Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 6197, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043663

ABSTRACT

Replication stress compromises genomic integrity. Fork blocking lesions such as those induced by cisplatin and other chemotherapeutic agents arrest replication forks. Repriming downstream of these lesions represents an important mechanism of replication restart, however the single stranded DNA (ssDNA) gaps left behind, unless efficiently filled, can serve as entry point for nucleases. Nascent strand gaps can be repaired by BRCA-mediated homology repair. Alternatively, gaps can also be filled by translesion synthesis (TLS) polymerases. How these events are regulated is still not clear. Here, we show that PARP10, a poorly-characterized mono-ADP-ribosyltransferase, is recruited to nascent strand gaps to promote their repair. PARP10 interacts with the ubiquitin ligase RAD18 and recruits it to these structures, resulting in the ubiquitination of the replication factor PCNA. PCNA ubiquitination, in turn, recruits the TLS polymerase REV1 for gap filling. We show that PARP10 recruitment to gaps and the subsequent REV1-mediated gap filling requires both the catalytic activity of PARP10, and its ability to interact with PCNA. We moreover show that PARP10 is hyperactive in BRCA-deficient cells, and its inactivation potentiates gap accumulations and cytotoxicity in these cells. Our work uncovers PARP10 as a regulator of ssDNA gap filling, which promotes genomic stability in BRCA-deficient cells.


Subject(s)
DNA Repair , DNA Replication , DNA, Single-Stranded , DNA-Binding Proteins , Poly(ADP-ribose) Polymerases , Proliferating Cell Nuclear Antigen , Ubiquitin-Protein Ligases , Ubiquitination , Humans , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/genetics , DNA Damage , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Cell Line, Tumor , HEK293 Cells , Translesion DNA Synthesis , DNA-Directed DNA Polymerase , Proto-Oncogene Proteins
2.
Nat Commun ; 14(1): 6265, 2023 10 07.
Article in English | MEDLINE | ID: mdl-37805499

ABSTRACT

Accumulation of single stranded DNA (ssDNA) gaps in the nascent strand during DNA replication has been associated with cytotoxicity and hypersensitivity to genotoxic stress, particularly upon inactivation of the BRCA tumor suppressor pathway. However, how ssDNA gaps contribute to genotoxicity is not well understood. Here, we describe a multi-step nucleolytic processing of replication stress-induced ssDNA gaps which converts them into cytotoxic double stranded DNA breaks (DSBs). We show that ssDNA gaps are extended bidirectionally by MRE11 in the 3'-5' direction and by EXO1 in the 5'-3' direction, in a process which is suppressed by the BRCA pathway. Subsequently, the parental strand at the ssDNA gap is cleaved by the MRE11 endonuclease generating a double strand break. We also show that exposure to bisphenol A (BPA) and diethylhexyl phthalate (DEHP), which are widespread environmental contaminants due to their use in plastics manufacturing, causes nascent strand ssDNA gaps during replication. These gaps are processed through the same mechanism described above to generate DSBs. Our work sheds light on both the relevance of ssDNA gaps as major determinants of genomic instability, as well as the mechanism through which they are processed to generate genomic instability and cytotoxicity.


Subject(s)
DNA Repair , Exodeoxyribonucleases , Humans , Exodeoxyribonucleases/metabolism , Endonucleases/genetics , Endonucleases/metabolism , DNA/genetics , Genomic Instability , DNA Replication , DNA, Single-Stranded/genetics , DNA Repair Enzymes/metabolism
3.
NAR Cancer ; 5(1): zcad009, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36814782

ABSTRACT

ADP-ribosylation is a post-translational modification involved in a variety of processes including DNA damage repair, transcriptional regulation, and cellular proliferation. Depending on the number of ADP moieties transferred to target proteins, ADP-ribosylation can be classified either as mono-ADP-ribosylation (MARylation) or poly-ADP-ribosylation (PARylation). This post-translational modification is catalyzed by enzymes known as ADP-ribosyltransferases (ARTs), which include the poly (ADP-ribose)-polymerase (PARP) superfamily of proteins. Certain members of the PARP family including PARP1 and PARP2 have been extensively studied and assessed as therapeutic targets. However, the other members of the PARP family of protein are not as well studied but have gained attention in recent years given findings suggesting their roles in an increasing number of cellular processes. Among these other members are PARP10 and PARP14, which have gradually emerged as key players in maintenance of genomic stability and carcinogenesis. PARP10 and PARP14 catalyze the transfer of a single ADP moiety to target proteins. Here, we summarize the current knowledge on MARylation in DNA repair and cancer, focusing on PARP10 and PARP14. We highlight the roles of PARP10 and PARP14 in cancer progression and response to chemotherapeutics and briefly discuss currently known PARP10 and PARP14 inhibitors.

4.
Oncotarget ; 13: 1078-1091, 2022.
Article in English | MEDLINE | ID: mdl-36187556

ABSTRACT

PARP10 is a mono-ADP-ribosyltransferase with multiple cellular functions, including proliferation, apoptosis, metabolism and DNA repair. PARP10 is overexpressed in a significant proportion of tumors, particularly breast and ovarian cancers. Identifying genetic susceptibilities based on PARP10 expression levels is thus potentially relevant for finding new targets for precision oncology. Here, we performed a series of CRISPR genome-wide loss-of-function screens in isogenic control and PARP10-overexpressing or PARP10-knockout cell lines, to identify genetic determinants of PARP10-mediated cellular survival. We found that PARP10-overexpressing cells rely on multiple DNA repair genes for survival, including ATM, the master regulator of the DNA damage checkpoint. Moreover, we show that PARP10 impacts the recruitment of ATM to nascent DNA upon replication stress. Finally, we identify the CDK2-Cyclin E1 complex as essential for proliferation of PARP10-knockout cells. Our work identifies a network of functionally relevant PARP10 synthetic interactions, and reveals a set of factors which can potentially be targeted in personalized cancer therapy.


Subject(s)
Neoplasms , Poly(ADP-ribose) Polymerases , ADP Ribose Transferases/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DNA , Humans , Neoplasms/genetics , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Precision Medicine , Proto-Oncogene Proteins/genetics
5.
Nat Commun ; 13(1): 5323, 2022 09 09.
Article in English | MEDLINE | ID: mdl-36085347

ABSTRACT

The inability to protect stalled replication forks from nucleolytic degradation drives genome instability and underlies chemosensitivity in BRCA-deficient tumors. An emerging hallmark of BRCA-deficiency is the inability to suppress replication-associated single-stranded DNA (ssDNA) gaps. Here, we report that lagging strand ssDNA gaps interfere with the ASF1-CAF-1 nucleosome assembly pathway, and drive fork degradation in BRCA-deficient cells. We show that CAF-1 function at replication forks is lost in BRCA-deficient cells, due to defects in its recycling during replication stress. This CAF-1 recycling defect is caused by lagging strand gaps which preclude PCNA unloading, causing sequestration of PCNA-CAF-1 complexes on chromatin. Importantly, correcting PCNA unloading defects in BRCA-deficient cells restores CAF-1-dependent fork stability. We further show that the activation of a HIRA-dependent compensatory histone deposition pathway restores fork stability to BRCA-deficient cells. We thus define lagging strand gap suppression and nucleosome assembly as critical enablers of BRCA-mediated fork stability.


Subject(s)
Chromatin Assembly and Disassembly , Nucleosomes , Chromatin Assembly Factor-1/genetics , DNA, Single-Stranded/genetics , Proliferating Cell Nuclear Antigen , Recycling
6.
Nat Commun ; 13(1): 5063, 2022 08 27.
Article in English | MEDLINE | ID: mdl-36030235

ABSTRACT

Suppression of nascent DNA degradation has emerged as an essential role of the BRCA pathway in genome protection. In BRCA-deficient cells, the MRE11 nuclease is responsible for both resection of reversed replication forks, and accumulation of single stranded DNA gaps behind forks. Here, we show that the mono-ADP-ribosyltransferase PARP14 is a critical co-factor of MRE11. PARP14 is recruited to nascent DNA upon replication stress in BRCA-deficient cells, and through its catalytic activity, mediates the engagement of MRE11. Loss or inhibition of PARP14 suppresses MRE11-mediated fork degradation and gap accumulation, and promotes genome stability and chemoresistance of BRCA-deficient cells. Moreover, we show that the KU complex binds reversed forks and protects them against EXO1-catalyzed degradation. KU recruits the PARP14-MRE11 complex, which initiates partial resection to release KU and allow long-range resection by EXO1. Our work identifies a multistep process of nascent DNA processing at stalled replication forks in BRCA-deficient cells.


Subject(s)
DNA Replication , Exodeoxyribonucleases , DNA , DNA, Single-Stranded , DNA-Binding Proteins , MRE11 Homologue Protein
7.
Oncogenesis ; 11(1): 33, 2022 Jun 18.
Article in English | MEDLINE | ID: mdl-35717336

ABSTRACT

Maintenance of replication fork stability is essential for genome preservation. Stalled replication forks can be reversed by translocases such as SMARCAL1, and unless protected through the activity of the BRCA pathway, are subsequently subjected to nucleolytic degradation. The ATM and ATR kinases are master regulators of the DNA damage response. ATM activation upon DNA damage is mediated by the acetyltransferase TIP60. Here, we show that the TIP60-ATM pathway promotes replication fork reversal by recruiting SMARCAL1 to stalled forks. This enables fork degradation in BRCA-deficient cells. We also show that this ATM activity is not shared by ATR. Moreover, we performed a series of genome-wide CRISPR knockout genetic screens to identify genetic determinants of the cellular sensitivity to ATM inhibition in wildtype and BRCA2-knockout cells, and validated the top hits from multiple screens. We provide a valuable list of common genes which regulate the response to multiple ATM inhibitors. Importantly, we identify a differential response of wildtype and BRCA2-deficient cells to these inhibitors. In BRCA2-knockout cells, DNA repair genes (including RAD17, MDC1, and USP28) were essential for survival upon ATM inhibitor treatment, which was not the case in wild-type cells. These findings may eventually help guide the way for rational deployment of ATM inhibitors in the clinic.

8.
Sci Rep ; 12(1): 6311, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35428820

ABSTRACT

Transient receptor potential channel melastatin 2 (TRPM2) is highly expressed in cancer and has an essential function in preserving viability through maintenance of mitochondrial function and antioxidant response. Here, the role of TRPM2 in cell survival was examined in neuroblastoma cells with TRPM2 deletion with CRISPR technology. Viability was significantly decreased in TRPM2 knockout after doxorubicin treatment. RNA sequence analysis and RT-qPCR revealed reduced RNAs encoding master transcription regulators FOXM1 and E2F1/2 and downstream cell cycle targets including Cyclin B1, CDK1, PLK1, and CKS1. CHIP analysis demonstrated decreased FOXM1 binding to their promoters. Western blotting confirmed decreased expression, and increased expression of CDK inhibitor p21, a CKS1 target. In cells with TRPM2 deletion, cell cycle progression to S and G2/M phases was reduced after treatment with doxorubicin. RNA sequencing also identified decreased DNA repair proteins in cells with TRPM2 deletion after doxorubicin treatment, and DNA damage was increased. Wild type TRPM2, but not Ca2+-impermeable mutant E960D, restored live cell number and reconstituted expression of E2F1, FOXM1, and cell cycle/DNA repair proteins. FOXM1 expression alone restored viability. TRPM2 is a potential therapeutic target to reduce tumor proliferation and increase doxorubicin sensitivity through modulation of FOXM1, E2F1, and cell cycle/DNA repair proteins.


Subject(s)
E2F1 Transcription Factor , Forkhead Box Protein M1 , Neuroblastoma , TRPM Cation Channels , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Doxorubicin/pharmacology , E2F1 Transcription Factor/metabolism , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Humans , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , TRPM Cation Channels/metabolism
9.
Nucleic Acids Res ; 49(22): 12855-12869, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34871431

ABSTRACT

Understanding chemoresistance mechanisms in BRCA-deficient cells will allow for identification of biomarkers for predicting tumor response to therapy, as well as the design of novel therapeutic approaches targeting this chemoresistance. Here, we show that the protein MED12, a component of the Mediator transcription regulation complex, plays an unexpected role in regulating chemosensitivity in BRCA-deficient cells. We found that loss of MED12 confers resistance to cisplatin and PARP inhibitors in both BRCA1- and BRCA2-deficient cells, which is associated with restoration of both homologous recombination and replication fork stability. Surprisingly, MED12-controlled chemosensitivity does not involve a function of the Mediator complex, but instead reflects a distinct role of MED12 in suppression of the TGFß pathway. Importantly, we show that ectopic activation of the TGFß pathway is enough to overcome the fork protection and DNA repair defects of BRCA-mutant cells, resulting in chemoresistance. Our work identifies the MED12-TGFß module as an important regulator of genomic stability and chemosensitivity in BRCA-deficient cells.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA Replication/genetics , Drug Resistance, Neoplasm/genetics , Mediator Complex/genetics , Transforming Growth Factor beta/genetics , Antineoplastic Agents/pharmacology , BRCA1 Protein/deficiency , BRCA1 Protein/metabolism , BRCA2 Protein/deficiency , BRCA2 Protein/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cisplatin/pharmacology , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Repair , HeLa Cells , Humans , Mediator Complex/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , RNA Interference , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism
10.
Nat Commun ; 11(1): 6118, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257658

ABSTRACT

Inhibitors of poly-ADP-ribose polymerase 1 (PARPi) are highly effective in killing cells deficient in homologous recombination (HR); thus, PARPi have been clinically utilized to successfully treat BRCA2-mutant tumors. However, positive response to PARPi is not universal, even among patients with HR-deficiency. Here, we present the results of genome-wide CRISPR knockout and activation screens which reveal genetic determinants of PARPi response in wildtype or BRCA2-knockout cells. Strikingly, we report that depletion of the ubiquitin ligase HUWE1, or the histone acetyltransferase KAT5, top hits from our screens, robustly reverses the PARPi sensitivity caused by BRCA2-deficiency. We identify distinct mechanisms of resistance, in which HUWE1 loss increases RAD51 levels to partially restore HR, whereas KAT5 depletion rewires double strand break repair by promoting 53BP1 binding to double-strand breaks. Our work provides a comprehensive set of putative biomarkers that advance understanding of PARPi response, and identifies novel pathways of PARPi resistance in BRCA2-deficient cells.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Poly(ADP-ribose) Polymerase Inhibitors/isolation & purification , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/drug effects , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Biomarkers , DNA Damage , DNA Repair , Gene Knockout Techniques , HeLa Cells , Homologous Recombination/drug effects , Humans , Lysine Acetyltransferase 5/metabolism , Mad2 Proteins/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
PLoS Genet ; 16(11): e1009176, 2020 11.
Article in English | MEDLINE | ID: mdl-33137164

ABSTRACT

The ataxia telangiectasia and Rad3-related (ATR) protein kinase is a key regulator of the cellular response to DNA damage. Due to increased amount of replication stress, cancer cells heavily rely on ATR to complete DNA replication and cell cycle progression. Thus, ATR inhibition is an emerging target in cancer therapy, with multiple ATR inhibitors currently undergoing clinical trials. Here, we describe dual genome-wide CRISPR knockout and CRISPR activation screens employed to comprehensively identify genes that regulate the cellular resistance to ATR inhibitors. Specifically, we investigated two different ATR inhibitors, namely VE822 and AZD6738, in both HeLa and MCF10A cells. We identified and validated multiple genes that alter the resistance to ATR inhibitors. Importantly, we show that the mechanisms of resistance employed by these genes are varied, and include restoring DNA replication fork progression, and prevention of ATR inhibitor-induced apoptosis. In particular, we describe a role for MED12-mediated inhibition of the TGFß signaling pathway in regulating replication fork stability and cellular survival upon ATR inhibition. Our dual genome-wide screen findings pave the way for personalized medicine by identifying potential biomarkers for ATR inhibitor resistance.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Biomarkers, Tumor/genetics , Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers, Tumor/metabolism , CRISPR-Cas Systems/genetics , DNA Replication/drug effects , DNA Replication/genetics , Drug Screening Assays, Antitumor , Gene Knockdown Techniques , HeLa Cells , Humans , Indoles , Mediator Complex/genetics , Mediator Complex/metabolism , Morpholines , Neoplasms/genetics , Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfonamides , Sulfoxides/pharmacology , Sulfoxides/therapeutic use , Transforming Growth Factor beta/metabolism
12.
Nucleic Acids Res ; 48(13): 7252-7264, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32542389

ABSTRACT

The DNA damage response is essential to maintain genomic stability, suppress replication stress, and protect against carcinogenesis. The ATR-CHK1 pathway is an essential component of this response, which regulates cell cycle progression in the face of replication stress. PARP14 is an ADP-ribosyltransferase with multiple roles in transcription, signaling, and DNA repair. To understand the biological functions of PARP14, we catalogued the genetic components that impact cellular viability upon loss of PARP14 by performing an unbiased, comprehensive, genome-wide CRISPR knockout genetic screen in PARP14-deficient cells. We uncovered the ATR-CHK1 pathway as essential for viability of PARP14-deficient cells, and identified regulation of DNA replication dynamics as an important mechanistic contributor to the synthetic lethality observed. Our work shows that PARP14 is an important modulator of the response to ATR-CHK1 pathway inhibitors.


Subject(s)
DNA Replication , Poly(ADP-ribose) Polymerases/metabolism , Synthetic Lethal Mutations , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Humans , Poly(ADP-ribose) Polymerases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...