Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Proc Natl Acad Sci U S A ; 116(3): 816-825, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30591567

ABSTRACT

G quadruplexes (G4s) and R loops are noncanonical DNA structures that can regulate basic nuclear processes and trigger DNA damage, genome instability, and cell killing. By different technical approaches, we here establish that specific G4 ligands stabilize G4s and simultaneously increase R-loop levels within minutes in human cancer cells. Genome-wide mapping of R loops showed that the studied G4 ligands likely cause the spreading of R loops to adjacent regions containing G4 structures, preferentially at 3'-end regions of expressed genes, which are partially ligand-specific. Overexpression of an exogenous human RNaseH1 rescued DNA damage induced by G4 ligands in BRCA2-proficient and BRCA2-silenced cancer cells. Moreover, even if the studied G4 ligands increased noncanonical DNA structures at similar levels in nuclear chromatin, their cellular effects were different in relation to cell-killing activity and stimulation of micronuclei, a hallmark of genome instability. Our findings therefore establish that G4 ligands can induce DNA damage by an R loop-dependent mechanism that can eventually lead to different cellular consequences depending on the chemical nature of the ligands.


Subject(s)
DNA Damage , G-Quadruplexes , Genomic Instability , Neoplasms/genetics , Aminoquinolines , Cell Line, Tumor , Genes, BRCA2 , Humans , Ligands , Picolinic Acids
2.
Int J Mol Sci ; 20(15)2019 Jul 24.
Article in English | MEDLINE | ID: mdl-31344837

ABSTRACT

The Rho GTPase family can be classified into classic and atypical members. Classic members cycle between an inactive Guanosine DiPhosphate -bound state and an active Guanosine TriPhosphate-bound state. Atypical Rho GTPases, such as RND1, are predominantly in an active GTP-bound conformation. The role of classic members in oncogenesis has been the subject of numerous studies, while that of atypical members has been less explored. Besides the roles of RND1 in healthy tissues, recent data suggest that RND1 is involved in oncogenesis and response to cancer therapeutics. Here, we present the current knowledge on RND1 expression, subcellular localization, and functions in healthy tissues. Then, we review data showing that RND1 expression is dysregulated in tumors, the molecular mechanisms involved in this deregulation, and the role of RND1 in oncogenesis. For several aggressive tumors, RND1 presents the features of a tumor suppressor gene. In these tumors, low expression of RND1 is associated with a bad prognosis for the patients. Finally, we highlight that RND1 expression is induced by anticancer agents and modulates their response. Of note, RND1 mRNA levels in tumors could be used as a predictive marker of both patient prognosis and response to anticancer agents.


Subject(s)
Carcinogenesis/genetics , Neoplasms/genetics , rho GTP-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor , Humans , Neoplasms/pathology
3.
Nucleic Acids Res ; 44(3): 1161-78, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26578593

ABSTRACT

Although defective repair of DNA double-strand breaks (DSBs) leads to neurodegenerative diseases, the processes underlying their production and signaling in non-replicating cells are largely unknown. Stabilized topoisomerase I cleavage complexes (Top1cc) by natural compounds or common DNA alterations are transcription-blocking lesions whose repair depends primarily on Top1 proteolysis and excision by tyrosyl-DNA phosphodiesterase-1 (TDP1). We previously reported that stabilized Top1cc produce transcription-dependent DSBs that activate ATM in neurons. Here, we use camptothecin (CPT)-treated serum-starved quiescent cells to induce transcription-blocking Top1cc and show that those DSBs are generated during Top1cc repair from Top1 peptide-linked DNA single-strand breaks generated after Top1 proteolysis and before excision by TDP1. Following DSB induction, ATM activates DNA-PK whose inhibition suppresses H2AX and H2A ubiquitination and the later assembly of activated ATM into nuclear foci. Inhibition of DNA-PK also reduces Top1 ubiquitination and proteolysis as well as resumption of RNA synthesis suggesting that DSB signaling further enhances Top1cc repair. Finally, we show that co-transcriptional DSBs kill quiescent cells. Together, these new findings reveal that DSB production and signaling by transcription-blocking Top1 lesions impact on non-replicating cell fate and provide insights on the molecular pathogenesis of neurodegenerative diseases such as SCAN1 and AT syndromes, which are caused by TDP1 and ATM deficiency, respectively.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA Topoisomerases, Type I/metabolism , DNA-Activated Protein Kinase/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Camptothecin/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Culture Media, Serum-Free/pharmacology , DNA Breaks, Single-Stranded , DNA Topoisomerases, Type I/genetics , DNA-Activated Protein Kinase/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Immunoblotting , Microscopy, Fluorescence , Nuclear Proteins/genetics , RNA Interference , Signal Transduction , Topoisomerase I Inhibitors/pharmacology , Ubiquitination/drug effects
4.
Cell Rep ; 43(5): 114214, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38761375

ABSTRACT

TDP1 removes transcription-blocking topoisomerase I cleavage complexes (TOP1ccs), and its inactivating H493R mutation causes the neurodegenerative syndrome SCAN1. However, the molecular mechanism underlying the SCAN1 phenotype is unclear. Here, we generate human SCAN1 cell models using CRISPR-Cas9 and show that they accumulate TOP1ccs along with changes in gene expression and genomic distribution of R-loops. SCAN1 cells also accumulate transcriptional DNA double-strand breaks (DSBs) specifically in the G1 cell population due to increased DSB formation and lack of repair, both resulting from abortive removal of transcription-blocking TOP1ccs. Deficient TDP1 activity causes increased DSB production, and the presence of mutated TDP1 protein hampers DSB repair by a TDP2-dependent backup pathway. This study provides powerful models to study TDP1 functions under physiological and pathological conditions and unravels that a gain of function of the mutated TDP1 protein, which prevents DSB repair, rather than a loss of TDP1 activity itself, could contribute to SCAN1 pathogenesis.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Mutation , Neurodegenerative Diseases , Phosphoric Diester Hydrolases , Humans , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Mutation/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/genetics , Transcription, Genetic , R-Loop Structures , CRISPR-Cas Systems/genetics
5.
Nat Commun ; 13(1): 2961, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35618715

ABSTRACT

RNase H2 is a specialized enzyme that degrades RNA in RNA/DNA hybrids and deficiency of this enzyme causes a severe neuroinflammatory disease, Aicardi GoutiĆØres syndrome (AGS). However, the molecular mechanism underlying AGS is still unclear. Here, we show that RNase H2 is associated with a subset of genes, in a transcription-dependent manner where it interacts with RNA Polymerase II. RNase H2 depletion impairs transcription leading to accumulation of R-loops, structures that comprise RNA/DNA hybrids and a displaced DNA strand, mainly associated with short and intronless genes. Importantly, accumulated R-loops are processed by XPG and XPF endonucleases which leads to DNA damage and activation of the immune response, features associated with AGS. Consequently, we uncover a key role for RNase H2 in the transcription of human genes by maintaining R-loop homeostasis. Our results provide insight into the mechanistic contribution of R-loops to AGS pathogenesis.


Subject(s)
R-Loop Structures , Ribonucleases , Autoimmune Diseases of the Nervous System , DNA/chemistry , DNA Breaks , Endoribonucleases/metabolism , Humans , Inflammation/genetics , Nervous System Malformations , R-Loop Structures/genetics , RNA/chemistry , Ribonuclease H/metabolism , Ribonuclease, Pancreatic/metabolism , Ribonucleases/metabolism
6.
EMBO Rep ; 10(8): 887-93, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19557000

ABSTRACT

Ataxia telangiectasia mutated (ATM), the deficiency of which causes a severe neurodegenerative disease, is a crucial mediator for the DNA damage response (DDR). As neurons have high rates of transcription that require topoisomerase I (TOP1), we investigated whether TOP1 cleavage complexes (TOP1cc)-which are potent transcription-blocking lesions-also produce transcription-dependent DNA double-strand breaks (DSBs) with ATM activation. We show the induction of DSBs and DDR activation in post-mitotic primary neurons and lymphocytes treated with camptothecin, with the induction of nuclear DDR foci containing activated ATM, gamma-H2AX (phosphorylated histone H2AX), activated CHK2 (checkpoint kinase 2), MDC1 (mediator of DNA damage checkpoint 1) and 53BP1 (p53 binding protein 1). The DSB-ATM-DDR pathway was suppressed by inhibiting transcription and gamma-H2AX signals were reduced by RNase H1 transfection, which removes transcription-mediated R-loops. Thus, we propose that Top1cc produce transcription arrests with R-loop formation and generate DSBs that activate ATM in post-mitotic cells.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Alpha-Amanitin/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins , Camptothecin/pharmacology , Cells, Cultured , Dichlororibofuranosylbenzimidazole/pharmacology , Enzyme Inhibitors/pharmacology , Flow Cytometry , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Neurons/drug effects , Neurons/metabolism , Nuclear Proteins/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Rats , Ribonuclease H/metabolism , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/physiology , Tumor Suppressor p53-Binding Protein 1
7.
Int Rev Cell Mol Biol ; 364: 195-240, 2021.
Article in English | MEDLINE | ID: mdl-34507784

ABSTRACT

Transcription is an essential cellular process but also a major threat to genome integrity. Transcription-associated DNA breaks are particularly detrimental as their defective repair can induce gene mutations and oncogenic chromosomal translocations, which are hallmarks of cancer. The past few years have revealed that transcriptional breaks mainly originate from DNA topological problems generated by the transcribing RNA polymerases. Defective removal of transcription-induced DNA torsional stress impacts on transcription itself and promotes secondary DNA structures, such as R-loops, which can induce DNA breaks and genome instability. Paradoxically, as they relax DNA during transcription, topoisomerase enzymes introduce DNA breaks that can also endanger genome integrity. Stabilization of topoisomerases on chromatin by various anticancer drugs or by DNA alterations, can interfere with transcription machinery and cause permanent DNA breaks and R-loops. Here, we review the role of transcription in mediating DNA breaks, and discuss how deregulation of topoisomerase activity can impact on transcription and DNA break formation, and its connection with cancer.


Subject(s)
DNA Breaks , DNA/chemistry , Neoplasms/genetics , Neoplasms/pathology , Transcription, Genetic , Animals , DNA Topoisomerases, Type I/metabolism , Genomic Instability , Humans , Neoplasms/enzymology
8.
Mol Cell Oncol ; 7(2): 1691905, 2020.
Article in English | MEDLINE | ID: mdl-32158914

ABSTRACT

Accumulation of DNA damage in resting cells is an emerging cause of human disease. We identified a mechanism of DNA double-strand break (DSB) formation in non-replicating cells, which strictly depends on transcription. These transcriptional DSBs arise from the twinned processing of R-loops and topoisomerase I and may underlie neurological disorders and cancers.

9.
Clin Cancer Res ; 14(20): 6449-55, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18927284

ABSTRACT

PURPOSE: Ecteinascidin 743 (Et743; trabectedin, Yondelis) has recently been approved in Europe for the treatment of soft tissue sarcomas and is undergoing clinical trials for other solid tumors. Et743 selectively targets cells proficient for TC-NER, which sets it apart from other DNA alkylating agents. In the present study, we examined the effects of Et743 on RNA Pol II. EXPERIMENTAL DESIGN AND RESULTS: We report that Et743 induces the rapid and massive degradation of transcribing Pol II in various cancer cell lines and normal fibroblasts. Pol II degradation was abrogated by the proteasome inhibitor MG132 and was dependent on TC-NER. Cockayne syndrome (CS) cells and xeroderma pigmentosum (XP) cells (XPD, XPA, XPG, and XPF) were defective in Pol II degradation, whereas XPC cells whose defect is limited to global genome NER in nontranscribing regions were proficient for Pol II degradation. Complementation of the CSB and XPD cells restored Pol II degradation. We also show that cells defective for the VHL complex were defective in Pol II degradation and that complementation of those cells restores Pol II degradation. Moreover, VHL deficiency rendered cells resistant to Et743-induced cell death, a similar effect to that of TC-NER deficiency. CONCLUSION: These results suggest that both TC-NER-induced and VHL-mediated Pol II degradation play a role in cell killing by Et743.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Repair/drug effects , Dioxoles/pharmacology , Neoplasms/enzymology , RNA Polymerase II/metabolism , Tetrahydroisoquinolines/pharmacology , Transcription, Genetic/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Cockayne Syndrome/enzymology , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , Cysteine Proteinase Inhibitors/pharmacology , DNA Damage/drug effects , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Genetic Complementation Test , Humans , Leupeptins/pharmacology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/drug effects , Poly-ADP-Ribose Binding Proteins , RNA Polymerase II/genetics , Sarcoma/enzymology , Sarcoma/genetics , Sarcoma/pathology , Trabectedin , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism , von Hippel-Lindau Disease/enzymology , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology
10.
Cell Rep ; 28(12): 3167-3181.e6, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31533039

ABSTRACT

Although accumulation of DNA damage and genomic instability in resting cells can cause neurodegenerative disorders, our understanding of how transcription produces DNA double-strand breaks (DSBs) isĀ limited. Transcription-blocking topoisomerase I cleavage complexes (TOP1ccs) are frequent events that prime DSB production in non-replicating cells. Here, we report a mechanism of their formation by showing that they arise from two nearby single-strand breaks (SSBs) on opposing DNA strands: one SSB from the removal of transcription-blocking TOP1ccs by the TDP1 pathway and the other from the cleavage of R-loops by endonucleases, including XPF, XPG, and FEN1. Genetic defects in TOP1cc removal (TDP1, PNKP, and XRCC1) or in the resolution of R-loops (SETX) enhance DSB formation and prevent their repair. Such deficiencies cause neurological disorders. Owing to the high frequency of TOP1cc trapping and the widespread distribution of R-loops, these persistent transcriptional DSBs could accumulate over time in neuronal cells, contributing to the neurodegenerative diseases.


Subject(s)
DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA Topoisomerases, Type I/metabolism , R-Loop Structures , Cell Line , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Flap Endonucleases/metabolism , Humans , Nuclear Proteins/metabolism , Transcription Factors/metabolism
11.
Cell Chem Biol ; 26(11): 1544-1558.e6, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31522999

ABSTRACT

The selective downregulation of activated intracellular proteins is a key challenge in cell biology. RHO small GTPases switch between a guanosine diphosphate (GDP)-bound and a guanosine triphosphate (GTP)-bound state that drives downstream signaling. At present, no tool is available to study endogenous RHO-GTPinduced conformational changes in live cells. Here, we established a cell-based screen to selectively degrade RHOB-GTP using F-box-intracellular single-domain antibody fusion. We identified one intracellular antibody (intrabody) that shows selective targeting of endogenous RHOB-GTP mediated by interactions between the CDR3 loop of the domain antibody and the GTP-binding pocket of RHOB. Our results suggest that, while RHOB is highly regulated at the expression level, only the GTP-bound pool, but not its global expression, mediates RHOB functions in genomic instability and in cell invasion. The F-box/intrabody-targeted protein degradation represents a unique approach to knock down the active form of small GTPases or other proteins with multiple cellular activities.


Subject(s)
Single-Domain Antibodies/metabolism , rhoB GTP-Binding Protein/metabolism , Binding Sites , Cell Movement/drug effects , Crystallography, X-Ray , Doxycycline/pharmacology , F-Box Proteins/genetics , F-Box Proteins/metabolism , Gene Expression/drug effects , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Mutagenesis , Protein Structure, Tertiary , RNA Interference , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , rhoB GTP-Binding Protein/antagonists & inhibitors , rhoB GTP-Binding Protein/genetics
12.
Mol Cancer Ther ; 6(12 Pt 1): 3229-38, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18089716

ABSTRACT

Camptothecin (CPT) analogues are powerful anticancer agents but are chemically unstable due to their alpha-hydroxylactone six-membered E-ring structure, which is essential for trapping topoisomerase I (Top1)-DNA cleavage complexes. To stabilize the E-ring, CPT keto analogues with a five-membered E-ring lacking the oxygen of the lactone ring (S38809 and S39625) have been synthesized. S39625 has been selected for advanced preclinical development based on its promising activity in tumor models. Here, we show that both keto analogues are active against purified Top1 and selective against Top1 in yeast and human cancer cells. The keto analogues show improved cytotoxicity toward colon, breast, and prostate cancer cells and leukemia cells compared with CPT. The drug-induced Top1-DNA cleavage complexes induced by the keto analogues show remarkable persistence both with purified Top1 and in cells following 1-h drug treatments. Moreover, we find that S39625 is not a substrate for either the ABCB1 (multidrug resistance-1/P-glycoprotein) or ABCG2 (mitoxantrone resistance/breast cancer resistance protein) drug efflux transporters, which sets S39625 apart from the clinically used CPT analogues topotecan or SN-38 (active metabolite of irinotecan). Finally, we show that nanomolar concentrations of S38809 or S39625 induce intense and persistent histone gamma-H2AX. The chemical stability of the keto analogues and the ability of S39625 to produce high levels of persistent Top1-DNA cleavage complex and its potent antiproliferative activity against human cancer cell lines make S39625 a promising new anticancer drug candidate. Histone gamma-H2AX could be used as a biomarker for the upcoming clinical trials of S39625.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Enzyme Inhibitors/pharmacology , Topoisomerase I Inhibitors , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Base Sequence , Biological Transport , Camptothecin/chemistry , Camptothecin/metabolism , Camptothecin/pharmacology , Cell Line, Tumor , DNA/metabolism , DNA Primers , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Hydrolysis , Molecular Structure
13.
Cell Death Dis ; 9(9): 931, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209297

ABSTRACT

RHO GTPases regulate essential functions such as the organization of the actin cytoskeleton. The classic members cycle between an active GTP-bound and an inactive GDP-bound conformation whereas atypical members are predominantly GTP-bound. Besides their well-established role, the classic RHO GTPases RHOB and RAC1, are rapidly induced and/or activated by genotoxic stress and contribute to the DNA damage response. Here we used camptothecin, a selective topoisomerase I (TOP1) inhibitor that stabilizes TOP1 cleavage complexes (TOP1cc), to search for other potential early DNA damage-inducible RHO GTPase genes. We identified that an atypical RHO GTPase, RND1, is rapidly induced by camptothecin. RND1 induction is closely associated with the presence of TOP1cc induced by camptothecin or by DNA lesions that elevate TOP1cc levels such as UV and hydrogen peroxide. We further demonstrated that camptothecin increases RND1 gene transcription and mRNA stability. Camptothecin also increases poly(ADP-ribose) polymerase 1 (PARP-1) activity, whose inhibition reduces RND1 transcription. In addition, overexpression of RND1 increases PARP-1, suggesting a cross-talk between PARP-1 and RND1. Finally, RND1 protects cells against camptothecin-induced apoptosis, and hence favors cellular resistance to camptothecin. Together, these findings highlight RND1 as an atypical RHO GTPase early induced by TOP1cc, and show that the TOP1cc-PARP-1-RND1 pathway protects cells against apoptosis induced by camptothecin.


Subject(s)
Camptothecin/pharmacology , DNA Topoisomerases, Type I/genetics , DNA/genetics , Drug Resistance, Neoplasm/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Transcription, Genetic/genetics , rho GTP-Binding Proteins/genetics , Animals , Cell Line , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/genetics , HCT116 Cells , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Mice , NIH 3T3 Cells , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Topoisomerase I Inhibitors/pharmacology
14.
DNA Repair (Amst) ; 5(12): 1489-94, 2006 Dec 09.
Article in English | MEDLINE | ID: mdl-16935573

ABSTRACT

Hereditary spinocerebellar ataxia with axonal neuropathy (SCAN1) is caused by an inactivating mutation (H493R) in the enzyme tyrosyl-DNA phosphodiesterase (Tdp1), which removes blocked 3'-termini at DNA strand breaks. Using SCAN1 cells treated with the specific topoisomerase I (Top1) inhibitor camptothecin, we find enhanced levels of Top1 cleavage complexes (Top1cc) and defective reversal of Top1cc in SCAN1 Tdp1-deficient cells, indicating a direct involvement of Tdp1 in the repair of Top1cc. Because the defective removal of Top1cc and the hypersensitivity of SCAN1 cells to camptothecin are not affected by aphidicolin, we propose that Tdp1 is involved in the repair of Top1cc associated with transcription damage in SCAN1 cells.


Subject(s)
Camptothecin/pharmacology , DNA Repair , DNA Topoisomerases, Type I/metabolism , Phosphoric Diester Hydrolases/metabolism , Spinocerebellar Ataxias/metabolism , Transcription, Genetic , Aphidicolin/pharmacology , Cell Line , DNA Replication/drug effects , Humans , Spinocerebellar Ataxias/genetics
15.
Cancer Res ; 65(11): 4844-51, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15930305

ABSTRACT

FdUMP[10], a 10mer of 5-fluoro-2'-deoxyuridine 5'-monophosphate (FdUMP), the thymidylate synthase inhibitory metabolite of 5-fluorouracil (FU), is most closely correlated with the DNA topoisomerase I (Top1) inhibitor camptothecin in the National Cancer Institute COMPARE analysis, but not with FU. FdUMP[10] exhibits more potent antiproliferative activity than FdUMP or 5-fluoro-2'-deoxyuridine (FdU) and is markedly more active than FU. Camptothecin-resistant P388/CPT45 cells lacking Top1 are cross-resistant to FdUMP[10] as well as to FdUMP, FdU, and the thymidylate synthase inhibitor raltitrexed (Tomudex). FdUMP[10] induces DNA single-strand breaks and cellular Top1-DNA complexes. Such complexes are also observed in response to FdUMP, FdU, raltitrexed, and FU. The FdUMP[10]-induced Top1-DNA complexes are not inhibited by the caspase inhibitor z-VAD-fmk and form independently of apoptotic DNA fragmentation, indicating that they do not correspond to apoptotic Top1-DNA complexes. In biochemical assay, Top1 is directly trapped at uracil and FdU misincorporation sites. We propose that FdUMP[10] damages DNA by trapping Top1 at uracil and FdU misincorporation sites resulting from thymidylate synthase inhibition and thymine depletion.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type I/metabolism , DNA, Neoplasm/metabolism , Fluorodeoxyuridylate/analogs & derivatives , Fluorodeoxyuridylate/pharmacology , Animals , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Death/drug effects , DNA Damage , DNA, Neoplasm/drug effects , DNA, Single-Stranded/metabolism , Drug Screening Assays, Antitumor , Fluorouracil/metabolism , Fluorouracil/pharmacology , Leukemia P388/drug therapy , Leukemia P388/enzymology , Mice , Quinazolines/pharmacology , Thiophenes/pharmacology , Thymidine/deficiency , Thymidine/metabolism , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/metabolism , Topoisomerase I Inhibitors
16.
Mol Cancer Ther ; 5(12): 3139-44, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172417

ABSTRACT

Topoisomerase I (Top1) is a ubiquitous enzyme that removes DNA supercoiling generated during transcription and replication. Top1 can be trapped on DNA as cleavage complexes by the anticancer drugs referred to as Top1 inhibitors as well as by alterations of the DNA structure. We reported recently that Top1 cleavage complexes (Top1cc) are trapped during apoptosis induced by arsenic trioxide and staurosporine. In the present study, we generalize the occurrence of apoptotic Top1cc in response to anticancer drugs, which by themselves do not directly interact with Top1: the topoisomerase II inhibitors etoposide, doxorubicin, and amsacrine, and the tubulin inhibitors vinblastine and Taxol. In all cases, the Top1cc form in the early phase of apoptosis and persist throughout the apoptotic process. Their formation is prevented by the caspase inhibitor benzyloxycarbonyl-Val-Ala-DL-Asp(OMe)-fluoromethylketone and the antioxidant N-acetyl-L-cysteine. We propose that the trapping of Top1cc is a general process of programmed cell death, which is caused by alterations of the DNA structure (oxidized bases and strand breaks) induced by caspases and reactive oxygen species.


Subject(s)
Apoptosis/drug effects , DNA Topoisomerases, Type I/metabolism , Enzyme Inhibitors/pharmacology , Topoisomerase II Inhibitors , Tubulin Modulators/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Amsacrine/pharmacology , Apoptosis/physiology , Caspase Inhibitors , Caspases/metabolism , Cell Line, Tumor , DNA Topoisomerases, Type I/biosynthesis , DNA Topoisomerases, Type II/metabolism , Doxorubicin/pharmacology , Etoposide/pharmacology , HL-60 Cells , Humans , Paclitaxel/pharmacology , Reactive Oxygen Species/metabolism , Vinblastine/pharmacology
17.
Cancer Res ; 64(4): 1475-82, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14983893

ABSTRACT

We have shown previously that the camptothecin analogue topotecan (TPT), a topoisomerase I (Top 1) poison, inhibits hypoxia-inducible factor 1 (HIF-1) transcriptional activity and HIF-1alpha protein accumulation in hypoxia-treated U251 human glioma cells. In this article, we demonstrate that TPT does not affect HIF-1alpha protein accumulation but inhibits its translation. In addition, we demonstrate that Top 1 is required for the inhibition of HIF-1alpha protein accumulation by TPT as shown by experiments performed using camptothecin-resistant cell lines with known Top 1 alterations. Experiments performed with aphidicolin indicated that TPT inhibited HIF-1alpha protein accumulation in the absence of DNA replication. DNA-damaging agents, such as ionizing radiation and doxorubicin, did not affect HIF-1alpha protein accumulation. Ongoing transcription was essential for the inhibition of HIF-1alpha protein accumulation by TPT. Our results demonstrate the existence of a novel pathway connecting Top 1-dependent signaling events and the regulation of HIF-1alpha protein expression and function. In addition, our findings dissociate the cytotoxic activity of TPT from the inhibition of the HIF-1 pathway and raise the possibility of novel clinical applications of TPT aimed at targeting HIF-1-dependent responses.


Subject(s)
DNA Topoisomerases, Type I/physiology , Protein Serine-Threonine Kinases , Transcription Factors/antagonists & inhibitors , Cell Line, Tumor , Cysteine Endopeptidases/physiology , DNA Damage , DNA Replication , Dose-Response Relationship, Drug , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Multienzyme Complexes/physiology , Phosphatidylinositol 3-Kinases/physiology , Proteasome Endopeptidase Complex , Protein Biosynthesis , Protein Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , RNA, Messenger/analysis , TOR Serine-Threonine Kinases , Topoisomerase I Inhibitors , Topotecan/pharmacology , Topotecan/therapeutic use , Transcription Factors/genetics , Transcription, Genetic
18.
PLoS One ; 11(1): e0147053, 2016.
Article in English | MEDLINE | ID: mdl-26784695

ABSTRACT

Topoisomerase I-DNA-cleavage complexes (Top1cc) stabilized by camptothecin (CPT) have specific effects at transcriptional levels. We recently reported that Top1cc increase antisense transcript (aRNAs) levels at divergent CpG-island promoters and, transiently, DNA/RNA hybrids (R-loop) in nuclear and mitochondrial genomes of colon cancer HCT116 cells. However, the relationship between R-loops and aRNAs was not established. Here, we show that aRNAs can form R-loops in N-TERA-2 cells under physiological conditions, and that promoter-associated R-loops are somewhat increased and extended in length immediately upon cell exposure to CPT. In contrast, persistent Top1ccs reduce the majority of R-loops suggesting that CPT-accumulated aRNAs are not commonly involved in R-loops. The enhancement of aRNAs by Top1ccs is present both in human colon cancer HCT116 cells and WI38 fibroblasts suggesting a common response of cancer and normal cells. Although Top1ccs lead to DSB and DDR kinases activation, we do not detect a dependence of aRNA accumulation on ATM or DNA-PK activation. However, we showed that the cell response to persistent Top1ccs can involve an impairment of aRNA turnover rather than a higher synthesis rate. Finally, a genome-wide analysis shows that persistent Top1ccs also determine an accumulation of sense transcripts at 5'-end gene regions suggesting an increased occurrence of truncated transcripts. Taken together, the results indicate that Top1 may regulate transcription initiation by modulating RNA polymerase-generated negative supercoils, which can in turn favor R-loop formation at promoters, and that transcript accumulation at TSS is a response to persistent transcriptional stress by Top1 poisoning.


Subject(s)
Camptothecin/pharmacology , DNA Replication , DNA Topoisomerases, Type I/chemistry , Promoter Regions, Genetic/genetics , Topoisomerase I Inhibitors/pharmacology , DNA Topoisomerases, Type I/genetics , HCT116 Cells , Humans , Real-Time Polymerase Chain Reaction , Transcription, Genetic
19.
Oncogene ; 23(16): 2934-49, 2004 Apr 12.
Article in English | MEDLINE | ID: mdl-15077155

ABSTRACT

Intrinsic (innate) and acquired (adaptive) resistance to chemotherapy critically limits the outcome of cancer treatments. For many years, it was assumed that the interaction of a drug with its molecular target would yield a lethal lesion, and that determinants of intrinsic drug resistance should therefore be sought either at the target level (quantitative changes or/and mutations) or upstream of this interaction, in drug metabolism or drug transport mechanisms. It is now apparent that independent of the factors above, cellular responses to a molecular lesion can determine the outcome of therapy. This review will focus on programmed cell death (apoptosis) and on survival pathways (Bcl-2, Apaf-1, AKT, NF-kappaB) involved in multidrug resistance. We will present our molecular interaction mapping conventions to summarize the AKT and IkappaB/NF-kappaB networks. They complement the p53, Chk2 and c-Abl maps published recently. We will also introduce the 'permissive apoptosis-resistance' model for the selection of multidrug-resistant cells.


Subject(s)
Apoptosis , Drug Resistance, Neoplasm , Neoplasms/pathology , Protein Serine-Threonine Kinases , Animals , Apoptotic Protease-Activating Factor 1 , Humans , NF-kappa B/metabolism , Neoplasms/drug therapy , Proteins/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/physiology
20.
Curr Pharm Des ; 11(22): 2855-72, 2005.
Article in English | MEDLINE | ID: mdl-16101442

ABSTRACT

Most anticancer drugs presently used clinically target genomic DNA. The selectivity of these anticancer drugs for tumor tissues is probably due to tumor-specific defects suppressing cell cycle checkpoints and DNA repair, and enhancing apoptotic response in the tumor. We will review the molecular interactions within the ATM-Chk2 pathway implicating the DNA damage sensor kinases (ATM, ATR and DNA-PK), the adaptor BRCT proteins (Nbs1, Brca1, 53BP1, MDC1) and the effector kinases (Chk2, Chk1, Plk3, JNK, p38). The molecular interaction map convention (MIM) will be used for presenting this molecular network (http://discover.nci.nih.gov/mim/). A characteristic of the ATM-Chk2 pathway is its redundancy. First, ATM and Chk2 phosphorylate common substrates including p53, E2F1, BRCA1, and Chk2 itself, which suggests that Chk2 (also known as CHECK2, Cds1 in fission yeast, and Dmchk2 or Dmnk or Loki in the fruit fly) acts as a relay for ATM and/or as a salvage pathway when ATM is inactivated. Secondly, redundancy is apparent for the substrates, which can be phosphorylated/activated at similar residues by Chk2, Chk1, and the polo kinases (Plk's). Functionally, Chk2 can activate both apoptosis (via p53, E2F1 and PML) and cell cycle checkpoint (via Cdc25A and Cdc25C, p53, and BRCA1). We will review the short list of published Chk2 inhibitors. We will also propose a novel paradigm for screening interfacial inhibitors of Chk2. Chk2 inhibitors might be used to enhance the tumor selectivity of DNA targeted agents in p53-deficient tumors, and for the treatment of tumors whose growth depends on enhanced Chk2 activity.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Checkpoint Kinase 2 , Drug Design , Enzyme Activation , Gene Expression Regulation, Enzymologic , Humans , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL