Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 9.764
1.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article En | MEDLINE | ID: mdl-38673980

Checkpoint kinase 1 (Chk1) is a key mediator of the DNA damage response that regulates cell cycle progression, DNA damage repair, and DNA replication. Small-molecule Chk1 inhibitors sensitize cancer cells to genotoxic agents and have shown preclinical activity as single agents in cancers characterized by high levels of replication stress. However, the underlying genetic determinants of Chk1-inhibitor sensitivity remain unclear. Although treatment options for advanced colorectal cancer are limited, radiotherapy is effective. Here, we report that exposure to a novel amidine derivative, K1586, leads to an initial reduction in the proliferative potential of colorectal cancer cells. Cell cycle analysis revealed that the length of the G2/M phase increased with K1586 exposure as a result of Chk1 instability. Exposure to K1586 enhanced the degradation of Chk1 in a time- and dose-dependent manner, increasing replication stress and sensitizing colorectal cancer cells to radiation. Taken together, the results suggest that a novel amidine derivative may have potential as a radiotherapy-sensitization agent that targets Chk1.


Amidines , Checkpoint Kinase 1 , Colorectal Neoplasms , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 1/antagonists & inhibitors , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/radiotherapy , Amidines/pharmacology , Cell Line, Tumor , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , DNA Replication/drug effects , Cell Proliferation/drug effects , DNA Damage/drug effects , Cell Cycle/drug effects
2.
Nature ; 628(8007): 433-441, 2024 Apr.
Article En | MEDLINE | ID: mdl-38509368

An important advance in cancer therapy has been the development of poly(ADP-ribose) polymerase (PARP) inhibitors for the treatment of homologous recombination (HR)-deficient cancers1-6. PARP inhibitors trap PARPs on DNA. The trapped PARPs are thought to block replisome progression, leading to formation of DNA double-strand breaks that require HR for repair7. Here we show that PARP1 functions together with TIMELESS and TIPIN to protect the replisome in early S phase from transcription-replication conflicts. Furthermore, the synthetic lethality of PARP inhibitors with HR deficiency is due to an inability to repair DNA damage caused by transcription-replication conflicts, rather than by trapped PARPs. Along these lines, inhibiting transcription elongation in early S phase rendered HR-deficient cells resistant to PARP inhibitors and depleting PARP1 by small-interfering RNA was synthetic lethal with HR deficiency. Thus, inhibiting PARP1 enzymatic activity may suffice for treatment efficacy in HR-deficient settings.


DNA Replication , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases , Transcription, Genetic , Humans , DNA Breaks, Double-Stranded , DNA Replication/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Recombinational DNA Repair , S Phase , Transcription, Genetic/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/metabolism
3.
J Biol Chem ; 300(4): 105786, 2024 Apr.
Article En | MEDLINE | ID: mdl-38401843

Histone proteins can become trapped on DNA in the presence of 5-formylcytosine (5fC) to form toxic DNA-protein conjugates. Their repair may involve proteolytic digestion resulting in DNA-peptide cross-links (DpCs). Here, we have investigated replication of a model DpC comprised of an 11-mer peptide (NH2-GGGKGLGK∗GGA) containing an oxy-lysine residue (K∗) conjugated to 5fC in DNA. Both CXG and CXT (where X = 5fC-DpC) sequence contexts were examined. Replication of both constructs gave low viability (<10%) in Escherichia coli, whereas TLS efficiency was high (72%) in HEK 293T cells. In E. coli, the DpC was bypassed largely error-free, inducing only 2 to 3% mutations, which increased to 4 to 5% with SOS. For both sequences, semi-targeted mutations were dominant, and for CXG, the predominant mutations were G→T and G→C at the 3'-base to the 5fC-DpC. In HEK 293T cells, 7 to 9% mutations occurred, and the dominant mutations were the semi-targeted G → T for CXG and T → G for CXT. These mutations were reduced drastically in cells deficient in hPol η, hPol ι or hPol ζ, suggesting a role of these TLS polymerases in mutagenic TLS. Steady-state kinetics studies using hPol η confirmed that this polymerase induces G → T and T → G transversions at the base immediately 3' to the DpC. This study reveals a unique replication pattern of 5fC-conjugated DpCs, which are bypassed largely error-free in both E. coli and human cells and induce mostly semi-targeted mutations at the 3' position to the lesion.


Cytosine , Cytosine/analogs & derivatives , DNA , Escherichia coli , Mutation , Humans , Escherichia coli/metabolism , Escherichia coli/genetics , HEK293 Cells , Cytosine/metabolism , Cytosine/chemistry , DNA/metabolism , DNA/chemistry , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , DNA Replication/drug effects
4.
Nucleic Acids Res ; 52(8): 4295-4312, 2024 May 08.
Article En | MEDLINE | ID: mdl-38416579

5-Fluorouracil (5-FU) is the first-line chemotherapeutic agent in colorectal cancer, and resistance to 5-FU easily emerges. One of the mechanisms of drug action and resistance of 5-FU is through DNA incorporation. Our quantitative reverse-transcription PCR data showed that one of the translesion synthesis (TLS) DNA polymerases, DNA polymerase η (polη), was upregulated within 72 h upon 5-FU administration at 1 and 10 µM, indicating that polη is one of the first responding polymerases, and the only TLS polymerase, upon the 5-FU treatment to incorporate 5-FU into DNA. Our kinetic studies revealed that 5-fluoro-2'-deoxyuridine triphosphate (5FdUTP) was incorporated across dA 41 and 28 times more efficiently than across dG and across inosine, respectively, by polη indicating that the mutagenicity of 5-FU incorporation is higher in the presence of inosine and that DNA lesions could lead to more mutagenic incorporation of 5-FU. Our polη crystal structures complexed with DNA and 5FdUTP revealed that dA:5FdUTP base pair is like dA:dTTP in the active site of polη, while 5FdUTP adopted 4-enol tautomer in the base pairs with dG and HX increasing the insertion efficiency compared to dG:dTTP for the incorrect insertions. These studies confirm that polη engages in the DNA incorporation and bypass of 5-FU.


Colorectal Neoplasms , DNA-Directed DNA Polymerase , Fluorouracil , Fluorouracil/pharmacology , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Humans , DNA Damage , DNA/metabolism , DNA/chemistry , DNA/biosynthesis , DNA Repair , Deoxyuracil Nucleotides/metabolism , Deoxyuracil Nucleotides/chemistry , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/chemistry , Kinetics , DNA Replication/drug effects , Drug Resistance, Neoplasm/genetics , Translesion DNA Synthesis
5.
J Biol Chem ; 299(12): 105385, 2023 Dec.
Article En | MEDLINE | ID: mdl-37890780

Cancer is a genetic disease requiring multiple mutations for its development. However, many carcinogens are DNA-unreactive and nonmutagenic and consequently described as nongenotoxic. One of such carcinogens is nickel, a global environmental pollutant abundantly emitted by burning of coal. We investigated activation of DNA damage responses by Ni and identified this metal as a replication stressor. Genotoxic stress markers indicated the accumulation of ssDNA and stalled replication forks, and Ni-treated cells were dependent on ATR for suppression of DNA damage and long-term survival. Replication stress by Ni resulted from destabilization of RRM1 and RRM2 subunits of ribonucleotide reductase and the resulting deficiency in dNTPs. Ni also increased DNA incorporation of rNMPs (detected by a specific fluorescent assay) and strongly enhanced their genotoxicity as a result of repressed repair of TOP1-DNA protein crosslinks (TOP1-DPC). The DPC-trap assay found severely impaired SUMOylation and K48-polyubiquitination of DNA-crosslinked TOP1 due to downregulation of specific enzymes. Our findings identified Ni as the human carcinogen inducing genome instability via DNA-embedded ribonucleotides and accumulation of TOP1-DPC which are carcinogenic abnormalities with poor detectability by the standard mutagenicity tests. The discovered mechanisms for Ni could also play a role in genotoxicity of other protein-reactive carcinogens.


Carcinogens , DNA Replication , Nickel , Nucleotides , Humans , Carcinogens/toxicity , DNA/metabolism , DNA Damage , DNA Repair , DNA Replication/drug effects , DNA Topoisomerases, Type I/metabolism , Nickel/toxicity , Saccharomyces cerevisiae/metabolism , Nucleotides/biosynthesis
6.
Nature ; 622(7981): 180-187, 2023 Oct.
Article En | MEDLINE | ID: mdl-37648864

Antibiotic binding sites are located in important domains of essential enzymes and have been extensively studied in the context of resistance mutations; however, their study is limited by positive selection. Using multiplex genome engineering1 to overcome this constraint, we generate and characterize a collection of 760 single-residue mutants encompassing the entire rifampicin binding site of Escherichia coli RNA polymerase (RNAP). By genetically mapping drug-enzyme interactions, we identify an alpha helix where mutations considerably enhance or disrupt rifampicin binding. We find mutations in this region that prolong antibiotic binding, converting rifampicin from a bacteriostatic to bactericidal drug by inducing lethal DNA breaks. The latter are replication dependent, indicating that rifampicin kills by causing detrimental transcription-replication conflicts at promoters. We also identify additional binding site mutations that greatly increase the speed of RNAP.Fast RNAP depletes the cell of nucleotides, alters cell sensitivity to different antibiotics and provides a cold growth advantage. Finally, by mapping natural rpoB sequence diversity, we discover that functional rifampicin binding site mutations that alter RNAP properties or confer drug resistance occur frequently in nature.


Anti-Bacterial Agents , Binding Sites , DNA-Directed RNA Polymerases , Escherichia coli , Mutation , Rifampin , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Binding Sites/drug effects , Binding Sites/genetics , DNA Breaks/drug effects , DNA Replication/drug effects , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Nucleotides/deficiency , Nucleotides/metabolism , Promoter Regions, Genetic , Rifampin/chemistry , Rifampin/metabolism , Rifampin/pharmacology , Time Factors , Transcription, Genetic/drug effects
7.
J Virol ; 97(6): e0037023, 2023 06 29.
Article En | MEDLINE | ID: mdl-37219458

DNA replication of E1-deleted first-generation adenoviruses (AdV) in cultured cancer cells has been reported repeatedly and it was suggested that certain cellular proteins could functionally compensate for E1A, leading to the expression of the early region 2 (E2)-encoded proteins and subsequently virus replication. Referring to this, the observation was named E1A-like activity. In this study, we investigated different cell cycle inhibitors with respect to their ability to increase viral DNA replication of dl70-3, an E1-deleted adenovirus. Our analyses of this issue revealed that in particular inhibition of cyclin-dependent kinases 4/6 (CDK4/6i) increased E1-independent adenovirus E2-expression and viral DNA replication. Detailed analysis of the E2-expression in dl70-3 infected cells by RT-qPCR showed that the increase in E2-expression originated from the E2-early promoter. Mutations of the two E2F-binding sites in the E2-early promoter (pE2early-LucM) caused a significant reduction in E2-early promoter activity in trans-activation assays. Accordingly, mutations of the E2F-binding sites in the E2-early promoter in a virus named dl70-3/E2Fm completely abolished CDK4/6i induced viral DNA replication. Thus, our data show that E2F-binding sites in the E2-early promoter are crucial for E1A independent adenoviral DNA replication of E1-deleted vectors in cancer cells. IMPORTANCE E1-deleted AdV vectors are considered replication deficient and are important tools for the study of virus biology, gene therapy, and large-scale vaccine development. However, deletion of the E1 genes does not completely abolish viral DNA replication in cancer cells. Here, we report, that the two E2F-binding sites in the adenoviral E2-early promoter contribute substantially to the so-called E1A-like activity in tumor cells. With this finding, on the one hand, the safety profile of viral vaccine vectors can be increased and, on the other hand, the oncolytic property for cancer therapy might be improved through targeted manipulation of the host cell.


Adenoviridae , Cell Cycle , DNA Replication , Virus Replication , Adenoviridae/genetics , Adenoviridae/metabolism , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Binding Sites , Cell Cycle/drug effects , Cell Line, Tumor , Cells/drug effects , Cells/virology , DNA Replication/drug effects , DNA, Viral/metabolism , Gene Expression Regulation, Viral/drug effects , Mutation , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/pharmacology , Virus Replication/physiology , Humans
8.
Nucleic Acids Res ; 51(16): 8532-8549, 2023 09 08.
Article En | MEDLINE | ID: mdl-37216608

Friedreich's ataxia (FRDA) is caused by expansions of GAA•TTC repeats in the first intron of the human FXN gene that occur during both intergenerational transmissions and in somatic cells. Here we describe an experimental system to analyze large-scale repeat expansions in cultured human cells. It employs a shuttle plasmid that can replicate from the SV40 origin in human cells or be stably maintained in S. cerevisiae utilizing ARS4-CEN6. It also contains a selectable cassette allowing us to detect repeat expansions that accumulated in human cells upon plasmid transformation into yeast. We indeed observed massive expansions of GAA•TTC repeats, making it the first genetically tractable experimental system to study large-scale repeat expansions in human cells. Further, GAA•TTC repeats stall replication fork progression, while the frequency of repeat expansions appears to depend on proteins implicated in replication fork stalling, reversal, and restart. Locked nucleic acid (LNA)-DNA mixmer oligonucleotides and peptide nucleic acid (PNA) oligomers, which interfere with triplex formation at GAA•TTC repeats in vitro, prevented the expansion of these repeats in human cells. We hypothesize, therefore, that triplex formation by GAA•TTC repeats stall replication fork progression, ultimately leading to repeat expansions during replication fork restart.


Friedreich Ataxia , Oligonucleotides , Peptide Nucleic Acids , Trinucleotide Repeat Expansion , Humans , DNA , DNA Replication/drug effects , Friedreich Ataxia/genetics , Iron-Binding Proteins/genetics , Oligonucleotides/pharmacology , Peptide Nucleic Acids/pharmacology , Saccharomyces cerevisiae/genetics
9.
Nat Commun ; 14(1): 381, 2023 01 24.
Article En | MEDLINE | ID: mdl-36693839

Fanconi Anemia (FA) is a rare, genome instability-associated disease characterized by a deficiency in repairing DNA crosslinks, which are known to perturb several cellular processes, including DNA transcription, replication, and repair. Formaldehyde, a by-product of metabolism, is thought to drive FA by generating DNA interstrand crosslinks (ICLs) and DNA-protein crosslinks (DPCs). However, the impact of formaldehyde on global cellular pathways has not been investigated thoroughly. Herein, using a pangenomic CRISPR-Cas9 screen, we identify EXO1 as a critical regulator of formaldehyde-induced DNA lesions. We show that EXO1 knockout cell lines exhibit formaldehyde sensitivity leading to the accumulation of replicative stress, DNA double-strand breaks, and quadriradial chromosomes, a typical feature of FA. After formaldehyde exposure, EXO1 is recruited to chromatin, protects DNA replication forks from degradation, and functions in parallel with the FA pathway to promote cell survival. In vitro, EXO1-mediated exonuclease activity is proficient in removing DPCs. Collectively, we show that EXO1 limits replication stress and DNA damage to counteract formaldehyde-induced genome instability.


CRISPR-Cas Systems , Drug Tolerance , Exodeoxyribonucleases , Fanconi Anemia , Formaldehyde , Humans , DNA , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair/drug effects , DNA Repair/genetics , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA Replication/drug effects , DNA Replication/genetics , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Fanconi Anemia/chemically induced , Fanconi Anemia/genetics , Formaldehyde/toxicity , Genomic Instability/drug effects , Genomic Instability/genetics , Drug Tolerance/genetics
10.
Sci Rep ; 12(1): 3093, 2022 02 23.
Article En | MEDLINE | ID: mdl-35197531

Development of chemotherapy has led to a high survival rate of cancer patients; however, the severe side effects of anticancer drugs, including organ hypoplasia, persist. To assume the side effect of anticancer drugs, we established a new ex vivo screening model and described a method for suppressing side effects. Cyclophosphamide (CPA) is a commonly used anticancer drug and causes severe side effects in developing organs with intensive proliferation, including the teeth and hair. Using the organ culture model, we found that treatment with CPA disturbed the growth of tooth germs by inducing DNA damage, apoptosis and suppressing cellular proliferation and differentiation. Furthermore, low temperature suppressed CPA-mediated inhibition of organ development. Our ex vivo and in vitro analysis revealed that low temperature impeded Rb phosphorylation and caused cell cycle arrest at the G1 phase during CPA treatment. This can prevent the CPA-mediated cell damage of DNA replication caused by the cross-linking reaction of CPA. Our findings suggest that the side effects of anticancer drugs on organ development can be avoided by maintaining the internal environment under low temperature.


Antineoplastic Agents/adverse effects , Cyclophosphamide/adverse effects , Temperature , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , DNA Damage/drug effects , DNA Replication/drug effects , G1 Phase/drug effects , Humans , Models, Biological , Organ Culture Techniques
11.
Cell Death Dis ; 13(2): 96, 2022 02 02.
Article En | MEDLINE | ID: mdl-35110528

Replication stress (RS) has a pivotal role in tumor initiation, progression, or therapeutic resistance. In this study, we depicted the mechanism of breast cancer stem cells' (bCSCs) response to RS and its clinical implication. We demonstrated that bCSCs present a limited level of RS compared with non-bCSCs in patient samples. We described for the first time that the spatial nuclear location of BMI1 protein triggers RS response in breast cancers. Hence, in bCSCs, BMI1 is rapidly located to stalled replication forks to recruit RAD51 and activate homologous-recombination machinery, whereas in non-bCSCs BMI1 is trapped on demethylated 1q12 megasatellites precluding effective RS response. We further demonstrated that BMI1/RAD51 axis activation is necessary to prevent cisplatin-induced DNA damage and that treatment of patient-derived xenografts with a RAD51 inhibitor sensitizes tumor-initiating cells to cisplatin. The comprehensive view of replicative-stress response in bCSC has profound implications for understanding and improving therapeutic resistance.


Breast Neoplasms/pathology , Cell Nucleus/metabolism , Drug Resistance, Neoplasm/drug effects , Neoplastic Stem Cells/drug effects , Polycomb Repressive Complex 1/metabolism , Rad51 Recombinase/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cisplatin/pharmacology , DNA Damage/drug effects , DNA Replication/drug effects , Drug Resistance, Neoplasm/genetics , Female , Homologous Recombination , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/antagonists & inhibitors , Rad51 Recombinase/antagonists & inhibitors
12.
Int J Mol Sci ; 23(4)2022 Feb 15.
Article En | MEDLINE | ID: mdl-35216252

Mosaicism is the most important limitation for one-step gene editing in embryos by CRISPR/Cas9 because cuts and repairs sometimes take place after the first DNA replication of the zygote. To try to minimize the risk of mosaicism, in this study a reversible DNA replication inhibitor was used after the release of CRISPR/Cas9 in the cell. There is no previous information on the use of aphidicolin in porcine embryos, so the reversible inhibition of DNA replication and the effect on embryo development of different concentrations of this drug was first evaluated. The effect of incubation with aphidicolin was tested with CRISPR/Cas9 at different concentrations and different delivery methodologies. As a result, the reversible inhibition of DNA replication was observed, and it was concentration dependent. An optimal concentration of 0.5 µM was established and used for subsequent experiments. Following the use of this drug with CRISPR/Cas9, a halving of mosaicism was observed together with a detrimental effect on embryo development. In conclusion, the use of reversible inhibition of DNA replication offers a way to reduce mosaicism. Nevertheless, due to the reduction in embryo development, it would be necessary to reach a balance for its use to be feasible.


Aphidicolin/pharmacology , CRISPR-Cas Systems/drug effects , Cell Nucleus/drug effects , DNA Replication/drug effects , Embryo, Mammalian/drug effects , Eukaryota/drug effects , Animals , Animals, Genetically Modified , Embryonic Development/drug effects , Gene Editing/methods , Mosaicism/drug effects , Swine , Zygote/drug effects
13.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article En | MEDLINE | ID: mdl-35055077

Whilst avoidance of chemical modifications of DNA bases is essential to maintain genome stability, during evolution eukaryotic cells have evolved a chemically reversible modification of the cytosine base. These dynamic methylation and demethylation reactions on carbon-5 of cytosine regulate several cellular and developmental processes such as embryonic stem cell pluripotency, cell identity, differentiation or tumourgenesis. Whereas these physiological processes are well characterized, very little is known about the toxicity of these cytosine analogues when they incorporate during replication. Here, we report a role of the base excision repair factor XRCC1 in protecting replication fork upon incorporation of 5-hydroxymethyl-2'-deoxycytosine (5hmC) and its deamination product 5-hydroxymethyl-2'-deoxyuridine (5hmU) during DNA synthesis. In the absence of XRCC1, 5hmC exposure leads to increased genomic instability, replication fork impairment and cell lethality. Moreover, the 5hmC deamination product 5hmU recapitulated the genomic instability phenotypes observed by 5hmC exposure, suggesting that 5hmU accounts for the observed by 5hmC exposure. Remarkably, 5hmC-dependent genomic instability and replication fork impairment seen in Xrcc1-/- cells were exacerbated by the trapping of Parp1 on chromatin, indicating that XRCC1 maintains replication fork stability during processing of 5hmC and 5hmU by the base excision repair pathway. Our findings uncover natural epigenetic DNA bases 5hmC and 5hmU as genotoxic nucleosides that threaten replication dynamics and genome integrity in the absence of XRCC1.


DNA Demethylation , DNA Replication , Deoxycytidine/analogs & derivatives , Thymidine/analogs & derivatives , X-ray Repair Cross Complementing Protein 1/genetics , 5-Methylcytosine/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage , DNA Replication/drug effects , Epigenesis, Genetic , Genomic Instability , Humans , Replication Origin , X-ray Repair Cross Complementing Protein 1/metabolism
14.
EMBO J ; 41(4): e108290, 2022 02 15.
Article En | MEDLINE | ID: mdl-35028974

Nucleotide metabolism fuels normal DNA replication and is also primarily targeted by the DNA replication checkpoint when replication stalls. To reveal a comprehensive interconnection between genome maintenance and metabolism, we analyzed the metabolomic changes upon replication stress in the budding yeast S. cerevisiae. We found that upon treatment of cells with hydroxyurea, glucose is rapidly diverted to the oxidative pentose phosphate pathway (PPP). This effect is mediated by the AMP-dependent kinase, SNF1, which phosphorylates the transcription factor Mig1, thereby relieving repression of the gene encoding the rate-limiting enzyme of the PPP. Surprisingly, NADPH produced by the PPP is required for efficient recruitment of replication protein A (RPA) to single-stranded DNA, providing the signal for the activation of the Mec1/ATR-Rad53/CHK1 checkpoint signaling kinase cascade. Thus, SNF1, best known as a central energy controller, determines a fast mode of replication checkpoint activation through a redox mechanism. These findings establish that SNF1 provides a hub with direct links to cellular metabolism, redox, and surveillance of DNA replication in eukaryotes.


DNA Replication , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA Replication/drug effects , DNA, Single-Stranded/metabolism , Glucose/genetics , Glucose/metabolism , Glycolysis/physiology , Hydroxyurea , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , NADP/metabolism , Pentose Phosphate Pathway , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Replication Protein A/genetics , Replication Protein A/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
15.
Theranostics ; 12(2): 657-674, 2022.
Article En | MEDLINE | ID: mdl-34976206

Rationale: Corneal neovascularization (CoNV) is a severe complication of various types of corneal diseases, that leads to permanent visual impairment. Current treatments for CoNV, such as steroids or anti-vascular endothelial growth factor agents, are argued over their therapeutic efficacy and adverse effects. Here, we demonstrate that transforming growth factor-ß (TGF-ß)-activated kinase 1 (TAK1) plays an important role in the pathogenesis of CoNV. Methods: Angiogenic activities were assessed in ex vivo and in vitro models subjected to TAK1 inhibition by 5Z-7-oxozeaenol, a selective inhibitor of TAK1. RNA-Seq was used to examine pathways that could be potentially affected by TAK1 inhibition. A gelatin-nanoparticles-encapsulated 5Z-7-oxozeaenol was developed as the eyedrop to treat CoNV in a rodent model. Results: We showed that 5Z-7-oxozeaenol reduced angiogenic processes through impeding cell proliferation. Transcriptome analysis suggested 5Z-7-oxozeaenol principally suppresses cell cycle and DNA replication, thereby restraining cell proliferation. In addition, inhibition of TAK1 by 5Z-7-oxozeaenol blocked TNFα-mediated NFκB signalling, and its downstream genes related to angiogenesis and inflammation. 5Z-7-oxozeaenol also ameliorated pro-angiogenic activity, including endothelial migration and tube formation. Furthermore, topical administration of the gelatin-nanoparticles-encapsulated 5Z-7-oxozeaenol led to significantly greater suppression of CoNV in a mouse model compared to the free form of 5Z-7-oxozeaenol, likely due to extended retention of 5Z-7-oxozeaenol in the cornea. Conclusion: Our study shows the potential of TAK1 as a therapeutic target for pathological angiogenesis, and the gelatin nanoparticle coupled with 5Z-7-oxozeaenol as a promising new eyedrop administration model in treatment of CoNV.


Corneal Neovascularization , Endothelium, Vascular , Lactones , MAP Kinase Kinase Kinases , Resorcinols , Animals , Humans , Male , Mice , Administration, Ophthalmic , Capsules , Cell Cycle/drug effects , Cell Line , Corneal Neovascularization/drug therapy , Cytokines/antagonists & inhibitors , DNA Replication/drug effects , Drug Delivery Systems , Endothelium, Vascular/drug effects , Gelatin , Lactones/administration & dosage , Lactones/pharmacology , Lactones/therapeutic use , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice, Inbred C57BL , Nanoparticles , Ophthalmic Solutions , Resorcinols/administration & dosage , Resorcinols/pharmacology , Resorcinols/therapeutic use , RNA-Seq
16.
Clin Exp Pharmacol Physiol ; 49(1): 134-144, 2022 01.
Article En | MEDLINE | ID: mdl-34448246

At present, there are still many problems in the treatment of lung cancer, such as high cost, side effects and low quality of life. The advantages of traditional Chinese medicine (TCM) in the treatment of lung cancer are reflected. Berberine has been increasingly popular in colorectal cancer treatment, but little is known about its bioactivity against non-small cell lung cancer (NSCLC). Cell proliferation, cell apoptosis, cDNA microarray, gene and protein expression, and NSCLC transplanted tumour growth were performed. Berberine suppressed NSCLC cell proliferation and colony formation in vitro and inhibited NSCLC tumour growth in subcutaneously transplanted tumour lung tumour models, leading to prolonged survival of tumour-bearing mice. However, berberine did not induce the cleavage of Caspase 3 and PARP1, and could not induce apoptosis in all NSCLC cells. Moreover, 646 genes were differentially expressed upon berberine administration, which were involved in seven signal pathways, such as DNA replication. In cDNA microarray, berberine downregulated the expression of RRM1, RRM2, LIG1, POLE2 that involving DNA repair and replication. Our findings demonstrate that berberine inhibits NSCLC cells growth through repressing DNA repair and replication rather than through apoptosis. Berberine could be used as a promising therapeutic candidate for NSCLC patients.


Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Berberine/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , DNA Repair/drug effects , DNA Replication/drug effects , Lung Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Berberine/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis
17.
J Virol ; 96(2): e0132621, 2022 01 26.
Article En | MEDLINE | ID: mdl-34669461

Parvovirus B19 (B19V) infection can cause hematological disorders and fetal hydrops during pregnancy. Currently, no antivirals or vaccines are available for the treatment or prevention of B19V infection. To identify novel small-molecule antivirals against B19V replication, we developed a high-throughput screening (HTS) assay, which is based on an in vitro nicking assay using recombinant N-terminal amino acids 1 to 176 of the viral large nonstructural protein (NS1N) and a fluorescently labeled DNA probe (OriQ) that spans the nicking site of the viral DNA replication origin. We collectively screened 17,040 compounds and identified 2,178 (12.78%) hits that possess >10% inhibition of the NS1 nicking activity, among which 84 hits were confirmed to inhibit nicking in a dose-dependent manner. Using ex vivo-expanded primary human erythroid progenitor cells (EPCs) infected by B19V, we validated 24 compounds that demonstrated >50% in vivo inhibition of B19V infection at 10 µM, which can be categorized into 7 structure scaffolds. Based on the therapeutic index (half-maximal cytotoxic concentration [CC50]/half-maximal effective concentration [EC50] ratio) in EPCs, the top 4 compounds were chosen to examine their inhibitions of B19V infection in EPCs at two times of the 90% maximal effective concentration (EC90). A purine derivative (P7) demonstrated an antiviral effect (EC50 = 1.46 µM) without prominent cytotoxicity (CC50 = 71.8 µM) in EPCs and exhibited 92% inhibition of B19V infection in EPCs at 3.32 µM, which can be used as the lead compound in future studies for the treatment of B19V infection-caused hematological disorders. IMPORTANCE B19V encodes a large nonstructural protein, NS1. Its N-terminal domain (NS1N) consisting of amino acids 1 to 176 binds to viral DNA and serves as an endonuclease to nick the viral DNA replication origins, which is a pivotal step in rolling-hairpin-dependent B19V DNA replication. For high-throughput screening (HTS) of anti-B19V antivirals, we miniaturized a fluorescence-based in vitro nicking assay, which employs a fluorophore-labeled probe spanning the terminal resolution site (trs) and the NS1N protein, into a 384-well-plate format. The HTS assay showed high reliability and capability in screening 17,040 compounds. Based on the therapeutic index (half-maximal cytotoxic concentration [CC50]/half-maximal effective concentration [EC50] ratio) in EPCs, a purine derivative demonstrated an antiviral effect of 92% inhibition of B19V infection in EPCs at 3.32 µM (two times the EC90). Our study demonstrated a robust HTS assay for screening antivirals against B19V infection.


Antiviral Agents/pharmacology , Erythroid Precursor Cells/virology , High-Throughput Screening Assays/methods , Parvovirus B19, Human/drug effects , Antiviral Agents/chemistry , Cell Survival/drug effects , DNA Replication/drug effects , DNA, Viral/metabolism , Erythroid Precursor Cells/drug effects , Fluorescent Dyes , Humans , Parvovirus B19, Human/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Origin , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
18.
Nat Commun ; 12(1): 6997, 2021 12 06.
Article En | MEDLINE | ID: mdl-34873176

Pathological variants of human mitochondrial DNA (mtDNA) typically co-exist with wild-type molecules, but the factors driving the selection of each are not understood. Because mitochondrial fitness does not favour the propagation of functional mtDNAs in disease states, we sought to create conditions where it would be advantageous. Glucose and glutamine consumption are increased in mtDNA dysfunction, and so we targeted the use of both in cells carrying the pathogenic m.3243A>G variant with 2-Deoxy-D-glucose (2DG), or the related 5-thioglucose. Here, we show that both compounds selected wild-type over mutant mtDNA, restoring mtDNA expression and respiration. Mechanistically, 2DG selectively inhibits the replication of mutant mtDNA; and glutamine is the key target metabolite, as its withdrawal, too, suppresses mtDNA synthesis in mutant cells. Additionally, by restricting glucose utilization, 2DG supports functional mtDNAs, as glucose-fuelled respiration is critical for mtDNA replication in control cells, when glucose and glutamine are scarce. Hence, we demonstrate that mitochondrial fitness dictates metabolite preference for mtDNA replication; consequently, interventions that restrict metabolite availability can suppress pathological mtDNAs, by coupling mitochondrial fitness and replication.


DNA Replication/drug effects , DNA, Mitochondrial/genetics , Deoxyglucose/pharmacology , Mitochondria/drug effects , Point Mutation , A549 Cells , Autophagy/drug effects , Autophagy/genetics , Cell Line, Tumor , Cells, Cultured , DNA, Mitochondrial/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Glucose/analogs & derivatives , Glucose/pharmacology , Glycolysis/drug effects , Glycolysis/genetics , Humans , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects
19.
Cells ; 10(12)2021 12 03.
Article En | MEDLINE | ID: mdl-34943918

Experiments on Vicia faba root meristem cells exposed to 150 µM cadmium chloride (CdCl2) were undertaken to analyse epigenetic changes, mainly with respect to DNA replication stress. Histone modifications examined by means of immunofluorescence labeling included: (1) acetylation of histone H3 on lysine 56 (H3K56Ac), involved in transcription, S phase, and response to DNA damage during DNA biosynthesis; (2) dimethylation of histone H3 on lysine 79 (H3K79Me2), correlated with the replication initiation; (3) phosphorylation of histone H3 on threonine 45 (H3T45Ph), engaged in DNA synthesis and apoptosis. Moreover, immunostaining using specific antibodies against 5-MetC-modified DNA was used to determine the level of DNA methylation. A significant decrease in the level of H3K79Me2, noted in all phases of the CdCl2-treated interphase cell nuclei, was found to correspond with: (1) an increase in the mean number of intranuclear foci of H3K56Ac histones (observed mainly in S-phase), (2) a plethora of nuclear and nucleolar labeling patterns (combined with a general decrease in H3T45Ph), and (3) a decrease in DNA methylation. All these changes correlate well with a general viewpoint that DNA modifications and post-translational histone modifications play an important role in gene expression and plant development under cadmium-induced stress conditions.


Cadmium/toxicity , DNA Replication/genetics , Epigenesis, Genetic , Meristem/cytology , Meristem/genetics , Stress, Physiological/genetics , Vicia faba/genetics , 5-Methylcytosine/metabolism , Acetylation/drug effects , Cell Cycle/drug effects , Cell Cycle/genetics , Chromatin/metabolism , DNA Replication/drug effects , DNA, Plant/metabolism , Epigenesis, Genetic/drug effects , Histones/metabolism , Lysine/metabolism , Meristem/drug effects , Methylation/drug effects , Phosphorylation/drug effects , Stress, Physiological/drug effects , Vicia faba/cytology , Vicia faba/drug effects
20.
Int J Mol Sci ; 22(24)2021 Dec 15.
Article En | MEDLINE | ID: mdl-34948245

Heme oxygenase-1 (HO-1) is one of the most powerful cytoprotective proteins known. The goal of this study was to explore the effects of HO-1 in c-kit-positive cardiac cells (CPCs). LinNEG/c-kitPOS CPCs were isolated and expanded from wild-type (WT), HO-1 transgenic (TG), or HO-1 knockout (KO) mouse hearts. Compared with WT CPCs, cell proliferation was significantly increased in HO-1TG CPCs and decreased in HO-1KO CPCs. HO-1TG CPCs also exhibited a marked increase in new DNA synthesis during the S-phase of cell division, not only under normoxia (21% O2) but after severe hypoxia (1% O2 for 16 h). These properties of HO-1TG CPCs were associated with nuclear translocation (and thus activation) of Nrf2, a key transcription factor that regulates antioxidant genes, and increased protein expression of Ec-SOD, the only extracellular antioxidant enzyme. These data demonstrate that HO-1 upregulates Ec-SOD in CPCs and suggest that this occurs via activation of Nrf2, which thus is potentially involved in the crosstalk between two antioxidants, HO-1 in cytoplasm and Ec-SOD in extracellular matrix. Overexpression of HO-1 in CPCs may improve the survival and reparative ability of CPCs after transplantation and thus may have potential clinical application to increase efficacy of cell therapy.


Heme Oxygenase-1/metabolism , Heme Oxygenase-1/physiology , Myocytes, Cardiac/metabolism , Animals , Antioxidants/pharmacology , Cell Proliferation , DNA Replication/drug effects , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Heart , Heme Oxygenase (Decyclizing)/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/physiology , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-kit/metabolism , S Phase , Superoxide Dismutase/metabolism
...