Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.869
Filter
2.
J Mol Cell Cardiol ; 191: 76-87, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718920

ABSTRACT

The reactivated adult epicardium produces epicardium-derived cells (EPDCs) via epithelial-mesenchymal transition (EMT) to benefit the recovery of the heart after myocardial infarction (MI). SMARCA4 is the core catalytic subunit of the chromatin re-modeling complex, which has the potential to target some reactivated epicardial genes in MI. However, the effects of epicardial SMARCA4 on MI remain uncertain. This study found that SMARCA4 was activated over time in epicardial cells following MI, and some of activated cells belonged to downstream differentiation types of EPDCs. This study used tamoxifen to induce lineage tracing and SMARCA4 deletion from epicardial cells in Wt1-CreER;Smarca4fl/fl;Rosa26-RFP adult mice. Epicardial SMARCA4 deletion reduces the number of epicardial cells in adult mice, which was related to changes in the activation, proliferation, and apoptosis of epicardial cells. Epicardial SMARCA4 deletion reduced collagen deposition and angiogenesis in the infarcted area, exacerbated cardiac injury in MI. The exacerbation of cardiac injury was related to the inhibition of generation and differentiation of EPDCs. The alterations in EPDCs were associated with inhibited transition between E-CAD and N-CAD during the epicardial EMT, coupled with the down-regulation of WT1, SNAIL1, and PDGF signaling. In conclusion, this study suggests that Epicardial SMARCA4 plays a critical role in cardiac injury caused by MI, and its regulatory mechanism is related to epicardial EMT. Epicardial SMARCA4 holds potential as a novel molecular target for treating MI.


Subject(s)
DNA Helicases , Epithelial-Mesenchymal Transition , Gene Deletion , Myocardial Infarction , Pericardium , Transcription Factors , Animals , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Epithelial-Mesenchymal Transition/genetics , Pericardium/pathology , Pericardium/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , Mice , Cell Differentiation , Apoptosis/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/deficiency , Cell Proliferation , Disease Models, Animal
3.
Front Immunol ; 15: 1358036, 2024.
Article in English | MEDLINE | ID: mdl-38690262

ABSTRACT

Background: It remains unclear whether BPIV3 infection leads to stress granules formation and whether G3BP1 plays a role in this process and in viral replication. This study aims to clarify the association between BPIV3 and stress granules, explore the effect of G3BP1 on BPIV3 replication, and provide significant insights into the mechanisms by which BPIV3 evades the host's antiviral immunity to support its own survival. Methods: Here, we use Immunofluorescence staining to observe the effect of BPIV3 infection on the assembly of stress granules. Meanwhile, the expression changes of eIF2α and G3BP1 were determined. Overexpression or siRNA silencing of intracellular G3BP1 levels was examined for its regulatory control of BPIV3 replication. Results: We identify that the BPIV3 infection elicited phosphorylation of the eIF2α protein. However, it did not induce the assembly of stress granules; rather, it inhibited the formation of stress granules and downregulated the expression of G3BP1. G3BP1 overexpression facilitated the formation of stress granules within cells and hindered viral replication, while G3BP1 knockdown enhanced BPIV3 expression. Conclusion: This study suggest that G3BP1 plays a crucial role in BPIV3 suppressing stress granule formation and viral replication.


Subject(s)
DNA Helicases , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins , Stress Granules , Virus Replication , Animals , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , RNA Recognition Motif Proteins/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , RNA Helicases/metabolism , RNA Helicases/genetics , Stress Granules/metabolism , Cattle , Eukaryotic Initiation Factor-2/metabolism , Respirovirus Infections/immunology , Respirovirus Infections/metabolism , Host-Pathogen Interactions/immunology , Phosphorylation , Cell Line , Cytoplasmic Granules/metabolism
4.
Nat Commun ; 15(1): 4127, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750080

ABSTRACT

Stress granules (SGs) are induced by various environmental stressors, resulting in their compositional and functional heterogeneity. SGs play a crucial role in the antiviral process, owing to their potent translational repressive effects and ability to trigger signal transduction; however, it is poorly understood how these antiviral SGs differ from SGs induced by other environmental stressors. Here we identify that TRIM25, a known driver of the ubiquitination-dependent antiviral innate immune response, is a potent and critical marker of the antiviral SGs. TRIM25 undergoes liquid-liquid phase separation (LLPS) and co-condenses with the SG core protein G3BP1 in a dsRNA-dependent manner. The co-condensation of TRIM25 and G3BP1 results in a significant enhancement of TRIM25's ubiquitination activity towards multiple antiviral proteins, which are mainly located in SGs. This co-condensation is critical in activating the RIG-I signaling pathway, thus restraining RNA virus infection. Our studies provide a conceptual framework for better understanding the heterogeneity of stress granule components and their response to distinct environmental stressors.


Subject(s)
DNA Helicases , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins , Signal Transduction , Stress Granules , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Ubiquitination , Humans , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , RNA Recognition Motif Proteins/metabolism , RNA Recognition Motif Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Stress Granules/metabolism , RNA Helicases/metabolism , DNA Helicases/metabolism , DEAD Box Protein 58/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Immunity, Innate , RNA, Double-Stranded/metabolism , HEK293 Cells , HeLa Cells , Cytoplasmic Granules/metabolism , RNA Virus Infections/virology , RNA Virus Infections/metabolism , RNA Virus Infections/immunology , Receptors, Immunologic/metabolism
5.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38717338

ABSTRACT

Senataxin is an evolutionarily conserved RNA-DNA helicase involved in DNA repair and transcription termination that is associated with human neurodegenerative disorders. Here, we investigated whether Senataxin loss affects protein homeostasis based on previous work showing R-loop-driven accumulation of DNA damage and protein aggregates in human cells. We find that Senataxin loss results in the accumulation of insoluble proteins, including many factors known to be prone to aggregation in neurodegenerative disorders. These aggregates are located primarily in the nucleolus and are promoted by upregulation of non-coding RNAs expressed from the intergenic spacer region of ribosomal DNA. We also map sites of R-loop accumulation in human cells lacking Senataxin and find higher RNA-DNA hybrids within the ribosomal DNA, peri-centromeric regions, and other intergenic sites but not at annotated protein-coding genes. These findings indicate that Senataxin loss affects the solubility of the proteome through the regulation of transcription-dependent lesions in the nucleus and the nucleolus.


Subject(s)
DNA Helicases , Multifunctional Enzymes , RNA Helicases , RNA, Untranslated , Humans , Cell Nucleolus/metabolism , Cell Nucleolus/genetics , DNA Damage , DNA Helicases/metabolism , DNA Helicases/genetics , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , Protein Aggregates , Proteostasis , R-Loop Structures/genetics , RNA Helicases/metabolism , RNA Helicases/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
6.
Genes (Basel) ; 15(5)2024 May 19.
Article in English | MEDLINE | ID: mdl-38790272

ABSTRACT

CHARGE syndrome, characterized by a distinct set of clinical features, has been linked primarily to mutations in the CHD7 gene. Initially defined by specific clinical criteria, including coloboma, heart defects, choanal atresia, delayed growth, and ear anomalies, CHARGE syndrome's diagnostic spectrum has broadened since the identification of CHD7. Variants in this gene exhibit considerable phenotypic variability, leading to the adoption of the term "CHD7 disorder" to encompass a wider range of associated symptoms. Recent research has identified CHD7 variants in individuals with isolated features such as autism spectrum disorder or gonadotropin-releasing hormone deficiency. In this study, we present three cases from two different families exhibiting audiovestibular impairment as the primary manifestation of a CHD7 variant. We discuss the expanding phenotypic variability observed in CHD7-related disorders, highlighting the importance of considering CHD7 in nonsyndromic hearing loss cases, especially when accompanied by inner ear malformations on MRI. Additionally, we underscore the necessity of genetic counseling and comprehensive clinical evaluation for individuals with CHD7 variants to ensure appropriate management of associated health concerns.


Subject(s)
CHARGE Syndrome , DNA Helicases , DNA-Binding Proteins , Humans , CHARGE Syndrome/genetics , CHARGE Syndrome/diagnosis , DNA Helicases/genetics , Male , DNA-Binding Proteins/genetics , Female , Mutation , Child , Adult , Phenotype , Pedigree , Child, Preschool , Adolescent
7.
Cancer Lett ; 592: 216929, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38697461

ABSTRACT

Small cell lung cancer (SCLC) is a recalcitrant cancer characterized by high frequency loss-of-function mutations in tumor suppressors with a lack of targeted therapy due to absence of high frequency gain-of-function abnormalities in oncogenes. SMARCAL1 is a member of the ATP-dependent chromatin remodeling protein SNF2 family that plays critical roles in DNA damage repair and genome stability maintenance. Here, we showed that SMARCAL1 was overexpressed in SCLC patient samples and was inversely associated with overall survival of the patients. SMARCAL1 was required for SCLC cell proliferation and genome integrity. Mass spectrometry revealed that PAR6B was a downstream SMARCAL1 signal molecule which rescued inhibitory effects caused by silencing of SMARCAL1. By screening of 36 FDA-approved clinically available agents related to DNA damage repair, we found that an aza-anthracenedione, pixantrone, was a potent SMARCAL1 inhibitor which suppressed the expression of SMARCAL1 and PAR6B at protein level. Pixantrone caused DNA damage and exhibited inhibitory effects on SCLC cells in vitro and in a patient-derived xenograft mouse model. These results indicated that SMARCAL1 functions as an oncogene in SCLC, and pixantrone as a SMARCAL1 inhibitor bears therapeutic potentials in this deadly disease.


Subject(s)
Cell Proliferation , DNA Helicases , Lung Neoplasms , Small Cell Lung Carcinoma , Xenograft Model Antitumor Assays , Humans , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , DNA Helicases/genetics , DNA Helicases/metabolism , Cell Proliferation/drug effects , Mice , Cell Line, Tumor , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , DNA Repair/drug effects
8.
J Gen Virol ; 105(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38572740

ABSTRACT

The herpes simplex virus 1 (HSV1) virion host shutoff (vhs) protein is an endoribonuclease that regulates the translational environment of the infected cell, by inducing the degradation of host mRNA via cellular exonuclease activity. To further understand the relationship between translational shutoff and mRNA decay, we have used ectopic expression to compare HSV1 vhs (vhsH) to its homologues from four other alphaherpesviruses - varicella zoster virus (vhsV), bovine herpesvirus 1 (vhsB), equine herpesvirus 1 (vhsE) and Marek's disease virus (vhsM). Only vhsH, vhsB and vhsE induced degradation of a reporter luciferase mRNA, with poly(A)+ in situ hybridization indicating a global depletion of cytoplasmic poly(A)+ RNA and a concomitant increase in nuclear poly(A)+ RNA and the polyA tail binding protein PABPC1 in cells expressing these variants. By contrast, vhsV and vhsM failed to induce reporter mRNA decay and poly(A)+ depletion, but rather, induced cytoplasmic G3BP1 and poly(A)+ mRNA- containing granules and phosphorylation of the stress response proteins eIF2α and protein kinase R. Intriguingly, regardless of their apparent endoribonuclease activity, all vhs homologues induced an equivalent general blockade to translation as measured by single-cell puromycin incorporation. Taken together, these data suggest that the activities of translational arrest and mRNA decay induced by vhs are separable and we propose that they represent sequential steps of the vhs host interaction pathway.


Subject(s)
Herpesvirus 1, Human , Viral Proteins , Viral Proteins/genetics , Viral Proteins/metabolism , Ribonucleases , DNA Helicases , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases , RNA Recognition Motif Proteins/metabolism , Herpesvirus 1, Human/genetics , Endoribonucleases/metabolism , RNA Stability , Virion/genetics , Virion/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38619323

ABSTRACT

Regulation of chromatin states is essential for proper temporal and spatial gene expression. Chromatin states are modulated by remodeling complexes composed of components that have enzymatic activities. CHD4 is the catalytic core of the nucleosome remodeling and deacetylase (NuRD) complex, which represses gene transcription. However, it remains to be determined how CHD4, a ubiquitous enzyme that remodels chromatin structure, functions in cardiomyocytes to maintain heart development. In particular, whether other proteins besides the NuRD components interact with CHD4 in the heart is controversial. Using quantitative proteomics, we identified that CHD4 interacts with SMYD1, a striated muscle-restricted histone methyltransferase that is essential for cardiomyocyte differentiation and cardiac morphogenesis. Comprehensive transcriptomic and chromatin accessibility studies of Smyd1 and Chd4 null embryonic mouse hearts revealed that SMYD1 and CHD4 repress a group of common genes and pathways involved in glycolysis, response to hypoxia, and angiogenesis. Our study reveals a mechanism by which CHD4 functions during heart development, and a previously uncharacterized mechanism regarding how SMYD1 represses cardiac transcription in the developing heart.


Subject(s)
DNA Helicases , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Heart , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Myocytes, Cardiac , Transcription Factors , Animals , Humans , Mice , Cell Differentiation/genetics , Chromatin/metabolism , Glycolysis/genetics , Heart/embryology , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice, Knockout , Muscle Proteins/metabolism , Muscle Proteins/genetics , Myocytes, Cardiac/metabolism , Proteomics , Transcription, Genetic
10.
J Mol Biol ; 436(11): 168589, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38677494

ABSTRACT

UvrD is a helicase vital for DNA replication and quality control processes. In its monomeric state, UvrD exhibits limited helicase activity, necessitating either dimerization or assistance from an accessory protein to efficiently unwind DNA. Within the DNA mismatch repair pathway, MutL plays a pivotal role in relaying the repair signal, enabling UvrD to unwind DNA from the strand incision site up to and beyond the mismatch. Although this interdependence is well-established, the precise mechanism of activation and the specific MutL-UvrD interactions that trigger helicase activity remain elusive. To address these questions, we employed site-specific crosslinking techniques using single-cysteine variants of MutL and UvrD followed by functional assays. Our investigation unveils that the C-terminal domain of MutL not only engages with UvrD but also acts as a self-sufficient activator of UvrD helicase activity on DNA substrates with 3'-single-stranded tails. Especially when MutL is covalently attached to the 2B or 1B domain the tail length can be reduced to a minimal substrate of 5 nucleotides without affecting unwinding efficiency.


Subject(s)
DNA Helicases , Escherichia coli Proteins , MutL Proteins , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/chemistry , MutL Proteins/metabolism , MutL Proteins/genetics , MutL Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Protein Binding , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Domains , DNA/metabolism , DNA/genetics
11.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(2): 184-192, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38686714

ABSTRACT

Objective To study the expression of SWI/SNF-related,matrix-associated,actin-dependent regulator of chromatin,subfamily A,member 4(SMARCA4)/Brahma-related gene 1,V-raf murine sarcoma viral oncogene homolog B(BRAF),P53,programmed cell death protein-1(PD-1),and programmed death-ligand 1(PD-L1),and changes in the expression of BRAF and neurotrophic tyrosine receptor kinase(NTRK) in the patients with colorectal cancer in Tibet,thereby providing a basis for targeted therapy and immunotherapy for this disease in Tibet. Methods A total of 64 patients with colorectal cancer resected in the Tibet Autonomous Region People's Hospital from January 2015 to July 2021 were enrolled in this study.The expression of SMARCA4,BRAF,P53,PD-1,and PD-L1 was detected by immunohistochemical staining.The gene fusion involving NTRK1,NTRK2,and NTRK3 was detected by fluorescence in situ hybridization,and the BRAF V600E gene mutation by polymerase chain reaction. Results The 64 patients with colorectal cancer were at a male-to-female ratio of 1.21∶1,with the mean age of (56.59±13.27) years.The tumors were located in the colon in 46(71.88%) patients and in the rectum in 18(28.12%) patients.Sixty(93.75%) patients presented adenocarcinoma,and 4(6.25%) patients presented other types of tumors.The patients in T1/T2 and T3/T4 phases accounted for 17.19%(n=11) and 82.81%(n=53),respectively.Lymph node metastasis occurred in 24(37.50%) patients.The immunohistochemical staining results showed partially down-regulated or absent expression of SMARCA4 in 1(1.56%) patient,positive BRAF expression in 4(6.25%) patients,and mutant expression of P53 in 35(54.69%) patients.The PD-1-expressing tumor associated immune cell was proportion score<10% in 45(70.31%) patients and≥10% in 19(29.69%) patients.The PD-L1 combined positive score was<10 in 52(81.25%) patients and≥10 in 12(18.75%) patients.The gene fusion of NTRK1,NTRK2,and NTRK3 was negative in all the patients,and BRAF V600E gene mutation was positive in 4(6.25%) patients.The SMARCA4 gene alteration was not detected in the patient with partial expression missing of SMARCA4.The PD-L1 combine positive score was correlated with the deficient mismatch repair(dMMR)/microsatellite instability-high (MSI-H) and the PD-1 expression (χ2=10.223,P=0.001;χ2=11.979,P=0.001). Conclusions The down-regulated or absent SMARCA4 expression and NTRK gene fusion are rare in the patients with colorectal cancer in Tibet.A few patients present BRAF V600E gene mutations,and Pan-TRK and BRAF expression can be used for the primary screening of NTRK gene fusion and BRAF gene mutation.The patients with dMMR/MSI-H are prone to high expression of PD-L1 and expected to benefit from immunotherapy.No significant correlation exists between P53 mutation and PD-L1 expression.The high expression of PD-1 is positively correlated with the high expression of PD-L1.


Subject(s)
B7-H1 Antigen , Biomarkers, Tumor , Colorectal Neoplasms , Immunotherapy , Proto-Oncogene Proteins B-raf , Humans , Male , Female , Middle Aged , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Tibet , Proto-Oncogene Proteins B-raf/genetics , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Transcription Factors/genetics , DNA Helicases/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Aged , Molecular Targeted Therapy , Mutation , Adult
12.
Plasmid ; 129-130: 102722, 2024.
Article in English | MEDLINE | ID: mdl-38631562

ABSTRACT

The predominant type of plasmids found in Acinetobacter species encode a Rep_3 initiation protein and many of these carry their accessory genes in dif modules. Here, available sequences of the 14 members of the group of Rep_3 plasmids typed as R3-T33, using a threshold of 95% identity in the repA gene, were compiled and compared. These plasmids were from various Acinetobacter species. The pdif sites were identified allowing the backbone and dif modules to be defined. As for other Rep_3 plasmids carrying dif modules, orfX encoding a protein of unknown function was found downstream of repA followed by a pdif site in the orientation XerC binding site-spacer-XerD binding site. Most backbones (n = 12) also included mobA and mobC genes but the two plasmids with the most diverged repA and orfX genes had different backbone contents. Although the gene content of the plasmid backbone was largely conserved, extensive recombinational exchange was detected and only two small groups carried identical or nearly identical backbones. Individual plasmids were associated with 1 to 13 dif modules. Many different dif modules were identified, including ones containing antibiotic or chromate resistance genes and several toxin/antitoxin gene pairs. In some cases, modules carrying the same genes were significantly diverged. Generally, the orientation of the pdif sites alternated such that C modules (XerC binding sites internal) alternated with D modules (XerD binding sites internal). However, fusions of two dif modules via mutational inactivation or loss of a pdif site were also detected.


Subject(s)
Acinetobacter , Plasmids , Acinetobacter/genetics , Plasmids/genetics , Plasmids/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , DNA, Bacterial/genetics , Base Sequence , Phylogeny , Trans-Activators/genetics , Trans-Activators/metabolism , DNA Helicases
14.
Expert Opin Ther Pat ; 34(3): 159-169, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38578210

ABSTRACT

INTRODUCTION: The multi-subunit SWI/SNF chromatin remodeling complex is a key epigenetic regulator for many cellular processes, and several subunits are found to be mutated in human cancers. The inactivating mutations of SMARCA4, the ATPase subunit of the complex, result in cellular dependency on the paralog SMARCA2 for survival. This observed synthetic lethal relationship posits targeting SMARCA2 in SMARCA4-deficient settings as an attractive therapeutic target in oncology. AREAS COVERED: This review covers patent literature disclosed during the 2019-30 June 2023 period which claim ATPase inhibitors and PROTAC degraders that bind to the ATPase domain of SMARCA2 and/or SMARCA4. A total of 16 documents from 6 applicants are presented. EXPERT OPINION: The demonstration of cellular dependence on SMARCA2 ATPase activity in SMARCA4-deficient settings has prompted substantial research toward SMARCA2-targeting therapies. Although selectively targeting the ATPase domain of SMARCA2 is viewed as challenging, several ATPase inhibitor scaffolds have been disclosed within the last five years. Most early compounds are weakly selective, but these efforts have culminated in the first dual SMARCA2/SMARCA4 ATPase inhibitor to enter clinical trials. Data from the ongoing clinical trials, as well as continued advancement of SMARCA2-selective ATPase inhibitors, are anticipated to significantly impact the field of therapies, targeting SMARCA4-deficient tumors.


Subject(s)
Antineoplastic Agents , DNA Helicases , Molecular Targeted Therapy , Neoplasms , Nuclear Proteins , Patents as Topic , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Antineoplastic Agents/pharmacology , DNA Helicases/metabolism , DNA Helicases/antagonists & inhibitors , DNA Helicases/genetics , Animals , Synthetic Lethal Mutations , Mutation , Adenosine Triphosphatases/metabolism
15.
Proc Natl Acad Sci U S A ; 121(19): e2317954121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38683976

ABSTRACT

Several microbial genomes lack textbook-defined essential genes. If an essential gene is absent from a genome, then an evolutionarily independent gene of unknown function complements its function. Here, we identified frequent nonhomologous replacement of an essential component of DNA replication initiation, a replicative helicase loader gene, in Vibrionaceae. Our analysis of Vibrionaceae genomes revealed two genes with unknown function, named vdhL1 and vdhL2, that were substantially enriched in genomes without the known helicase-loader genes. These genes showed no sequence similarities to genes with known function but encoded proteins structurally similar with a viral helicase loader. Analyses of genomic syntenies and coevolution with helicase genes suggested that vdhL1/2 encodes a helicase loader. The in vitro assay showed that Vibrio harveyi VdhL1 and Vibrio ezurae VdhL2 promote the helicase activity of DnaB. Furthermore, molecular phylogenetics suggested that vdhL1/2 were derived from phages and replaced an intrinsic helicase loader gene of Vibrionaceae over 20 times. This high replacement frequency implies the host's advantage in acquiring a viral helicase loader gene.


Subject(s)
DNA Helicases , DNA Replication , Phylogeny , Vibrionaceae , Vibrionaceae/genetics , Vibrionaceae/enzymology , DNA Helicases/metabolism , DNA Helicases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Bacteriophages/genetics , Bacteriophages/enzymology , Evolution, Molecular , Genome, Bacterial , DnaB Helicases/metabolism , DnaB Helicases/genetics , Vibrio/genetics , Vibrio/enzymology
16.
Vet Microbiol ; 293: 110070, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593624

ABSTRACT

Stress granules (SGs), the main component is GTPase-activating protein-binding protein 1 (G3BP1), which are assembled during viral infection and function to sequester host and viral mRNAs and proteins, are part of the antiviral responses. In this study, we found that porcine deltacoronavirus (PDCoV) infection induced stable formation of robust SGs in cells through a PERK (protein kinase R-like endoplasmic reticulum kinase)-dependent mechanism. Overexpression of SGs marker proteins G3BP1 significantly reduced PDCoV replication in vitro, while inhibition of endogenous G3BP1 enhanced PDCoV replication. Moreover, PDCoV infected LLC-PK1 cells raise the phosphorylation level of G3BP1. By overexpression of the G3BP1 phosphorylated protein or the G3BP1 dephosphorylated protein, we found that phosphorylation of G3BP1 is involved in the regulation of PDCoV-induced inflammatory response. Taken together, our study presents a vital aspect of the host innate response to invading pathogens and reveals attractive host targets for antiviral target.


Subject(s)
DNA Helicases , Inflammation , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins , Animals , Swine , RNA Recognition Motif Proteins/genetics , RNA Recognition Motif Proteins/metabolism , Phosphorylation , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/metabolism , RNA Helicases/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Virus Replication , Coronavirus/immunology , Coronavirus/physiology , Cell Line , Swine Diseases/virology , Swine Diseases/immunology , Swine Diseases/genetics , Immunity, Innate
17.
Mol Cell ; 84(9): 1684-1698.e9, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38593805

ABSTRACT

The Bloom syndrome (BLM) helicase is critical for alternative lengthening of telomeres (ALT), a homology-directed repair (HDR)-mediated telomere maintenance mechanism that is prevalent in cancers of mesenchymal origin. The DNA substrates that BLM engages to direct telomere recombination during ALT remain unknown. Here, we determine that BLM helicase acts on lagging strand telomere intermediates that occur specifically in ALT-positive cells to assemble a replication-associated DNA damage response. Loss of ATRX was permissive for BLM localization to ALT telomeres in S and G2, commensurate with the appearance of telomere C-strand-specific single-stranded DNA (ssDNA). DNA2 nuclease deficiency increased 5'-flap formation in a BLM-dependent manner, while telomere C-strand, but not G-strand, nicks promoted ALT. These findings define the seminal events in the ALT DNA damage response, linking aberrant telomeric lagging strand DNA replication with a BLM-directed HDR mechanism that sustains telomere length in a subset of human cancers.


Subject(s)
DNA Damage , DNA Replication , RecQ Helicases , Telomere Homeostasis , Telomere , RecQ Helicases/metabolism , RecQ Helicases/genetics , Humans , Telomere/metabolism , Telomere/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Bloom Syndrome/enzymology , Bloom Syndrome/pathology , Cell Line, Tumor
18.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38652118

ABSTRACT

Chromatin-remodeling protein BRG1/SMARCA4 is pivotal for establishing oligodendrocyte (OL) lineage identity. However, its functions for oligodendrocyte-precursor cell (OPC) differentiation within the postnatal brain and during remyelination remain elusive. Here, we demonstrate that Brg1 loss profoundly impairs OPC differentiation in the brain with a comparatively lesser effect in the spinal cord. Moreover, BRG1 is critical for OPC remyelination after injury. Integrative transcriptomic/genomic profiling reveals that BRG1 exhibits a dual role by promoting OPC differentiation networks while repressing OL-inhibitory cues and proneuronal programs. Furthermore, we find that BRG1 interacts with EED/PRC2 polycomb-repressive-complexes to enhance H3K27me3-mediated repression at gene loci associated with OL-differentiation inhibition and neurogenesis. Notably, BRG1 depletion decreases H3K27me3 deposition, leading to the upregulation of BMP/WNT signaling and proneurogenic genes, which suppresses OL programs. Thus, our findings reveal a hitherto unexplored spatiotemporal-specific role of BRG1 for OPC differentiation in the developing CNS and underscore a new insight into BRG1/PRC2-mediated epigenetic regulation that promotes and safeguards OL lineage commitment and differentiation.


Subject(s)
Cell Differentiation , DNA Helicases , Oligodendroglia , Polycomb Repressive Complex 2 , Animals , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , Epigenesis, Genetic , Histones/metabolism , Histones/genetics , Mice, Inbred C57BL , Neurogenesis/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/genetics , Remyelination , Transcription Factors/metabolism , Transcription Factors/genetics
19.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 40-44, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38678629

ABSTRACT

This study aimed to investigate the expression of long non-coding ribonucleic acid (lncRNA) DDX11 antisense RNA 1 (DDX11-AS1) in breast cancer (BC) tissues and cells and investigate its biological function and potential molecular mechanism through in vitro experiments. Tissue specimens were obtained from 44 BC patients. TRIzol method was used to extract RNAs from the tissues. The relative expression of DDX11-AS1 in BC tissues and the expression of DDX11-AS1 in BC cells were detected via quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The effect of DDX11-AS1 on the proliferation ability of BC cells was detected via cell counting kit-8 (CCK-8) assay. Flow cytometry was adopted to study the effect of DDX11-AS1 on the distribution of BC cell cycle. Transwell assays were performed to analyze the effects of DDX11-AS1 on the migration and invasion abilities of BC cells. Finally, after interfering with the expression of DDX11-AS1 in BC cells, changes in the expressions of molecular markers for epithelial-mesenchymal transition (EMT) were detected via Western blotting. According to the results of qRT-PCR, the expression of DDX11-AS1 was up-regulated in 38 out of 44 cases of BC tissues compared with that in the para-carcinoma tissues, and the expression of DDX11-AS1 in BC cells was up-regulated as well. After interference with the expression of DDX11-AS1 in BC cells, it was found via CCK-8 assay that the proliferation ability of BC cells was restrained, flow cytometry results showed that the BC cell cycle was arrested at G1/G0 phase, and the results of transwell assays revealed that the cell invasion and migration abilities were suppressed in experimental group compared with those in control group. According to the results of Western blotting, after interfering with the expression of DDX11-AS1 in BC cells, there were changes in the expressions of molecular markers for EMT. In BC, the expression of lncRNA DDX11-AS1 is up-regulated, which promotes the proliferation, migration and invasion of BC cells by regulating EMT.


Subject(s)
Breast Neoplasms , Cell Movement , Cell Proliferation , DNA Helicases , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness , RNA, Long Noncoding , Humans , Epithelial-Mesenchymal Transition/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Female , Cell Movement/genetics , Cell Proliferation/genetics , Neoplasm Invasiveness/genetics , Cell Line, Tumor , Middle Aged , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Cell Cycle/genetics
20.
Genes (Basel) ; 15(4)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38674442

ABSTRACT

(1) Background: Cockayne syndrome (CS) is an ultra-rare multisystem disorder, classically subdivided into three forms and characterized by a clinical spectrum without a clear genotype-phenotype correlation for both the two causative genes ERCC6 (CS type B) and ERCC8 (CS type A). We assessed this, presenting a series of patients with genetically confirmed CSB. (2) Materials and Methods: We retrospectively collected demographic, clinical, genetic, neuroimaging, and serum neurofilament light-chain (sNFL) data about CSB patients; diagnostic and severity scores were also determined. (3) Results: Data of eight ERCC6/CSB patients are presented. Four patients had CS I, three patients CS II, and one patient CS III. Various degrees of ataxia and spasticity were cardinal neurologic features, with variably combined systemic characteristics. Mean age at diagnosis was lower in the type II form, in which classic CS signs were more evident. Interestingly, sNFL determination appeared to reflect clinical classification. Two novel premature stop codon and one novel missense variants were identified. All CS I subjects harbored the p.Arg735Ter variant; the milder CS III subject carried the p.Leu764Ser missense change. (4) Conclusion: Our work confirms clinical variability also in the ERCC6/CSB type, where manifestations may range from severe involvement with prenatal or neonatal onset to normal psychomotor development followed by progressive ataxia. We propose, for the first time in CS, sNFL as a useful peripheral biomarker, with increased levels compared to currently available reference values and with the potential ability to reflect disease severity.


Subject(s)
Cockayne Syndrome , DNA Helicases , DNA Repair Enzymes , Poly-ADP-Ribose Binding Proteins , Transcription Factors , Humans , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , Cockayne Syndrome/diagnosis , Poly-ADP-Ribose Binding Proteins/genetics , DNA Repair Enzymes/genetics , Female , Male , DNA Helicases/genetics , Child , Child, Preschool , Adolescent , Retrospective Studies , Adult , Infant , Genetic Association Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...