Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 217
Filter
1.
Redox Rep ; 29(1): 2345455, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38723197

ABSTRACT

OBJECTIVES: Cancer cells undergo metabolic reprogramming to adapt to high oxidative stress, but little is known about how metabolic remodeling enables gastric cancer cells to survive stress associated with aberrant reactive oxygen species (ROS) production. Here, we aimed to identify the key metabolic enzymes that protect gastric cancer (GC) cells from oxidative stress. METHODS: ROS level was detected by DCFH-DA probes. Multiple cell biological studies were performed to identify the underlying mechanisms. Furthermore, cell-based xenograft and patient-derived xenograft (PDX) model were performed to evaluate the role of MTHFD2 in vivo. RESULTS: We found that overexpression of MTHFD2, but not MTHFD1, is associated with reduced overall and disease-free survival in gastric cancer. In addition, MTHFD2 knockdown reduces the cellular NADPH/NADP+ ratio, colony formation and mitochondrial function, increases cellular ROS and cleaved PARP levels and induces in cell death under hypoxia, a hallmark of solid cancers and a common inducer of oxidative stress. Moreover, genetic or pharmacological inhibition of MTHFD2 reduces tumor burden in both tumor cell lines and patient-derived xenograft-based models. DISCUSSION: our study highlights the crucial role of MTHFD2 in redox regulation and tumor progression, demonstrating the therapeutic potential of targeting MTHFD2.


Subject(s)
Methylenetetrahydrofolate Dehydrogenase (NADP) , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species , Stomach Neoplasms , Humans , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Animals , Mice , Reactive Oxygen Species/metabolism , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , Cell Line, Tumor , Homeostasis , Aminohydrolases/metabolism , Aminohydrolases/genetics , Disease Progression , Xenograft Model Antitumor Assays
2.
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
3.
J Bioenerg Biomembr ; 56(3): 333-345, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38488992

ABSTRACT

Ovarian cancer (OC) is a deadliest gynecological cancer with the highest mortality rate. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), a crucial tumor-promoting factor, is over-expressed in several malignancies including OC. The present study aimed to explore the role and mechanisms of MTHFD2 in OC malignant progression. Thus, cell proliferation, cycling, apoptosis, migration, and invasion were evaluated by CCK-8 assay, EdU assay, flow cytometry, wound healing, transwell assay and western blotting. Additionally, glycolysis was assessed by measuring the level of glucose and lactate production, as well as the expressions of GLUT1, HK2 and PKM2. Then the expression of ferroptosis-related proteins and ERK signaling was detected using western blotting. Ferroptosis was detected through the measurement of iron level, GSH, MDA and ROS activities. The results revealed that MTHFD2 was highly expressed in OC cells. Besides, interference with MTHFD2 induced ferroptosis, promoted ROS accumulation, destroyed mitochondrial function, reduced ATP content and inhibited glycolysis in OC cells. Subsequently, we further found that interference with MTHFD2 affected mitochondrial function and glycolysis in OC cells through ERK signaling. Moreover, interference with MTHFD2 affected ferroptosis to inhibit the malignant progression of OC cells. Collectively, our present study disclosed that interference with MTHFD2 induced ferroptosis in OC to inhibit tumor malignant progression through regulating ERK signaling.


Subject(s)
Ferroptosis , MAP Kinase Signaling System , Methylenetetrahydrofolate Dehydrogenase (NADP) , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Ferroptosis/physiology , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multifunctional Enzymes/metabolism , Cell Line, Tumor , Aminohydrolases/metabolism , Aminohydrolases/genetics , Disease Progression , Mice
4.
J Mol Biol ; 436(9): 168542, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38492718

ABSTRACT

PrimPol is a human DNA primase-polymerase which restarts DNA synthesis beyond DNA lesions and non-B DNA structures blocking replication. Disfunction of PrimPol in cells leads to slowing of DNA replication rates in mitochondria and nucleus, accumulation of chromosome aberrations, cell cycle delay, and elevated sensitivity to DNA-damaging agents. A defective PrimPol has been suggested to be associated with the development of ophthalmic diseases, elevated mitochondrial toxicity of antiviral drugs and increased cell resistance to chemotherapy. Here, we describe a rare missense PrimPol variant V102A with altered biochemical properties identified in patients suffering from ovarian and cervical cancer. The Val102 to Ala substitution dramatically reduced both the primase and DNA polymerase activities of PrimPol as well as specifically decreased its ability to incorporate ribonucleotides. Structural analysis indicates that the V102A substitution can destabilize the hydrophobic pocket adjacent to the active site, affecting dNTP binding and catalysis.


Subject(s)
DNA Primase , DNA-Directed DNA Polymerase , Multifunctional Enzymes , Mutation, Missense , Ovarian Neoplasms , Uterine Cervical Neoplasms , Female , Humans , Amino Acid Substitution , Catalytic Domain , Crystallography, X-Ray , DNA Primase/metabolism , DNA Primase/chemistry , DNA Primase/genetics , DNA Replication , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/chemistry , Models, Molecular , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/chemistry , Protein Conformation , Uterine Cervical Neoplasms/genetics , Ovarian Neoplasms/genetics
5.
Nucleic Acids Res ; 52(7): 3740-3760, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38321962

ABSTRACT

It is well-established that, through canonical functions in transcription and DNA repair, the tumor suppressor p53 plays a central role in safeguarding cells from the consequences of DNA damage. Recent data retrieved in tumor and stem cells demonstrated that p53 also carries out non-canonical functions when interacting with the translesion synthesis (TLS) polymerase iota (POLι) at DNA replication forks. This protein complex triggers a DNA damage tolerance (DDT) mechanism controlling the DNA replication rate. Given that the levels of p53 trigger non-binary rheostat-like functions in response to stress or during differentiation, we explore the relevance of the p53 levels for its DDT functions at the fork. We show that subtle changes in p53 levels modulate the contribution of some DDT factors including POLι, POLη, POLζ, REV1, PCNA, PRIMPOL, HLTF and ZRANB3 to the DNA replication rate. Our results suggest that the levels of p53 are central to coordinate the balance between DDT pathways including (i) fork-deceleration by the ZRANB3-mediated fork reversal factor, (ii) POLι-p53-mediated fork-slowing, (iii) POLι- and POLη-mediated TLS and (iv) PRIMPOL-mediated fork-acceleration. Collectively, our study reveals the relevance of p53 protein levels for the DDT pathway choice in replicating cells.


Subject(s)
DNA Damage , DNA Polymerase iota , DNA Replication , DNA-Directed DNA Polymerase , Tumor Suppressor Protein p53 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , DNA-Directed DNA Polymerase/metabolism , Humans , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/genetics , DNA Repair , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , DNA Primase/metabolism , DNA Primase/genetics , DNA Damage Tolerance
6.
Nucleic Acids Res ; 52(7): 3778-3793, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38348929

ABSTRACT

DNA replication stress, caused by various endogenous and exogenous agents, halt or stall DNA replication progression. Cells have developed diverse mechanisms to tolerate and overcome replication stress, enabling them to continue replication. One effective strategy to overcome stalled replication involves skipping the DNA lesion using a specialized polymerase known as PrimPol, which reinitiates DNA synthesis downstream of the damage. However, the mechanism regulating PrimPol repriming is largely unclear. In this study, we observe that knockdown of STN1 or CTC1, components of the CTC1/STN1/TEN1 complex, leads to enhanced replication progression following UV exposure. We find that such increased replication is dependent on PrimPol, and PrimPol recruitment to stalled forks increases upon CST depletion. Moreover, we find that p21 is upregulated in STN1-depleted cells in a p53-independent manner, and p21 depletion restores normal replication rates caused by STN1 deficiency. We identify that p21 interacts with PrimPol, and STN1 depletion stimulates p21-PrimPol interaction and facilitates PrimPol recruitment to stalled forks. Our findings reveal a previously undescribed interplay between CST, PrimPol and p21 in promoting repriming in response to stalled replication, and shed light on the regulation of PrimPol repriming at stalled forks.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21 , DNA Primase , DNA Replication , DNA-Directed DNA Polymerase , Multifunctional Enzymes , Telomere-Binding Proteins , Ultraviolet Rays , Humans , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Primase/metabolism , DNA Primase/genetics , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Telomere-Binding Proteins/metabolism , Telomere-Binding Proteins/genetics , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , DNA Damage
7.
Biochimie ; 217: 10-19, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37558082

ABSTRACT

The RNA/DNA helicase senataxin (SETX) has been involved in multiple crucial processes related to genome expression and integrity such us transcription termination, the regulation of transcription-replication conflicts and the resolution of R-loops. SETX has been the focus of numerous studies since the discovery that mutations in its coding gene are the root cause of two different neurodegenerative diseases: Ataxia with Oculomotor Apraxia type 2 (AOA2) and a juvenile form of Amyotrophic Lateral Sclerosis (ALS4). A plethora of cellular phenotypes have been described as the result of SETX deficiency, yet the precise molecular function of SETX as well as the molecular pathways leading from SETX mutations to AOA2 and ALS4 pathologies have remained unclear. However, recent data have shed light onto the biochemical activities and biological roles of SETX, thus providing new clues to understand the molecular consequences of SETX mutation. In this review we summarize near two decades of scientific effort to elucidate SETX function, we discuss strengths and limitations of the approaches and models used thus far to investigate SETX-associated diseases and suggest new possible research avenues for the study of AOA2 and ALS4 pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Humans , RNA Helicases/genetics , RNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Neurodegenerative Diseases/genetics , Transcription, Genetic , Mutation , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , RNA
8.
J Mol Biol ; 436(1): 168275, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37714300

ABSTRACT

Translesion DNA synthesis (TLS) is a DNA damage tolerance pathway utilized by cells to overcome lesions encountered throughout DNA replication. During replication stress, cancer cells show increased dependency on TLS proteins for cellular survival and chemoresistance. TLS proteins have been described to be involved in various DNA repair pathways. One of the major emerging roles of TLS is single-stranded DNA (ssDNA) gap-filling, primarily after the repriming activity of PrimPol upon encountering a lesion. Conversely, suppression of ssDNA gap accumulation by TLS is considered to represent a mechanism for cancer cells to evade the toxicity of chemotherapeutic agents, specifically in BRCA-deficient cells. Thus, TLS inhibition is emerging as a potential treatment regimen for DNA repair-deficient tumors.


Subject(s)
DNA Primase , DNA Repair , DNA, Single-Stranded , DNA-Directed DNA Polymerase , Multifunctional Enzymes , Translesion DNA Synthesis , DNA Damage , DNA, Single-Stranded/genetics , DNA-Directed DNA Polymerase/metabolism , Humans , Animals , DNA Primase/metabolism , Multifunctional Enzymes/metabolism
9.
Nucleic Acids Res ; 52(1): 243-258, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-37971291

ABSTRACT

The primase/polymerase PRIMPOL restarts DNA synthesis when replication is arrested by template impediments. However, we do not have a comprehensive view of how PRIMPOL-dependent repriming integrates with the main pathways of damage tolerance, REV1-dependent 'on-the-fly' lesion bypass at the fork and PCNA ubiquitination-dependent post-replicative gap filling. Guided by genome-wide CRISPR/Cas9 screens to survey the genetic interactions of PRIMPOL in a non-transformed and p53-proficient human cell line, we find that PRIMPOL is needed for cell survival following loss of the Y-family polymerases REV1 and POLη in a lesion-dependent manner, while it plays a broader role in promoting survival of cells lacking PCNA K164-dependent post-replicative gap filling. Thus, while REV1- and PCNA K164R-bypass provide two layers of protection to ensure effective damage tolerance, PRIMPOL is required to maximise the effectiveness of the interaction between them. We propose this is through the restriction of post-replicative gap length provided by PRIMPOL-dependent repriming.


Subject(s)
DNA Damage , DNA Primase , DNA-Directed DNA Polymerase , Humans , DNA Primase/genetics , DNA Primase/metabolism , DNA Replication , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , DNA-Directed DNA Polymerase/metabolism
10.
Biochemistry (Mosc) ; 88(11): 1933-1943, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38105210

ABSTRACT

Human DNA primase/polymerase PrimPol synthesizes DNA primers de novo after replication fork stalling at the sites of DNA damage, thus contributing to the DNA damage tolerance. The role of PrimPol in response to the different types of DNA damage is poorly understood. We knocked out the PRIMPOL gene in the lung carcinoma A549 cell line and characterized the response of the obtained cells to the DNA damage caused by hydrogen peroxide, methyl methanesulfonate (MMS), cisplatin, bleomycin, and ionizing radiation. The PRIMPOL knockout reduced the number of proliferating cells and cells in the G2 phase after treatment with MMS and caused a more pronounced delay of the S phase in the cisplatin-treated cells. Ionizing radiation at a dose of 10 Gy significantly increased the content of apoptotic cells among the PRIMPOL-deficient cells, while the proportion of cells undergoing necroptosis increased in both parental and knockout cells at any radiation dose. The viability of PRIMPOL-deficient cells upon the hydrogen peroxide-induced oxidative stress increased compared to the control cells, as determined by the methyl tetrazolium (MTT) assay. The obtained data indicate the involvement of PRIMPOL in the modulation of adaptive cell response to various types of genotoxic stress.


Subject(s)
Adenocarcinoma of Lung , DNA-Directed DNA Polymerase , Humans , DNA-Directed DNA Polymerase/metabolism , A549 Cells , Cisplatin/pharmacology , Hydrogen Peroxide/pharmacology , DNA Replication , DNA Damage , Adenocarcinoma of Lung/genetics , DNA Primase/genetics , DNA Primase/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism
11.
J Mol Neurosci ; 73(11-12): 996-1009, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37982993

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive, uncurable neurodegenerative disorder characterized by the degradation of motor neurons leading to muscle impairment, failure, and death. Senataxin, encoded by the SETX gene, is a human helicase protein whose mutations have been linked with ALS onset, particularly in its juvenile ALS4 form. Using senataxin's yeast homolog Sen1 as a model for study, it is suggested that senataxin's N-terminus interacts with RNA polymerase II, whilst its C-terminus engages in helicase activity. Senataxin is heavily involved in transcription regulation, termination, and R-loop resolution, enabled by recruitment and interactions with enzymes such as ubiquitin protein ligase SAN1 and ribonuclease H (RNase H). Senataxin also engages in DNA damage response (DDR), primarily interacting with the exosome subunit Rrp45. The Sen1 mutation E1597K, alongside the L389S and R2136H gain-of-function mutations to senataxin, is shown to cause negative structural and thus functional effects to the protein, thus contributing to a disruption in WT functions, motor neuron (MN) degeneration, and the manifestation of ALS clinical symptoms. This review corroborates and summarizes published papers concerning the structure and function of senataxin as well as the effects of their mutations in ALS pathology in order to compile current knowledge and provide a reference for future research. The findings compiled in this review are indicative of the experimental and therapeutic potential of senataxin and its mutations as a target in future ALS treatment/cure discovery, with some potential therapeutic routes also being discussed in the review.


Subject(s)
Amyotrophic Lateral Sclerosis , Humans , Amyotrophic Lateral Sclerosis/metabolism , Motor Neurons/metabolism , Gene Expression Regulation , Mutation , DNA Helicases/genetics , RNA Helicases/genetics , RNA Helicases/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism
12.
J Mol Biol ; 435(24): 168338, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37923120

ABSTRACT

To facilitate the eukaryotic repriming pathway of DNA damage tolerance, PrimPol synthesises de novo oligonucleotide primers downstream of polymerase-stalling obstacles. These primers enable replicative polymerases to resume synthesis and ensure the timely completion of DNA replication. Initiating synthesis de novo requires the coordination of single-stranded DNA, initiating nucleotides, and metal ions within PrimPol's active site to catalyze the formation of the first phosphodiester bond. Here we examine the interactions between human PrimPol's catalytic domain, nucleotides, and DNA template during each of the various catalytic steps to determine the 'choreography' of primer synthesis, where substrates bind in an ordered manner. Our findings show that the ability of PrimPol to conduct de novo primer synthesis is underpinned by a network of stabilising interactions between the enzyme, template, and nucleotides, as we previously observed for related primase CRISPR-Associated Prim-Pol (CAPP). Together, these findings establish a detailed model for the initiation of DNA synthesis by human PrimPol, which appears highly conserved.


Subject(s)
Catalytic Domain , DNA Replication , DNA-Directed DNA Polymerase , Humans , DNA Primase/metabolism , DNA, Single-Stranded/genetics , DNA-Directed DNA Polymerase/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Nucleotides
13.
Biochemistry (Mosc) ; 88(8): 1139-1155, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37758313

ABSTRACT

Transmission of genetic information depends on successful completion of DNA replication. Genomic DNA is subjected to damage on a daily basis. DNA lesions create obstacles for DNA polymerases and can lead to the replication blockage, formation of DNA breaks, cell cycle arrest, and apoptosis. Cells have evolutionary adapted to DNA damage by developing mechanisms allowing elimination of lesions prior to DNA replication (DNA repair) and helping to bypass lesions during DNA synthesis (DNA damage tolerance). The second group of mechanisms includes the restart of DNA synthesis at the sites of DNA damage by DNA primase-polymerase PrimPol. Human PrimPol was described in 2013. The properties and functions of this enzyme have been extensively studied in recent years, but very little is known about the regulation of PrimPol and association between the enzyme dysfunction and diseases. In this review, we described the mechanisms of human PrimPol regulation in the context of DNA replication, discussed in detail interactions of PrimPol with other proteins, and proposed possible pathways for the regulation of human PrimPol activity. The article also addresses the association of PrimPol dysfunction with human diseases.


Subject(s)
DNA Primase , DNA-Directed DNA Polymerase , Humans , DNA Primase/genetics , DNA Primase/metabolism , DNA-Directed DNA Polymerase/metabolism , DNA Replication , DNA/metabolism , DNA Damage , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism
14.
World J Microbiol Biotechnol ; 39(8): 214, 2023 May 31.
Article in English | MEDLINE | ID: mdl-37256388

ABSTRACT

Studying the straw lignocellulose strengthening mechanism during simultaneous degradation has important practical significance for improving resource utilization and reducing environmental pollution. In this paper, the degradation ability of four straw lignocellulose-degrading enzymes was evaluated by molecular docking and molecular dynamics. Using the significantly binds to straw lignocellulose-degrading enzyme as a template, a multifunctional lignocellulose-degrading enzyme 3CBH-1KS5-4XQD-1B85 was constructed based on amino acid recombination and homologous modeling. Five efficient degrading enzymes (3CBH-1, 3CBH-2, 3CBH-3, 3CBH-4, and 3CBH-5) were designed by site-directed mutagenesis of 3CBH-1KS5-4XQD-1B85 amino acid at position 346. Molecular dynamics showed that the degradation ability of 3CBH-1 was significant and it was 1.45 times higher than 3CBH-1KS5-4XQD-1B85. Moreover, the mechanism of enhanced degradability and the stability of the enzymes were explored. With the aid of Taguchi experiments, the suitable external environment for degrading straw was determined. In the presence of inhibitors (organic acids and phenolic compounds), the binding energy of 3CBH-1 (238.46 ± 30.96 kJ/mol) is 36.42% higher than that of 3CBH-1KS5-4XQD-1B85 (174.79 ± 20.35 kJ/mol) without external environmental stimulation. Based on homology modeling, this paper constructed a site-directed mutagenesis scheme of multifunctional enzymes, and the aim was to obtain multifunctional and efficient straw lignocellulose-degrading enzymes through protein engineering, which provided a feasible scheme for straw biodegradation.


Subject(s)
Molecular Dynamics Simulation , Multifunctional Enzymes , Multifunctional Enzymes/metabolism , Molecular Docking Simulation , Lignin/metabolism , Amino Acids
15.
Sci Adv ; 9(15): eade7997, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37058556

ABSTRACT

Recent studies have described a DNA damage tolerance pathway choice that involves a competition between PrimPol-mediated repriming and fork reversal. Screening different translesion DNA synthesis (TLS) polymerases by the use of tools for their depletion, we identified a unique role of Pol ι in regulating such a pathway choice. Pol ι deficiency unleashes PrimPol-dependent repriming, which accelerates DNA replication in a pathway that is epistatic with ZRANB3 knockdown. In Pol ι-depleted cells, the excess participation of PrimPol in nascent DNA elongation reduces replication stress signals, but thereby also checkpoint activation in S phase, triggering chromosome instability in M phase. This TLS-independent function of Pol ι requires its PCNA-interacting but not its polymerase domain. Our findings unravel an unanticipated role of Pol ι in protecting the genome stability of cells from detrimental changes in DNA replication dynamics caused by PrimPol.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase , Humans , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , DNA/genetics , DNA/metabolism , DNA Repair , DNA Damage , Chromosomal Instability , DNA Primase/genetics , DNA Primase/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism
16.
Nucleic Acids Res ; 51(6): 2818-2837, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36864660

ABSTRACT

Prolonged pausing of the transcription machinery may lead to the formation of three-stranded nucleic acid structures, called R-loops, typically resulting from the annealing of the nascent RNA with the template DNA. Unscheduled persistence of R-loops and RNA polymerases may interfere with transcription itself and other essential processes such as DNA replication and repair. Senataxin (SETX) is a putative helicase, mutated in two neurodegenerative disorders, which has been implicated in the control of R-loop accumulation and in transcription termination. However, understanding the precise role of SETX in these processes has been precluded by the absence of a direct characterisation of SETX biochemical activities. Here, we purify and characterise the helicase domain of SETX in parallel with its yeast orthologue, Sen1. Importantly, we show that SETX is a bona fide helicase with the ability to resolve R-loops. Furthermore, SETX has retained the transcription termination activity of Sen1 but functions in a species-specific manner. Finally, subsequent characterisation of two SETX variants harbouring disease-associated mutations shed light into the effect of such mutations on SETX folding and biochemical properties. Altogether, these results broaden our understanding of SETX function in gene expression and the maintenance of genome integrity and provide clues to elucidate the molecular basis of SETX-associated neurodegenerative diseases.


Subject(s)
DNA Helicases , RNA Helicases , Transcription Termination, Genetic , Humans , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Expression Regulation , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Neurodegenerative Diseases , R-Loop Structures , RNA Helicases/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
17.
Int J Mol Sci ; 24(6)2023 Mar 18.
Article in English | MEDLINE | ID: mdl-36982902

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is regarded as a fatal neurodegenerative disease that is featured by progressive damage of the upper and lower motor neurons. To date, over 45 genes have been found to be connected with ALS pathology. The aim of this work was to computationally identify unique sets of protein hydrolysate peptides that could serve as therapeutic agents against ALS. Computational methods which include target prediction, protein-protein interaction, and peptide-protein molecular docking were used. The results showed that the network of critical ALS-associated genes consists of ATG16L2, SCFD1, VAC15, VEGFA, KEAP1, KIF5A, FIG4, TUBA4A, SIGMAR1, SETX, ANXA11, HNRNPL, NEK1, C9orf72, VCP, RPSA, ATP5B, and SOD1 together with predicted kinases such as AKT1, CDK4, DNAPK, MAPK14, and ERK2 in addition to transcription factors such as MYC, RELA, ZMIZ1, EGR1, TRIM28, and FOXA2. The identified molecular targets of the peptides that support multi-metabolic components in ALS pathogenesis include cyclooxygenase-2, angiotensin I-converting enzyme, dipeptidyl peptidase IV, X-linked inhibitor of apoptosis protein 3, and endothelin receptor ET-A. Overall, the results showed that AGL, APL, AVK, IIW, PVI, and VAY peptides are promising candidates for further study. Future work would be needed to validate the therapeutic properties of these hydrolysate peptides by in vitro and in vivo approaches.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Humans , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Molecular Docking Simulation , NF-E2-Related Factor 2/metabolism , Peptides/pharmacology , Peptides/metabolism , Superoxide Dismutase-1/genetics , DNA Helicases/metabolism , RNA Helicases/metabolism , Multifunctional Enzymes/metabolism , Kinesins/metabolism , Flavoproteins/metabolism
18.
Nature ; 613(7942): 187-194, 2023 01.
Article in English | MEDLINE | ID: mdl-36544021

ABSTRACT

R-loops are RNA-DNA-hybrid-containing nucleic acids with important cellular roles. Deregulation of R-loop dynamics can lead to DNA damage and genome instability1, which has been linked to the action of endonucleases such as XPG2-4. However, the mechanisms and cellular consequences of such processing have remained unclear. Here we identify a new population of RNA-DNA hybrids in the cytoplasm that are R-loop-processing products. When nuclear R-loops were perturbed by depleting the RNA-DNA helicase senataxin (SETX) or the breast cancer gene BRCA1 (refs. 5-7), we observed XPG- and XPF-dependent cytoplasmic hybrid formation. We identify their source as a subset of stable, overlapping nuclear hybrids with a specific nucleotide signature. Cytoplasmic hybrids bind to the pattern recognition receptors cGAS and TLR3 (ref. 8), activating IRF3 and inducing apoptosis. Excised hybrids and an R-loop-induced innate immune response were also observed in SETX-mutated cells from patients with ataxia oculomotor apraxia type 2 (ref. 9) and in BRCA1-mutated cancer cells10. These findings establish RNA-DNA hybrids as immunogenic species that aberrantly accumulate in the cytoplasm after R-loop processing, linking R-loop accumulation to cell death through the innate immune response. Aberrant R-loop processing and subsequent innate immune activation may contribute to many diseases, such as neurodegeneration and cancer.


Subject(s)
Cytoplasm , DNA , Innate Immunity Recognition , Nucleic Acid Heteroduplexes , R-Loop Structures , RNA , Humans , Apoptosis , Cytoplasm/immunology , Cytoplasm/metabolism , DNA/chemistry , DNA/immunology , DNA Helicases/genetics , DNA Helicases/metabolism , Genes, BRCA1 , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Mutation , Neoplasms , Nucleic Acid Heteroduplexes/chemistry , Nucleic Acid Heteroduplexes/immunology , R-Loop Structures/immunology , RNA/chemistry , RNA/immunology , RNA Helicases/genetics , RNA Helicases/metabolism , Spinocerebellar Ataxias/genetics
19.
Commun Biol ; 5(1): 1395, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36543851

ABSTRACT

Replication stress (RS) is a leading cause of genome instability and cancer development. A substantial source of endogenous RS originates from the encounter between the transcription and replication machineries operating on the same DNA template. This occurs predominantly under specific contexts, such as oncogene activation, metabolic stress, or a deficiency in proteins that specifically act to prevent or resolve those transcription-replication conflicts (TRCs). One such protein is Senataxin (SETX), an RNA:DNA helicase involved in resolution of TRCs and R-loops. Here we identify a synthetic lethal interaction between SETX and proteins of the Fanconi anemia (FA) pathway. Depletion of SETX induces spontaneous under-replication and chromosome fragility due to active transcription and R-loops that persist in mitosis. These fragile loci are targeted by the Fanconi anemia protein, FANCD2, to facilitate the resolution of under-replicated DNA, thus preventing chromosome mis-segregation and allowing cells to proliferate. Mechanistically, we show that FANCD2 promotes mitotic DNA synthesis that is dependent on XPF and MUS81 endonucleases. Importantly, co-depleting FANCD2 together with SETX impairs cancer cell proliferation, without significantly affecting non-cancerous cells. Therefore, we uncovered a synthetic lethality between SETX and FA proteins for tolerance of transcription-mediated RS that may be exploited for cancer therapy.


Subject(s)
DNA Helicases , Fanconi Anemia Complementation Group D2 Protein , Neoplasms , RNA Helicases , Humans , DNA , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Replication , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Neoplasms/genetics , Neoplasms/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism
20.
mBio ; 13(6): e0308722, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36468882

ABSTRACT

Expression of the fission yeast Schizosaccharomyces pombe phosphate regulon is sensitive to the intracellular level of the inositol pyrophosphate signaling molecule 1,5-IP8. IP8 dynamics are determined by Asp1, a bifunctional enzyme consisting of an N-terminal kinase domain and a C-terminal pyrophosphatase domain that catalyze IP8 synthesis and catabolism, respectively. Here, we report structures of the Asp1 kinase domain, crystallized with two protomers in the asymmetric unit, one of which was complexed with ligands (ADPNP, ADP, or ATP; Mg2+ or Mn2+; IP6, 5-IP7, or 1,5-IP8) and the other which was ligand-free. The ligand-free enzyme adopts an "open" conformation that allows ingress of substrates and egress of products. ADPNP, ADP, and ATP and associated metal ions occupy a deep phospho-donor pocket in the active site. IP6 or 5-IP7 engagement above the nucleotide favors adoption of a "closed" conformation, in which surface protein segments undergo movement and a disordered-to-ordered transition to form an inositol polyphosphate-binding site. In a structure mimetic of the kinase Michaelis complex, the anionic 5-IP7 phosphates are encaged by an ensemble of nine cationic amino acids: Lys43, Arg223, Lys224, Lys260, Arg274, Arg285, Lys290, Arg293, and Lys341. Alanine mutagenesis of amino acids that contact the adenosine nucleoside of the ATP donor underscored the contributions of Asp258 interaction with the ribose 3'-OH and of Glu248 with adenine-N6. Changing Glu248 to Gln elicited a gain of function whereby the kinase became adept at using GTP as phosphate donor. Wild-type Asp1 kinase can utilize N6-benzyl-ATP as phosphate donor. IMPORTANCE The inositol pyrophosphate signaling molecule 1,5-IP8 modulates fission yeast phosphate homeostasis via its action as an agonist of RNA 3'-processing and transcription termination. Cellular IP8 levels are determined by Asp1, a bifunctional enzyme composed of an N-terminal kinase and a C-terminal pyrophosphatase domain. Here, we present a series of crystal structures of the Asp1 kinase domain, in a ligand-free state and in complexes with nucleotides ADPNP, ADP, and ATP, divalent cations magnesium and manganese, and inositol polyphosphates IP6, 5-IP7, and 1,5-IP8. Substrate binding elicits a switch from open to closed conformations, entailing a disordered-to-ordered transition and a rearrangement or movement of two peptide segments that form a binding site for the phospho-acceptor. Our structures, along with structure-guided mutagenesis, fortify understanding of the mechanism and substrate specificity of Asp1 kinase, and they extend and complement structural and functional studies of the orthologous human kinase PPIP5K2.


Subject(s)
Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Humans , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Diphosphates/metabolism , Inositol Phosphates/metabolism , Multifunctional Enzymes/metabolism , Phosphotransferases (Phosphate Group Acceptor)/genetics , Pyrophosphatases/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...