Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.444
Filter
1.
Medicine (Baltimore) ; 103(31): e39045, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093728

ABSTRACT

RATIONALE: SMARCA4-deficient undifferentiated tumor (SMARCA4-UT) is a recently reported rare malignancy that can rapidly metastasize to tissues and organs throughout the body. The tumor is characterized by a lower response to platinum-based chemotherapy. More regrettably, the mean survival time of patients with this disease after diagnosis is only 4 to 7 months. PATIENT CONCERNS: A 58-year-old man was admitted to a hospital for fatigue, sudden syncope, and a mass-like shadow of his left upper lobe demonstrated by a pulmonary computed tomographic. Based on his subsequent clinical and pathological features, he was highly suspected of SMARCA4-UT. DIAGNOSES: Combined with next-generation sequencing genetic testing and immunohistochemical examination results, the patient was diagnosed with SMARCA4-UT. INTERVENTIONS: The patient received a left upper lobectomy and lymph node dissection, four-course chemotherapy divided into 8 sessions with the use of paclitaxel simply, and a proper post-discharge self-care. OUTCOMES: The patient's operation and chemotherapy were all successful and he maintained a high quality of life after surgery that far exceeded his predicted survival. LESSONS: Early diagnosis, higher education level, attention to the disease and complications, reducing chemotherapy damage, adequate nutrient intake, relieving symptoms, controlling depression, and maintaining immunity and the ability to perform activities of daily living may all be the positive factors that can prolong the survival of patients with SMARCA4-UT.


Subject(s)
DNA Helicases , Lung Neoplasms , Quality of Life , Transcription Factors , Humans , Male , Middle Aged , DNA Helicases/genetics , DNA Helicases/deficiency , Transcription Factors/genetics , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Pneumonectomy
2.
Nat Commun ; 15(1): 7003, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143110

ABSTRACT

DNA polymerase theta (Polθ) is a DNA helicase-polymerase protein that facilitates DNA repair and is synthetic lethal with homology-directed repair (HDR) factors. Thus, Polθ is a promising precision oncology drug-target in HDR-deficient cancers. Here, we characterize the binding and mechanism of action of a Polθ helicase (Polθ-hel) small-molecule inhibitor (AB25583) using cryo-EM. AB25583 exhibits 6 nM IC50 against Polθ-hel, selectively kills BRCA1/2-deficient cells, and acts synergistically with olaparib in cancer cells harboring pathogenic BRCA1/2 mutations. Cryo-EM uncovers predominantly dimeric Polθ-hel:AB25583 complex structures at 3.0-3.2 Å. The structures reveal a binding-pocket deep inside the helicase central-channel, which underscores the high specificity and potency of AB25583. The cryo-EM structures in conjunction with biochemical data indicate that AB25583 inhibits the ATPase activity of Polθ-hel helicase via an allosteric mechanism. These detailed structural data and insights about AB25583 inhibition pave the way for accelerating drug development targeting Polθ-hel in HDR-deficient cancers.


Subject(s)
Cryoelectron Microscopy , DNA Helicases , DNA Polymerase theta , DNA-Directed DNA Polymerase , Humans , DNA Helicases/metabolism , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Helicases/antagonists & inhibitors , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/chemistry , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/chemistry , Piperazines/pharmacology , Piperazines/chemistry , Cell Line, Tumor , Phthalazines/pharmacology , Phthalazines/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Models, Molecular , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Protein Binding
3.
Sci Rep ; 14(1): 18923, 2024 08 15.
Article in English | MEDLINE | ID: mdl-39143142

ABSTRACT

Chromodomain helicase DNA-binding protein (CHD) gene family, an ATP (adenosine triphosphate) -dependent chromatin remodeler family, is involved in multiple developmental process and tumor development. However, there have been none pan-cancer analyses of this family. The expression levels, survival profiles, mutation profiles and immune infiltration of the CHD family genes from TCGA and TARGET database were analyzed using online tools or R packages. Interestingly, all types of CHD gene expressions were associated with the prognosis of Neuroblastoma, Acute lymphoblastic leukemia-Phase 3 and Acute Myeloid Leukemia (All P < 0.05). Knock down of CHD7 and CHD9 in K562 (human erythromyeloblastoid leukemia) and HEC-1-B (human endometrial adenocarcinoma) cells significantly inhibit cell proliferation and migration (P < 0.05). Proliferation, colony formation and migration assays were performed in CHD7 and CHD9 knockdown K562 and HBC-1-B cell lines. Mechanisms were also analyzed by PPI and GO ontology for our experiments. Histone modification, especially the methylation of H3K4, might be involved in CHD7 and CHD9 related oncogenesis. Through bioinformatic analysis, we showed CHD genes significantly affected the prognosis of different tumor types, including childhood tumor. Our findings provide new insights into the function and mechanism of CHD gene family, especially in CHD7 and CHD9.


Subject(s)
Computational Biology , DNA Helicases , DNA-Binding Proteins , Neoplasms , Humans , Computational Biology/methods , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Neoplasms/genetics , Neoplasms/pathology , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Prognosis , Cell Line, Tumor , Mutation
4.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(8): 962-965, 2024 Aug 10.
Article in Chinese | MEDLINE | ID: mdl-39097280

ABSTRACT

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.


Subject(s)
CHARGE Syndrome , DNA Helicases , DNA-Binding Proteins , Mutation, Missense , Humans , CHARGE Syndrome/genetics , DNA Helicases/genetics , Female , DNA-Binding Proteins/genetics , Exome Sequencing , Infant , Amino Acid Sequence
5.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125997

ABSTRACT

The transmembrane protein ß-amyloid precursor protein (APP) is central to the pathophysiology of Alzheimer's disease (AD). The ß-amyloid hypothesis posits that aberrant processing of APP forms neurotoxic ß-amyloid aggregates, which lead to the cognitive impairments observed in AD. Although numerous additional factors contribute to AD, there is a need to better understand the synaptic function of APP. We have found that Drosophila APP-like (APPL) has both shared and non-shared roles at the synapse with Kismet (Kis), a chromatin helicase binding domain (CHD) protein. Kis is the homolog of CHD7 and CHD8, both of which are implicated in neurodevelopmental disorders including CHARGE Syndrome and autism spectrum disorders, respectively. Loss of function mutations in kis and animals expressing human APP and BACE in their central nervous system show reductions in the glutamate receptor subunit, GluRIIC, the GTPase Rab11, and the bone morphogenetic protein (BMP), pMad, at the Drosophila larval neuromuscular junction (NMJ). Similarly, processes like endocytosis, larval locomotion, and neurotransmission are deficient in these animals. Our pharmacological and epistasis experiments indicate that there is a functional relationship between Kis and APPL, but Kis does not regulate appl expression at the larval NMJ. Instead, Kis likely influences the synaptic localization of APPL, possibly by promoting rab11 transcription. These data identify a potential mechanistic connection between chromatin remodeling proteins and aberrant synaptic function in AD.


Subject(s)
Amyloid beta-Protein Precursor , Drosophila Proteins , Neuromuscular Junction , rab GTP-Binding Proteins , Animals , Neuromuscular Junction/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Synaptic Transmission , Synapses/metabolism , Receptors, Glutamate/metabolism , Receptors, Glutamate/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Humans , DNA Helicases/metabolism , DNA Helicases/genetics , Membrane Proteins , Nerve Tissue Proteins , Homeodomain Proteins , Receptors, Ionotropic Glutamate
6.
Cell Mol Life Sci ; 81(1): 339, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39120648

ABSTRACT

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.


Subject(s)
Autophagy , DNA Damage , DNA Helicases , Inflammation , Multifunctional Enzymes , Nucleotidyltransferases , R-Loop Structures , RNA Helicases , Humans , Autophagy/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/deficiency , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/deficiency , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/deficiency , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Immunity, Innate , Inflammation/pathology , Inflammation/metabolism , Inflammation/genetics , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Phosphoproteins , RNA Helicases/metabolism , RNA Helicases/genetics
7.
Nat Commun ; 15(1): 6419, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39079955

ABSTRACT

Multiple Sclerosis (MS) is a heterogeneous inflammatory and neurodegenerative disease with an unpredictable course towards progressive disability. Treating progressive MS is challenging due to limited insights into the underlying mechanisms. We examined the molecular changes associated with primary progressive MS (PPMS) using a cross-tissue (blood and post-mortem brain) and multilayered data (genetic, epigenetic, transcriptomic) from independent cohorts. In PPMS, we found hypermethylation of the 1q21.1 locus, controlled by PPMS-specific genetic variations and influencing the expression of proximal genes (CHD1L, PRKAB2) in the brain. Evidence from reporter assay and CRISPR/dCas9 experiments supports a causal link between methylation and expression and correlation network analysis further implicates these genes in PPMS brain processes. Knock-down of CHD1L in human iPSC-derived neurons and knock-out of chd1l in zebrafish led to developmental and functional deficits of neurons. Thus, several lines of evidence suggest a distinct genetic-epigenetic-transcriptional interplay in the 1q21.1 locus potentially contributing to PPMS pathogenesis.


Subject(s)
Brain , Chromosomes, Human, Pair 1 , DNA Methylation , DNA-Binding Proteins , Epigenesis, Genetic , Zebrafish , Humans , Zebrafish/genetics , Animals , DNA Methylation/genetics , Chromosomes, Human, Pair 1/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Brain/metabolism , Brain/pathology , DNA Helicases/genetics , DNA Helicases/metabolism , Neurons/metabolism , Multiple Sclerosis, Chronic Progressive/genetics , Induced Pluripotent Stem Cells/metabolism , Male , Female , Middle Aged , Genetic Predisposition to Disease , Adult
8.
J Hematol Oncol ; 17(1): 58, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080761

ABSTRACT

INTRODUCTION: Small Cell Lung Cancer (SCLC) can be classified into transcriptional subtypes with distinct degrees of neuroendocrine (NE) differentiation. Recent evidence supports plasticity among subtypes with a bias toward adoption of low-NE states during disease progression or upon acquired chemotherapy resistance. Here, we identify a role for SMARCA4, the catalytic subunit of the SWI/SNF complex, as a regulator of subtype shift in SCLC. METHODS: ATACseq and RNAseq experiments were performed in SCLC cells after pharmacological inhibition of SMARCA4. DNA binding of SMARCA4 was characterized by ChIPseq in high-NE SCLC patient derived xenografts (PDXs). Enrichment analyses were applied to transcriptomic data. Combination of FHD-286 and afatinib was tested in vitro and in a set of chemo-resistant SCLC PDXs in vivo. RESULTS: SMARCA4 expression positively correlates with that of NE genes in both SCLC cell lines and patient tumors. Pharmacological inhibition of SMARCA4 with FHD-286 induces the loss of NE features and downregulates neuroendocrine and neuronal signaling pathways while activating non-NE factors. SMARCA4 binds to gene loci encoding NE-lineage transcription factors ASCL1 and NEUROD1 and alters chromatin accessibility, enhancing NE programs. Enrichment analysis applied to high-confidence SMARCA4 targets confirmed neuron related pathways as the top GO Biological processes regulated by SMARCA4 in SCLC. In parallel, SMARCA4 also controls REST, a known suppressor of the NE phenotype, by regulating SRRM4-dependent REST transcript splicing. Furthermore, SMARCA4 inhibition drives ERBB pathway activation in SCLC, rendering SCLC tumors sensitive to afatinib. CONCLUSIONS: This study nominates SMARCA4 as a key regulator of the NE state plasticity and defines a novel therapeutic strategy for SCLC.


Subject(s)
DNA Helicases , Lung Neoplasms , Nuclear Proteins , Small Cell Lung Carcinoma , Transcription Factors , Humans , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , DNA Helicases/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Cell Line, Tumor , Animals , Mice , Gene Expression Regulation, Neoplastic , Repressor Proteins
9.
Nat Commun ; 15(1): 6104, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030241

ABSTRACT

G-quadruplexes (G4s) formed by guanine-rich nucleic acids induce genome instability through impeding DNA replication fork progression. G4s are stable DNA structures, the unfolding of which require the functions of DNA helicases. Pif1 helicase binds preferentially to G4 DNA and plays multiple roles in maintaining genome stability, but the mechanism by which Pif1 unfolds G4s is poorly understood. Here we report the co-crystal structure of Saccharomyces cerevisiae Pif1 (ScPif1) bound to a G4 DNA with a 5' single-stranded DNA (ssDNA) segment. Unlike the Thermus oshimai Pif1-G4 structure, in which the 1B and 2B domains confer G4 recognition, ScPif1 recognizes G4 mainly through the wedge region in the 1A domain that contacts the 5' most G-tetrad directly. A conserved Arg residue in the wedge is required for Okazaki fragment processing but not for mitochondrial function or for suppression of gross chromosomal rearrangements. Multiple substitutions at this position have similar effects on resolution of DNA duplexes and G4s, suggesting that ScPif1 may use the same wedge to unwind G4 and dsDNA. Our results reveal the mechanism governing dsDNA unwinding and G4 unfolding by ScPif1 helicase that can potentially be generalized to other eukaryotic Pif1 helicases and beyond.


Subject(s)
DNA Helicases , G-Quadruplexes , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , DNA Helicases/metabolism , DNA Helicases/chemistry , DNA Helicases/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , DNA/metabolism , DNA/chemistry , DNA/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Binding , DNA Replication , Genomic Instability
10.
Nat Commun ; 15(1): 6200, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043686

ABSTRACT

Cell fate is likely regulated by a common machinery, while components of this machine remain to be identified. Here we report the design and testing of engineered cell fate controller NanogBiD, fusing BiD or BRG1 interacting domain of SS18 with Nanog. NanogBiD promotes mouse somatic cell reprogramming efficiently in contrast to the ineffective native protein under multiple testing conditions. Mechanistic studies further reveal that it facilitates cell fate transition by recruiting the intended Brg/Brahma-associated factor (BAF) complex to modulate chromatin accessibility and reorganize cell state specific enhancers known to be occupied by canonical Nanog, resulting in precocious activation of multiple genes including Sall4, miR-302, Dppa5a and Sox15 towards pluripotency. Although we have yet to test our approach in other species, our findings suggest that engineered chromatin regulators may provide much needed tools to engineer cell fate in the cells as drugs era.


Subject(s)
Nanog Homeobox Protein , Transcription Factors , Animals , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Cellular Reprogramming/genetics , Chromatin/metabolism , Chromatin/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Cell Differentiation , Cell Engineering/methods , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
11.
Cancer Biol Ther ; 25(1): 2375440, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38978225

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most lethal malignant tumors worldwide. Brahma-related gene 1 (BRG1), as a catalytic ATPase, is a major regulator of gene expression and is known to mutate and overexpress in HCC. The purpose of this study was to investigate the mechanism of action of BRG1 in HCC cells. In our study, BRG1 was silenced or overexpressed in human HCC cell lines. Transwell and wound healing assays were used to analyze cell invasiveness and migration. Mitochondrial membrane potential (MMP) and mitochondrial permeability transition pore (mPTP) detection were used to evaluate mitochondrial function in HCC cells. Colony formation and cell apoptosis assays were used to evaluate the effect of BRG1/TOMM40/ATP5A1 on HCC cell proliferation and apoptosis/death. Immunocytochemistry (ICC), immunofluorescence (IF) staining and western blot analysis were used to determine the effect of BRG1 on TOMM40, ATP5A1 pathway in HCC cells. As a result, knockdown of BRG1 significantly inhibited cell proliferation and invasion, promoted apoptosis in HCC cells, whereas BRG1 overexpression reversed the above effects. Overexpression of BRG1 can up-regulate MMP level, inhibit mPTP opening and activate TOMM40, ATP5A1 expression. Our results suggest that BRG1, as an oncogene, promotes HCC progression by regulating TOMM40 affecting mitochondrial function and ATP5A1 synthesis. Targeting BRG1 may represent a new and effective way to prevent HCC development.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular , Cell Proliferation , DNA Helicases , Liver Neoplasms , Mitochondria , Mitochondrial Precursor Protein Import Complex Proteins , Nuclear Proteins , Transcription Factors , Humans , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , DNA Helicases/metabolism , DNA Helicases/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Neoplasm Metastasis , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
12.
Microb Biotechnol ; 17(7): e14524, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38980956

ABSTRACT

The coral reef microbiome plays a vital role in the health and resilience of reefs. Previous studies have examined phage therapy for coral pathogens and for modifying the coral reef microbiome, but defence systems against coral-associated bacteria have received limited attention. Phage defence systems play a crucial role in helping bacteria fight phage infections. In this study, we characterized a new defence system, Hma (HmaA-HmaB-HmaC), in the coral-associated Halomonas meridiana derived from the scleractinian coral Galaxea fascicularis. The Swi2/Snf2 helicase HmaA with a C-terminal nuclease domain exhibits antiviral activity against Escherichia phage T4. Mutation analysis revealed the nickase activity of the nuclease domain (belonging to PDD/EXK superfamily) of HmaA is essential in phage defence. Additionally, HmaA homologues are present in ~1000 bacterial and archaeal genomes. The high frequency of HmaA helicase in Halomonas strains indicates the widespread presence of these phage defence systems, while the insertion of defence genes in the hma region confirms the existence of a defence gene insertion hotspot. These findings offer insights into the diversity of phage defence systems in coral-associated bacteria and these diverse defence systems can be further applied into designing probiotics with high-phage resistance.


Subject(s)
Anthozoa , DNA Helicases , Halomonas , Halomonas/genetics , Halomonas/enzymology , Animals , Anthozoa/microbiology , Anthozoa/virology , DNA Helicases/genetics , DNA Helicases/metabolism , Bacteriophages/genetics , Bacteriophages/enzymology , Bacteriophages/physiology , Deoxyribonucleases/genetics , Deoxyribonucleases/metabolism
13.
Cell Rep ; 43(7): 114464, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38985669

ABSTRACT

Poly(ADP-ribose) polymerase inhibitors (PARPis) exhibit remarkable anticancer activity in tumors with homologous recombination (HR) gene mutations. However, the role of other DNA repair proteins in PARPi-induced lethality remains elusive. Here, we reveal that FANCM promotes PARPi resistance independent of the core Fanconi anemia (FA) complex. FANCM-depleted cells retain HR proficiency, acting independently of BRCA1 in response to PARPis. FANCM depletion leads to increased DNA damage in the second S phase after PARPi exposure, driven by elevated single-strand DNA (ssDNA) gap formation behind replication forks in the first S phase. These gaps arise from both 53BP1- and primase and DNA directed polymerase (PRIMPOL)-dependent mechanisms. Notably, FANCM-depleted cells also exhibit reduced resection of collapsed forks, while 53BP1 deletion restores resection and mitigates PARPi sensitivity. Our results suggest that FANCM counteracts 53BP1 to repair PARPi-induced DNA damage. Furthermore, FANCM depletion leads to increased chromatin bridges and micronuclei formation after PARPi treatment, elucidating the mechanism underlying extensive cell death in FANCM-depleted cells.


Subject(s)
DNA, Single-Stranded , Poly(ADP-ribose) Polymerase Inhibitors , Tumor Suppressor p53-Binding Protein 1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Humans , DNA, Single-Stranded/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Damage , DNA Repair/drug effects , Homologous Recombination/drug effects , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor
14.
J Cell Biol ; 223(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39007803

ABSTRACT

Stress triggers the formation of two distinct cytoplasmic biomolecular condensates: stress granules (SGs) and processing bodies (PBs), both of which may contribute to stress-responsive translation regulation. Though PBs can be present constitutively, stress can increase their number and size and lead to their interaction with stress-induced SGs. The mechanism of such interaction, however, is largely unknown. Formation of canonical SGs requires the RNA binding protein Ubiquitin-Associated Protein 2-Like (UBAP2L), which is a central SG node protein in the RNA-protein interaction network of SGs and PBs. UBAP2L binds to the essential SG and PB proteins G3BP and DDX6, respectively. Research on UBAP2L has mostly focused on its role in SGs, but not its connection to PBs. We find that UBAP2L is not solely an SG protein but also localizes to PBs in certain conditions, contributes to PB biogenesis and SG-PB interactions, and can nucleate hybrid granules containing SG and PB components in cells. These findings inform a new model for SG and PB formation in the context of UBAP2L's role.


Subject(s)
RNA Helicases , RNA Recognition Motif Proteins , Stress Granules , Humans , Stress Granules/metabolism , Stress Granules/genetics , RNA Helicases/metabolism , RNA Helicases/genetics , RNA Recognition Motif Proteins/metabolism , RNA Recognition Motif Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Processing Bodies/metabolism , Processing Bodies/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Cytoplasmic Granules/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , HeLa Cells , DNA Helicases/metabolism , DNA Helicases/genetics , HEK293 Cells , Protein Binding , Carrier Proteins/metabolism , Carrier Proteins/genetics , Proto-Oncogene Proteins
15.
ACS Chem Biol ; 19(7): 1433-1439, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38959478

ABSTRACT

Most of the human cancers are dependent on telomerase to extend the telomeres. But ∼10% of all cancers use a telomerase-independent, homologous recombination mediated pathway called alternative lengthening of telomeres (ALT). Due to the poor prognosis, ALT status is not being considered yet in the diagnosis of cancer. No such specific treatment is available to date for ALT positive cancers. ALT positive cancers are dependent on replication stress to deploy DNA repair pathways to the telomeres to execute homologous recombination mediated telomere extension. SMARCAL1 (SWI/SNF related, matrix-associated, actin-dependent regulator of chromatin, subfamily A-like 1) is associated with the ALT telomeres to resolve replication stress thus providing telomere stability. Thus, the dependency on replication stress regulatory factors like SMARCAL1 made it a suitable therapeutic target for the treatment of ALT positive cancers. In this study, we found a significant downregulation of SMARCAL1 expression by stabilizing the G-quadruplex (G4) motif found in the promoter of SMARCAL1 by potent G4 stabilizers, like TMPyP4 and BRACO-19. SMARCAL1 downregulation led toward the increased localization of PML (promyelocytic leukemia) bodies in ALT telomeres and triggered the formation of APBs (ALT-associated promyelocytic leukemia bodies) in ALT positive cell lines, increasing telomere replication stress and DNA damage at a genomic level. Induction of replication stress and hyper-recombinogenic phenotype in ALT positive cells mediated by G4 stabilizing molecules already highlighted their possible application as a new therapeutic window to target ALT positive tumors. In accordance with this, our study will also provide a valuable insight toward the development of G4-based ALT therapeutics targeting SMARCAL1.


Subject(s)
DNA Helicases , G-Quadruplexes , Neoplasms , Promoter Regions, Genetic , Telomere , Humans , Telomere/genetics , Telomere/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , Neoplasms/genetics , Cell Line, Tumor , DNA Replication , Telomere Homeostasis
16.
Pathologica ; 116(3): 163-169, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38979590

ABSTRACT

The 5th WHO classification of thoracic tumours includes thoracic SMARCA4-deficient undifferentiated tumour (SMARCA4-UT) among the "other epithelial tumours of the lung" chapter. Herein, we present a case of undifferentiated thoracic neoplasm with retention of SMARCA4 expression, lack of NUT fusion protein and loss of SMARCB1/INI1 expression. After presenting the clinical and pathological features of the tumour, we carried out a review of the literature on the same topic. Albeit very rare, we believe this entity should be included in the heterogeneous group of undifferentiated neoplasms of the thorax.


Subject(s)
DNA Helicases , SMARCB1 Protein , Thoracic Neoplasms , Transcription Factors , Humans , SMARCB1 Protein/deficiency , SMARCB1 Protein/genetics , Transcription Factors/genetics , Transcription Factors/deficiency , Thoracic Neoplasms/pathology , Thoracic Neoplasms/genetics , DNA Helicases/deficiency , DNA Helicases/genetics , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Male , Female , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Middle Aged , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/diagnosis
17.
Front Biosci (Landmark Ed) ; 29(7): 262, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39082357

ABSTRACT

BACKGROUND: The switching/sucrose non-fermentable (SWI/SNF) Related, Matrix Associated, Actin Dependent Regulator Of Chromatin, Subfamily A (SMARCA) member 2 and member 4 (SMARCA2/4) are paralogs and act as the key enzymatic subunits in the SWI/SNF complex for chromatin remodeling. However, the role of SMARCA2/4 in DNA damage response remains unclear. METHODS: Laser microirradiation assays were performed to examine the key domains of SMARCA2/4 for the relocation of the SWI/SNF complex to DNA lesions. To examine the key factors that mediate the recruitment of SMARCA2/4, the relocation of SMARCA2/4 to DNA lesions was examined in HeLa cells treated with inhibitors of Ataxia-telangiectasia-mutated (ATM), Ataxia telangiectasia and Rad3-related protein (ATR), CREB-binding protein (CBP) and its homologue p300 (p300/CBP), or Poly (ADP-ribose) polymerase (PARP) 1/2 as well as in H2AX-deficient HeLa cells. Moreover, by concomitantly suppressing SMARCA2/4 with the small molecule inhibitor FHD286 or Compound 14, the function of SMARCA2/4 in Radiation sensitive 51 (RAD51) foci formation and homologous recombination repair was examined. Finally, using a colony formation assay, the synergistic effect of PARP inhibitors and SMARCA2/4 inhibitors on the suppression of tumor cell growth was examined. RESULTS: We show that SMARCA2/4 relocate to DNA lesions in response to DNA damage, which requires their ATPase activities. Moreover, these ATPase activities are also required for the relocation of other subunits in the SWI/SNF complex to DNA lesions. Interestingly, the relocation of SMARCA2/4 is independent of γH2AX, ATM, ATR, p300/CBP, or PARP1/2, indicating that it may directly recognize DNA lesions as a DNA damage sensor. Lacking SMARCA2/4 prolongs the retention of γH2AX, Ring Finger Protein 8 (RNF8) and Breast cancer susceptibility gene 1 (BRCA1) at DNA lesions and impairs RAD51-dependent homologous recombination repair. Furthermore, the treatment of an SMARCA2/4 inhibitor sensitizes tumor cells to PARP inhibitor treatment. CONCLUSIONS: This study reveals SMARCA2/4 as a DNA damage repair factor for double-strand break repair.


Subject(s)
DNA Damage , DNA Helicases , DNA Repair , Nuclear Proteins , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , HeLa Cells , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Histones/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , BRCA1 Protein/metabolism , BRCA1 Protein/genetics
18.
Matrix Biol ; 132: 72-86, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39009171

ABSTRACT

Post-mitotic, non-proliferative dermal fibroblasts have crucial functions in maintenance and restoration of tissue homeostasis. They are involved in essential processes such as wound healing, pigmentation and hair growth, but also tumor development and aging-associated diseases. These processes are energetically highly demanding and error prone when mitochondrial damage occurs. However, mitochondrial function in fibroblasts and the influence of mitochondrial dysfunction on fibroblast-specific demands are still unclear. To address these questions, we created a mouse model in which accelerated cell-specific mitochondrial DNA (mtDNA) damage accumulates. We crossed mice carrying a dominant-negative mutant of the mitochondrial replicative helicase Twinkle (RosaSTOP system) with mice that express fibroblast-specific Cre Recombinase (Collagen1A2 CreERT) which can be activated by Tamoxifen (TwinkleFIBRO). Thus, we are able to induce mtDNA deletions and duplications in specific cells, a process which resembles the physiological aging process in humans, where this damage accumulates in all tissues. Upon proliferation in vitro, Tamoxifen induced Twinkle fibroblasts deplete most of their mitochondrial DNA which, although not disturbing the stoichiometry of the respiratory chain complexes, leads to reduced ROS production and mitochondrial membrane potential as well as an anti-inflammatory and anti-fibrotic profile of the cells. In Sodium Azide treated wildtype fibroblasts, without a functioning respiratory chain, we observe the opposite, a rather pro-inflammatory and pro-fibrotic signature. Upon accumulation of mitochondrial DNA mutations in vivo the TwinkleFIBRO mice are protected from fibrosis development induced by intradermal Bleomycin injections. This is due to dampened differentiation of the dermal fibroblasts into α-smooth-muscle-actin positive myofibroblasts in TwinkleFIBRO mice. We thus provide evidence for striking differences of the impact that mtDNA mutations have in contrast to blunted mitochondrial function in dermal fibroblasts and skin homeostasis. These data contribute to improved understanding of mitochondrial function and dysfunction in skin and provide mechanistic insight into potential targets to treat skin fibrosis in the future.


Subject(s)
Bleomycin , Cell Differentiation , DNA, Mitochondrial , Fibrosis , Mutation , Myofibroblasts , Animals , Bleomycin/adverse effects , Bleomycin/toxicity , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Mice , Myofibroblasts/metabolism , Myofibroblasts/pathology , Myofibroblasts/drug effects , Cell Differentiation/drug effects , DNA Helicases/genetics , DNA Helicases/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Tamoxifen/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Disease Models, Animal , Reactive Oxygen Species/metabolism , Humans , Skin/pathology , Skin/metabolism , Skin/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Collagen Type I
19.
Nat Commun ; 15(1): 6031, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39019869

ABSTRACT

Mutations in the Cockayne Syndrome group B (CSB) gene cause cancer in mice, but premature aging and severe neurodevelopmental defects in humans. CSB, a member of the SWI/SNF family of chromatin remodelers, plays diverse roles in regulating gene expression and transcription-coupled nucleotide excision repair (TC-NER); however, these functions do not explain the distinct phenotypic differences observed between CSB-deficient mice and humans. During investigating Cockayne Syndrome-associated genome instability, we uncover an intrinsic mechanism that involves elongating RNA polymerase II (RNAPII) undergoing transient pauses at internal T-runs where CSB is required to propel RNAPII forward. Consequently, CSB deficiency retards RNAPII elongation in these regions, and when coupled with G-rich sequences upstream, exacerbates genome instability by promoting R-loop formation. These R-loop prone motifs are notably abundant in relatively long genes related to neuronal functions in the human genome, but less prevalent in the mouse genome. These findings provide mechanistic insights into differential impacts of CSB deficiency on mice versus humans and suggest that the manifestation of the Cockayne Syndrome phenotype in humans results from the progressive evolution of mammalian genomes.


Subject(s)
Cockayne Syndrome , DNA Helicases , DNA Repair Enzymes , Genomic Instability , Poly-ADP-Ribose Binding Proteins , R-Loop Structures , RNA Polymerase II , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , Cockayne Syndrome/metabolism , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Animals , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , R-Loop Structures/genetics , DNA Repair , Transcription Elongation, Genetic , Mice, Knockout
20.
Nucleic Acids Res ; 52(14): 8609-8627, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-38967005

ABSTRACT

High spontaneous mutation rate is crucial for obtaining ideal phenotype and exploring the relationship between genes and phenotype. How to break the genetic stability of organisms and increase the mutation frequency has become a research hotspot. Here, we present a practical and controllable evolutionary tool (oMut-Cgts) based on dual genetic level modification engineering for Corynebacterium glutamicum. Firstly, the modification engineering of transcription and replication levels based on RNA polymerase α subunit and DNA helicase Cgl0854 as the 'dock' of cytidine deaminase (pmCDA1) significantly increased the mutation rate, proving that the localization of pmCDA1 around transient ssDNA is necessary for genome mutation. Then, the combined modification and optimization of engineering at dual genetic level achieved 1.02 × 104-fold increased mutation rate. The genome sequencing revealed that the oMut-Cgts perform uniform and efficient C:G→T:A transitions on a genome-wide scale. Furthermore, oMut-Cgts-mediated rapid evolution of C. glutamicum with stress (acid, oxidative and ethanol) tolerance proved that the tool has powerful functions in multi-dimensional biological engineering (rapid phenotype evolution, gene function mining and protein evolution). The strategies for rapid genome evolution provided in this study are expected to be applicable to a variety of applications in all prokaryotic cells.


Subject(s)
Corynebacterium glutamicum , Genome, Bacterial , Corynebacterium glutamicum/genetics , Genetic Engineering/methods , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Mutation Rate , Evolution, Molecular , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Replication/genetics , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL