Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Cell ; 151(7): 1474-87, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23260137

ABSTRACT

DNA Ligase IV is responsible for sealing of double-strand breaks (DSBs) during nonhomologous end-joining (NHEJ). Inhibiting Ligase IV could result in amassing of DSBs, thereby serving as a strategy toward treatment of cancer. Here, we identify a molecule, SCR7 that inhibits joining of DSBs in cell-free repair system. SCR7 blocks Ligase IV-mediated joining by interfering with its DNA binding but not that of T4 DNA Ligase or Ligase I. SCR7 inhibits NHEJ in a Ligase IV-dependent manner within cells, and activates the intrinsic apoptotic pathway. More importantly, SCR7 impedes tumor progression in mouse models and when coadministered with DSB-inducing therapeutic modalities enhances their sensitivity significantly. This inhibitor to target NHEJ offers a strategy toward the treatment of cancer and improvement of existing regimens.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair/drug effects , DNA Ligases/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , Pyrimidines/therapeutic use , Schiff Bases/therapeutic use , Amino Acid Sequence , Animals , Cell Line, Tumor , DNA Ligase ATP , DNA Ligases/chemistry , DNA Ligases/genetics , Disease Models, Animal , Drug Design , Drug Resistance, Neoplasm , Humans , Lymphocytes/drug effects , Lymphoma/drug therapy , Lymphoma/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Radiation Tolerance , Rats , Schiff Bases/chemical synthesis , Schiff Bases/chemistry , Sequence Alignment
2.
EMBO J ; 40(17): e107776, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34232536

ABSTRACT

Host-virus protein-protein interactions play key roles in the life cycle of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We conducted a comprehensive interactome study between the virus and host cells using tandem affinity purification and proximity-labeling strategies and identified 437 human proteins as the high-confidence interacting proteins. Further characterization of these interactions and comparison to other large-scale study of cellular responses to SARS-CoV-2 infection elucidated how distinct SARS-CoV-2 viral proteins participate in its life cycle. With these data mining, we discovered potential drug targets for the treatment of COVID-19. The interactomes of two key SARS-CoV-2-encoded viral proteins, NSP1 and N, were compared with the interactomes of their counterparts in other human coronaviruses. These comparisons not only revealed common host pathways these viruses manipulate for their survival, but also showed divergent protein-protein interactions that may explain differences in disease pathology. This comprehensive interactome of SARS-CoV-2 provides valuable resources for the understanding and treating of this disease.


Subject(s)
COVID-19/genetics , Coronavirus Nucleocapsid Proteins/genetics , SARS-CoV-2/genetics , Viral Nonstructural Proteins/genetics , COVID-19/pathology , COVID-19/virology , Host-Pathogen Interactions/genetics , Humans , Protein Interaction Maps/genetics , SARS-CoV-2/pathogenicity , Virus Replication/genetics
3.
Nucleic Acids Res ; 49(13): 7476-7491, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34197614

ABSTRACT

Poly (ADP-ribose) polymerase inhibitor (PARPi)-based therapies initially reduce tumor burden but eventually lead to acquired resistance in cancer patients with BRCA1 or BRCA2 mutation. To understand the potential PARPi resistance mechanisms, we performed whole-genome CRISPR screens to discover genetic alterations that change the gene essentiality in cells with inducible depletion of BRCA2. We identified that several RNA Polymerase II transcription Mediator complex components, especially Cyclin C (CCNC) as synthetic survival targets upon BRCA2 loss. Total mRNA sequencing demonstrated that loss of CCNC could activate the transforming growth factor (TGF)-beta signaling pathway and extracellular matrix (ECM)-receptor interaction pathway, however the inhibition of these pathways could not reverse cell survival in BRCA2 depleted CCNC-knockout cells, indicating that the activation of these pathways is not required for the resistance. Moreover, we showed that the improved survival is not due to restoration of homologous recombination repair although decreased DNA damage signaling was observed. Interestingly, loss of CCNC could restore replication fork stability in BRCA2 deficient cells, which may contribute to PARPi resistance. Taken together, our data reveal CCNC as a critical genetic determinant upon BRCA2 loss of function, which may help the development of novel therapeutic strategies that overcome PARPi resistance.


Subject(s)
BRCA2 Protein/genetics , Cyclin C/genetics , BRCA2 Protein/metabolism , CRISPR-Cas Systems , Cell Survival , DNA Damage , DNA Replication , Gene Expression Regulation , Gene Knockout Techniques , HEK293 Cells , Humans , Mediator Complex/genetics , Mediator Complex/physiology , Recombinational DNA Repair , Stress, Physiological/genetics
4.
EMBO Rep ; 21(6): e49123, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32307824

ABSTRACT

Replication across oxidative DNA lesions can give rise to mutations that pose a threat to genome integrity. How such lesions, which escape base excision repair, get removed without error during replication remains unknown. Our PCNA-based screen to uncover changes in replisome composition under different replication stress conditions had revealed a previously unknown PCNA-interacting protein, HMCES/C3orf37. Here, we show that HMCES is a critical component of the replication stress response, mainly upon base misincorporation. We further demonstrate that the absence of HMCES imparts resistance to pemetrexed treatment due to error-prone bypass of oxidative damage. Furthermore, based on genetic screening, we show that homologous recombination repair proteins, such as CtIP, BRCA2, BRCA1, and PALB2, are indispensable for the survival of HMCES KO cells. Hence, HMCES, which is the sole member of the SRAP superfamily in higher eukaryotes known so far, acts as a proofreader on replication forks, facilitates resolution of oxidative base damage, and therefore ensures faithful DNA replication.


Subject(s)
DNA Repair , DNA Replication , DNA Damage , DNA Repair/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Oxidative Stress/genetics
5.
Mol Cell Proteomics ; 19(3): 467-477, 2020 03.
Article in English | MEDLINE | ID: mdl-31900314

ABSTRACT

Adenosine monophosphate-activated protein kinase (AMPK) is an obligate heterotrimer that consists of a catalytic subunit (α) and two regulatory subunits (ß and γ). AMPK is a key enzyme in the regulation of cellular energy homeostasis. It has been well studied and is known to function in many cellular pathways. However, the interactome of AMPK has not yet been systematically established, although protein-protein interaction is critically important for protein function and regulation. Here, we used tandem-affinity purification, coupled with mass spectrometry (TAP-MS) analysis, to determine the interactome of AMPK and its functions. We conducted a TAP-MS analysis of all seven AMPK subunits. We identified 138 candidate high-confidence interacting proteins (HCIPs) of AMPK, which allowed us to build an interaction network of AMPK complexes. Five candidate AMPK-binding proteins were experimentally validated, underlining the reliability of our data set. Furthermore, we demonstrated that AMPK acts with a strong AMPK-binding protein, Artemis, in non-homologous end joining. Collectively, our study established the first AMPK interactome and uncovered a new function of AMPK in DNA repair.


Subject(s)
AMP-Activated Protein Kinases/metabolism , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Protein Subunits/metabolism , DNA-Binding Proteins/genetics , Endonucleases/genetics , HEK293 Cells , Humans , Protein Interaction Mapping
6.
Mol Carcinog ; 60(9): 627-643, 2021 09.
Article in English | MEDLINE | ID: mdl-34192388

ABSTRACT

Nonhomologous end joining (NHEJ), one of the major DNA double-strand break repair pathways, plays a significant role in cancer cell proliferation and resistance to radio and chemotherapeutic agents. Previously, we had described a small molecule inhibitor, SCR7, which inhibited NHEJ in a DNA Ligase IV dependent manner. Here, we report that SCR7 potentiates the effect of γ-radiation (IR) that induces DNA breaks as intermediates to eradicate cancer cells. Dose fractionation studies revealed that coadministration of SCR7 and IR (0.5 Gy) in mice Dalton's lymphoma (DLA) model led to a significant reduction in mice tumor cell proliferation, which was equivalent to that observed for 2 Gy dose when both solid and liquid tumor models were used. Besides, co-treatment with SCR7 and 1 Gy of IR further improved the efficacy. Notably, there was no significant change in blood parameters, kidney and liver functions upon combinatorial treatment of SCR7 and IR. Further, the co-treatment of SCR7 and IR resulted in a significant increase in unrepaired DSBs within cancer cells compared to either of the agent alone. Anatomy, histology, and other studies in tumor models confirmed the cumulative effects of both agents in activating apoptotic pathways to induce cytotoxicity by modulating DNA damage response and repair pathways. Thus, we report that SCR7 has the potential to reduce the side effects of radiotherapy by lowering its effective dose ex vivo and in mice tumor models, with implications in cancer therapy.


Subject(s)
DNA End-Joining Repair/drug effects , DNA End-Joining Repair/radiation effects , Pyrimidines/pharmacology , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , Schiff Bases/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Ligase ATP/metabolism , Disease Models, Animal , Humans , Mice , Xenograft Model Antitumor Assays
7.
Nucleic Acids Res ; 47(19): 10181-10201, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31495888

ABSTRACT

Interstrand crosslinks (ICLs) are highly toxic DNA lesions that are repaired via a complex process requiring the coordination of several DNA repair pathways. Defects in ICL repair result in Fanconi anemia, which is characterized by bone marrow failure, developmental abnormalities, and a high incidence of malignancies. SLX4, also known as FANCP, acts as a scaffold protein and coordinates multiple endonucleases that unhook ICLs, resolve homologous recombination intermediates, and perhaps remove unhooked ICLs. In this study, we explored the role of SLX4IP, a constitutive factor in the SLX4 complex, in ICL repair. We found that SLX4IP is a novel regulatory factor; its depletion sensitized cells to treatment with ICL-inducing agents and led to accumulation of cells in the G2/M phase. We further discovered that SLX4IP binds to SLX4 and XPF-ERCC1 simultaneously and that disruption of one interaction also disrupts the other. The binding of SLX4IP to both SLX4 and XPF-ERCC1 not only is vital for maintaining the stability of SLX4IP protein, but also promotes the interaction between SLX4 and XPF-ERCC1, especially after DNA damage. Collectively, these results demonstrate a new regulatory role for SLX4IP in maintaining an efficient SLX4-XPF-ERCC1 complex in ICL repair.


Subject(s)
Carrier Proteins/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Homologous Recombination/genetics , Recombinases/genetics , DNA/chemistry , DNA/genetics , DNA-Binding Proteins/chemistry , HEK293 Cells , Humans , Protein Binding/genetics
8.
EMBO Rep ; 17(1): 94-109, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26658330

ABSTRACT

High expression levels of SLFN11 correlate with the sensitivity of human cancer cells to DNA-damaging agents. However, little is known about the underlying mechanism. Here, we show that SLFN11 interacts directly with RPA1 and is recruited to sites of DNA damage in an RPA1-dependent manner. Furthermore, we establish that SLFN11 inhibits checkpoint maintenance and homologous recombination repair by promoting the destabilization of the RPA-ssDNA complex, thereby sensitizing cancer cell lines expressing high endogenous levels of SLFN11 to DNA-damaging agents. Finally, we demonstrate that the RPA1-binding ability of SLFN11 is required for its function in the DNA damage response. Our findings not only provide novel insight into the molecular mechanisms underlying the drug sensitivity of cancer cell lines expressing SLFN11 at high levels, but also suggest that SLFN11 expression can serve as a biomarker to predict responses to DNA-damaging therapeutic agents.


Subject(s)
Cell Cycle Checkpoints , DNA Damage , Nuclear Proteins/metabolism , Recombinational DNA Repair , Replication Protein A/metabolism , Biomarkers , Cell Line, Tumor , DNA Replication , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Genes, cdc , HeLa Cells , Humans , Nuclear Proteins/genetics , Replication Protein A/genetics
9.
Mol Carcinog ; 56(2): 550-566, 2017 02.
Article in English | MEDLINE | ID: mdl-27312791

ABSTRACT

The terminal step of ligation of single and/or double-strand breaks during physiological processes such as DNA replication, repair and recombination requires participation of DNA ligases in all mammals. DNA Ligase I has been well characterised to play vital roles during these processes. Considering the indispensable role of DNA Ligase I, a therapeutic strategy to impede proliferation of cancer cells is by using specific small molecule inhibitors against it. In the present study, we have designed and chemically synthesised putative DNA Ligase I inhibitors. Based on various biochemical and biophysical screening approaches, we identify two prospective DNA Ligase I inhibitors, SCR17 and SCR21. Both the inhibitors blocked ligation of nicks on DNA in a concentration-dependent manner, when catalysed by cell-free extracts or purified Ligase I. Docking studies in conjunction with biolayer interferometry and gel shift assays revealed that both SCR17 and SCR21 can bind to Ligase I, particularly to the DNA Binding Domain of Ligase I with KD values in nanomolar range. The inhibitors did not show significant affinity towards DNA Ligase III and DNA Ligase IV. Further, addition of Ligase I could restore the joining, when the inhibitors were treated with testicular cell-free extracts. Ex vivo studies using multiple assays showed that even though cell death was limited in the presence of inhibitors in cancer cells, their proliferation was compromised. Hence, we identify two promising DNA Ligase I inhibitors, which can be used in biochemical and cellular assays, and could be further modified and optimised to target cancer cells. © 2016 Wiley Periodicals, Inc.


Subject(s)
DNA Ligase ATP/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Ligase ATP/chemistry , DNA Ligase ATP/metabolism , DNA Replication/drug effects , Drug Design , HEK293 Cells , Humans , Male , Molecular Docking Simulation , Rats , Rats, Wistar
10.
Mol Cell Biochem ; 426(1-2): 149-160, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27882441

ABSTRACT

Pyrazole moiety represents an important category of heterocyclic compound in pharmaceutical and medicinal chemistry. The novel 1-aryl-3, 5-bis (het) aryl pyrazole derivatives were synthesized with complementary regioselectivity. The chemical structures were confirmed by IR, 1H NMR, 13C NMR, and mass spectral analysis. The chemical entities were screened in various cancer cell lines to assess their cell viability activity. Results showed that the compound 3-(1-(4-bromophenyl)-5-phenyl-1H-pyrazol-3-yl) pyridine (5d) possessed maximum cytotoxic effect against breast cancer and leukemic cells. The cytotoxicity was confirmed by live-dead cell assay and cell cycle analysis. Mitochondrial membrane potential, Annexin V-FITC staining, DNA fragmentation, Hoechst staining, and western blot assays revealed the ability of compound 5d to induce cell death by activating apoptosis in cancer cells. Thus, the present study demonstrates that compound 5d could be an attractive chemical entity for the development of small molecule inhibitors for treatment of leukemia and breast cancer.


Subject(s)
Antineoplastic Agents , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Cytotoxins , Leukemia/drug therapy , Pyrazoles , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cell Death/drug effects , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacology , Female , Humans , K562 Cells , Leukemia/metabolism , MCF-7 Cells , Mass Spectrometry , Mice , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacology
11.
Bioorg Med Chem Lett ; 24(19): 4682-4688, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25205189

ABSTRACT

A series of 2,5,6-substituted imidazo[2,1-b][1,3,4]thiadiazole derivatives have been prepared and were tested for antiproliferative activity on cancer cells at the National Cancer Institute. Results showed that molecules with a benzyl group at position 2, exhibited an increase in activity for the introduction of a formyl group at the 5 position. The compound 2-benzyl-5-formyl-6-(4-bromophenyl)imidazo[2,1-b][1,3,4]thiadiazole 22 has been chosen for understanding the mechanism of action by various molecular and cellular biology studies. Results obtained from cell cycle evaluation analysis, analysis of mitochondrial membrane potential and Annexin V-FITC by flow cytometric analysis, ROS production and expression of apoptotic and DNA-repair proteins suggested that compound 22 induced cytotoxicity by activating extrinsic pathway of apoptosis, however, without affecting cell cycle progression.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Thiadiazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Molecular Structure , Structure-Activity Relationship , Thiadiazoles/chemical synthesis , Thiadiazoles/chemistry
12.
J Periodontol ; 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38946115

ABSTRACT

BACKGROUND: Periodontal diseases are associated with dysbiosis in the oral microbial communities. Managing oral biofilms is therefore key for preventing these diseases. Management protocols often include over-the-counter antimicrobial mouth rinses, which lack data on their effects on the oral microbiome's ecology, bacterial composition, metabolic activity, and dysbiosis resilience. This study examined the efficacy of antimicrobial mouth rinses to halt dysbiosis in in vitro oral biofilms under periodontitis-simulating conditions. METHODS: Multispecies oral biofilms were grown on hydroxyapatite discs (HADs) and rinsed daily with one of six mouth rinses. Positive and negative controls were included. After three rinses, biofilms were analyzed with viability quantitative polymerase chain reaction and visualized using scanning electron microscopy. Supernatants of rinsed biofilms were used for metabolic activity analysis. In addition, human oral keratinocytes were exposed to rinsed biofilms to assess their inflammatory response. All outputs were analyzed for correlation using Spearman coefficient. RESULTS: Product-related changes were observed in the rinsed biofilms. Three of the six tested mouth rinses could significantly prevent dysbiosis with ≥30% reduction in pathobiont abundance relative to the control. These biofilms had lower metabolic activity, and the exposed human oral keratinocyte produced less interleukin-8. Interleukin-8 production correlated to both pathobiont quantity and the metabolic activity of the biofilms. CONCLUSION: Some mouth rinses could support biofilm resilience and stop dysbiosis evolution in the biofilm model, with a clear product-related effect. Such mouth rinses can be considered for patients under maintenance/supportive periodontal therapy to prevent/delay disease recurrence. Others are more useful for different periodontal therapy stages.

13.
Pharmaceutics ; 16(3)2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38543271

ABSTRACT

Peri-implantitis is a growing pathological concern for dental implants which aggravates the occurrence of revision surgeries. This increases the burden on both hospitals and the patients themselves. Research is now focused on the development of materials and accompanying implants designed to resist biofilm formation. To enhance this endeavor, a smart method of biofilm inhibition coupled with limiting toxicity to the host cells is crucial. Therefore, this research aims to establish a proof-of-concept for the pH-triggered release of chlorhexidine (CHX), an antiseptic commonly used in mouth rinses, from a titanium (Ti) substrate to inhibit biofilm formation on its surface. To this end, a macroporous Ti matrix is filled with mesoporous silica (together referred to as Ti/SiO2), which acts as a diffusion barrier for CHX from the CHX feed side to the release side. To limit release to acidic conditions, the release side of Ti/SiO2 is coated with crosslinked chitosan (CS), a pH-responsive and antimicrobial natural polymer. Scanning electron microscopy coupled with energy dispersive X-ray spectroscopy (SEM/EDX) and Fourier transform infrared (FTIR) spectroscopy confirmed successful CS film formation and crosslinking on the Ti/SiO2 disks. The presence of the CS coating reduced CHX release by 33% as compared to non-coated Ti/SiO2 disks, thus reducing the antiseptic exposure to the environment in normal conditions. Simultaneous differential scanning calorimetry and thermogravimetric analyzer (SDT) results highlighted the thermal stability of the crosslinked CS films. Quartz crystal microbalance with dissipation monitoring (QCM-D) indicated a clear pH response for crosslinked CS coatings in an acidic medium. This pH response also influenced CHX release through a Ti/SiO2/CS disk where the CHX release was higher than the average trend in the neutral medium. Finally, the antimicrobial study revealed a significant reduction in biofilm formation for the CS-coated samples compared to the control sample using viability quantitative polymerase chain reaction (v-qPCR) measurements, which were also corroborated using SEM imaging. Overall, this study investigates the smart triggered release of pharmaceutical agents aimed at inhibiting biofilm formation, with potential applicability to implant-like structures.

14.
Mol Carcinog ; 52(6): 413-25, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22213363

ABSTRACT

Polycyclic aromatic molecules such as ellipticine intercalate into double-stranded DNA and interfere with physiological functions. In the present study, we evaluate the chemotherapeutic potential of MPTQ on animal models and its mode of action. In order to test the antitumor activity, monohydrochloride of MPTQ was orally administered in mice bearing tumor. Results showed a significant inhibition of tumor growth compared to that of untreated controls. More importantly, mean lifespan of tumor bearing animals treated with MPTQ was significantly higher as compared to that of untreated tumor bearing mice suggesting that the treatment affected viability of cancerous cells, but not of normal cells. Consistent with this, we find that administration of MPTQ to normal mice did not cause any major side effects as observed upon hematological and serum profiling. We also found that MPTQ induces cytotoxicity in cancer cell lines, by activating apoptosis both by intrinsic and extrinsic pathways. Thus, MPTQ could be used as a potential cancer therapeutic agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Intercalating Agents/therapeutic use , Neoplasms/drug therapy , Quinolines/therapeutic use , Thiophenes/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , DNA/genetics , DNA Breaks, Double-Stranded/drug effects , Humans , Intercalating Agents/pharmacology , Leukemia/drug therapy , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , Mice , Mice, Inbred BALB C , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Quinolines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology
15.
Sci Adv ; 9(49): eadi6681, 2023 12 08.
Article in English | MEDLINE | ID: mdl-38055811

ABSTRACT

Type II topoisomerases (TOP2) form transient TOP2 cleavage complexes (TOP2ccs) during their catalytic cycle to relieve topological stress. TOP2ccs are covalently linked TOP2-DNA intermediates that are reversible but can be trapped by TOP2 poisons. Trapped TOP2ccs block transactions on DNA and generate genotoxic stress, which are the mechanisms of action of TOP2 poisons. How cells avoid TOP2cc accumulation remains largely unknown. In this study, we uncovered RAD54 like 2 (RAD54L2) as a key factor that mediates a TOP2-specific DNA damage avoidance pathway. RAD54L2 deficiency conferred unique sensitivity to treatment with TOP2 poisons. RAD54L2 interacted with TOP2A/TOP2B and ZATT/ZNF451 and promoted the turnover of TOP2 from DNA with or without TOP2 poisons. Additionally, inhibition of proteasome activity enhanced the chromatin binding of RAD54L2, which in turn led to the removal of TOP2 from chromatin. In conclusion, we propose that RAD54L2-mediated TOP2 turnover is critically important for the avoidance of potential TOP2-linked DNA damage under physiological conditions and in response to TOP2 poisons.


Subject(s)
Poisons , DNA Topoisomerases, Type II/genetics , DNA Damage , DNA Repair , DNA/chemistry , Chromatin/genetics
16.
FEBS J ; 289(11): 3093-3096, 2022 06.
Article in English | MEDLINE | ID: mdl-35212156

ABSTRACT

In response to double-strand breaks (DSBs) in the DNA, cells undergo transcriptional, translational and post-translational reprogramming to tackle the damage. In this study by Riepe et al., the authors have shown that the global translation inhibition of proteins is concomitant to DNA damage response. Treatment with various DSB-generating agents can cause a major downregulation in the translation of cellular proteins except for the ISR (integrated stress response) proteins. Authors report a specific and significant reduction in the level of a core ribosomal RPS27A protein coupled to kinetics of DSB induction and repair. The study proposes that molecular alterations generated as a by-product of DNA damage may inadvertently impact phenotypic responses of the cells and a cautious approach must be followed when utilizing DSB-based genome editing techniques. Comment on: https://doi.org/10.1111/febs.16321.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA , DNA End-Joining Repair , Gene Editing/methods
17.
Nat Commun ; 13(1): 4240, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35869071

ABSTRACT

Anticancer drugs, such as camptothecin (CPT), trap topoisomerase I (TOP1) on DNA and form TOP1 cleavage complexes (TOP1cc). Alternative repair pathways have been suggested in the repair of TOP1cc. However, how these pathways work with TDP1, a key repair enzyme that specifically hydrolyze the covalent bond between TOP1 catalytic tyrosine and the 3'-end of DNA and contribute to the repair of TOP1cc is poorly understood. Here, using unbiased whole-genome CRISPR screens and generation of co-deficient cells with TDP1 and other genes, we demonstrate that MUS81 is an important factor that mediates the generation of excess double-strand breaks (DSBs) in TDP1 KO cells. APEX1/2 are synthetic lethal with TDP1. However, deficiency of APEX1/2 does not reduce DSB formation in TDP1 KO cells. Together, our data suggest that TOP1cc can be either resolved directly by TDP1 or be converted into DSBs and repaired further by the Homologous Recombination (HR) pathway.


Subject(s)
Antineoplastic Agents , DNA Topoisomerases, Type I , Camptothecin/pharmacology , DNA Damage , DNA Repair , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , Phosphoric Diester Hydrolases/metabolism
18.
FEBS J ; 288(15): 4503-4506, 2021 08.
Article in English | MEDLINE | ID: mdl-33811455

ABSTRACT

Cancer immunotherapy represents a very encouraging mode of treatment for cancer where one's immune system is utilized to eliminate tumor cells. Wayne et al. explore inhibition of DNA damage response (DDR) pathways with small molecule inhibitors as a means to prime cells with immune response. These findings suggest that a one-size-fits-all approach cannot be used when harnessing immune response via DDR inhibitors and genotoxic agents, which are required ultimately for the success of immunotherapy. Comment on: https://doi.org/10.1111/febs.15747.


Subject(s)
Neoplasms , DNA Damage , Humans , Immunity , Immunotherapy , Neoplasms/genetics , Neoplasms/therapy
19.
Cell Rep ; 36(2): 109390, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260911

ABSTRACT

Recombination activating genes (RAGs), consisting of RAG1 and RAG2, are stringently regulated lymphoid-specific genes, which initiate V(D)J recombination in developing lymphocytes. We report the regulation of RAG1 through a microRNA (miRNA), miR-29c, in a B cell stage-specific manner in mice and humans. Various lines of experimentation, including CRISPR-Cas9 genome editing, demonstrate the target specificity and direct interaction of miR-29c to RAG1. Modulation of miR-29c levels leads to change in V(D)J recombination efficiency in pre-B cells. The miR-29c expression is inversely proportional to RAG1 in a B cell developmental stage-specific manner, and miR-29c null mice exhibit a reduction in mature B cells. A negative correlation of miR-29c and RAG1 levels is also observed in leukemia patients, suggesting the potential use of miR-29c as a biomarker and a therapeutic target. Thus, our results reveal the role of miRNA in the regulation of RAG1 and its relevance in cancer.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , MicroRNAs/metabolism , V(D)J Recombination/genetics , 3' Untranslated Regions/genetics , Animals , B-Lymphocytes/cytology , Base Sequence , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Homeodomain Proteins/metabolism , Humans , Luciferases/metabolism , Lymphocytes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , RNA Processing, Post-Transcriptional/genetics
20.
Bioorg Med Chem Lett ; 20(21): 6297-301, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20832305

ABSTRACT

We have designed and synthesized three novel compounds, 5-isopropylidiene derivatives of 3-dimethyl-2-thio-hydantoin (ITH-1), 3-ethyl-2-thio-2,4-oxazolidinedione (ITO-1), and 5-benzilidene-3-ethyl rhodanine (BTR-1), and have tested their chemotherapeutic properties. Our results showed that all three compounds induced cytotoxicity in a time- and concentration-dependent manner on leukemic cell line, CEM. Among the compounds tested, BTR-1 was 5- to 7-fold more potent than ITH-1 and ITO-1 when compared by trypan blue and MTT assays. IC(50) value of BTR-1 was estimated to be <10µM. Both cell cycle analysis and tritiated thymidine assays revealed that BTR-1 affects DNA replication by inducing a block at S phase. BTR-1 treatment led to increased level of ROS production and DNA strand breaks suggesting activation of apoptosis for induction of cell death.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Rhodanine/analogs & derivatives , Rhodanine/pharmacology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cell Survival/drug effects , Coloring Agents , DNA Fragmentation , Dose-Response Relationship, Drug , Humans , L-Lactate Dehydrogenase/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Reactive Oxygen Species , Rhodanine/chemical synthesis , Tetrazolium Salts , Thiazoles , Trypan Blue
SELECTION OF CITATIONS
SEARCH DETAIL