the motor module, the ARP module, and the TRRAP module. The RUVBL1-RUVBL2 hexamer serves as a rigid core for the assembly of EP400 ATPase and YL1 in the motor module. In the ARP module, an ACTL6A-ACTB heterodimer and an extra ACTL6A make hydrophobic contacts with EP400 HSA helix, buttressed by network interactions among DMAP1, EPC1, and EP400. The ARP module stably associates with the motor module but is flexibly tethered to the TRRAP module, exhibiting a unique feature of human TIP60. The architecture of the nucleosome-bound human TIP60 reveals an unengaged nucleosome that is located between the core subcomplex and the TRRAP module. Our work illustrates the molecular architecture of human TIP60 and provides architectural insights into how this complex is bound by the nucleosome.


Assuntos
Microscopia Crioeletrônica , Lisina Acetiltransferase 5 , Humanos , Lisina Acetiltransferase 5/metabolismo , Lisina Acetiltransferase 5/química , Lisina Acetiltransferase 5/genética , Nucleossomos/metabolismo , Nucleossomos/ultraestrutura , Nucleossomos/química , DNA Helicases/metabolismo , DNA Helicases/química , Modelos Moleculares , ATPases Associadas a Diversas Atividades Celulares/metabolismo , ATPases Associadas a Diversas Atividades Celulares/química , ATPases Associadas a Diversas Atividades Celulares/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/química , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/química , Ligação Proteica , Multimerização Proteica , Proteínas que Contêm Bromodomínio , Proteínas Adaptadoras de Transdução de Sinal
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(8): 962-965, 2024 Aug 10.
Artigo em Chinês | MEDLINE | ID: mdl-39097280

RESUMO

OBJECTIVE: To explore the genetic basis for child with CHARGE syndrome. METHODS: A child who was diagnosed at Ningbo Women and Children's Hospital on September 29, 2022 was selected as the study subject. Relevant clinical data were collected. The child and her parents were subjected to whole exome sequencing (WES), and candidate variant was verified by Sanger sequencing and bioinformatic analysis. RESULTS: The child was found to harbor a de novo c.2972T>C (p.L991S) missense variant of the CHD7 gene, which was detected in neither of her parents. Based on the guidelines from the American College of Medical Genetics and Genomics (ACMG), the variant was predicted to be likely pathogenic (PM6+PM2_Supporting+PP2+PP3+PP4). Bioinformatic analysis predicted that amino acid 991 is highly conserved among various species, and a hydrogen bond has formed between Asp993 and the mutant Ser991. CONCLUSION: The heterozygous c.2972T>C (p.L991S) missense variant of the CHD7 gene probably underlay the pathogenesis of CHARGE syndrome in this child. Above finding has also enriched the mutational spectrum for CHARGE syndrome.


Assuntos
Síndrome CHARGE , DNA Helicases , Proteínas de Ligação a DNA , Mutação de Sentido Incorreto , Humanos , Síndrome CHARGE/genética , DNA Helicases/genética , Feminino , Proteínas de Ligação a DNA/genética , Sequenciamento do Exoma , Lactente , Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA