Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 896
Filter
Add more filters

Publication year range
1.
Cell ; 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38971151

ABSTRACT

Homologous recombination deficiency (HRD) is prevalent in cancer, sensitizing tumor cells to poly (ADP-ribose) polymerase (PARP) inhibition. However, the impact of HRD and related therapies on the tumor microenvironment (TME) remains elusive. Our study generates single-cell gene expression and T cell receptor profiles, along with validatory multimodal datasets from >100 high-grade serous ovarian cancer (HGSOC) samples, primarily from a phase II clinical trial (NCT04507841). Neoadjuvant monotherapy with the PARP inhibitor (PARPi) niraparib achieves impressive 62.5% and 73.6% response rates per RECIST v.1.1 and GCIG CA125, respectively. We identify effector regulatory T cells (eTregs) as key responders to HRD and neoadjuvant therapies, co-occurring with other tumor-reactive T cells, particularly terminally exhausted CD8+ T cells (Tex). TME-wide interferon signaling correlates with cancer cells upregulating MHC class II and co-inhibitory ligands, potentially driving Treg and Tex fates. Depleting eTregs in HRD mouse models, with or without PARP inhibition, significantly suppresses tumor growth without observable toxicities, underscoring the potential of eTreg-focused therapeutics for HGSOC and other HRD-related tumors.

2.
Cell ; 170(6): 1079-1095.e20, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28823558

ABSTRACT

Loss-of-function mutations in TET2 occur frequently in patients with clonal hematopoiesis, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML) and are associated with a DNA hypermethylation phenotype. To determine the role of TET2 deficiency in leukemia stem cell maintenance, we generated a reversible transgenic RNAi mouse to model restoration of endogenous Tet2 expression. Tet2 restoration reverses aberrant hematopoietic stem and progenitor cell (HSPC) self-renewal in vitro and in vivo. Treatment with vitamin C, a co-factor of Fe2+ and α-KG-dependent dioxygenases, mimics TET2 restoration by enhancing 5-hydroxymethylcytosine formation in Tet2-deficient mouse HSPCs and suppresses human leukemic colony formation and leukemia progression of primary human leukemia PDXs. Vitamin C also drives DNA hypomethylation and expression of a TET2-dependent gene signature in human leukemia cell lines. Furthermore, TET-mediated DNA oxidation induced by vitamin C treatment in leukemia cells enhances their sensitivity to PARP inhibition and could provide a safe and effective combination strategy to selectively target TET deficiency in cancer. PAPERCLIP.


Subject(s)
Ascorbic Acid/pharmacology , DNA-Binding Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Proto-Oncogene Proteins/metabolism , Vitamins/pharmacology , Animals , Ascorbic Acid/administration & dosage , Cell Death , Cell Line, Tumor , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , Gene Knockdown Techniques , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myelodysplastic Syndromes/genetics , Neoplasm Transplantation , Poly (ADP-Ribose) Polymerase-1/genetics , Proto-Oncogene Proteins/genetics , Transcription, Genetic , Transplantation, Heterologous , Vitamins/administration & dosage
3.
Mol Cell ; 83(7): 1043-1060.e10, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36854302

ABSTRACT

Repair of DNA double-strand breaks (DSBs) elicits three-dimensional (3D) chromatin topological changes. A recent finding reveals that 53BP1 assembles into a 3D chromatin topology pattern around DSBs. How this formation of a higher-order structure is configured and regulated remains enigmatic. Here, we report that SLFN5 is a critical factor for 53BP1 topological arrangement at DSBs. Using super-resolution imaging, we find that SLFN5 binds to 53BP1 chromatin domains to assemble a higher-order microdomain architecture by driving damaged chromatin dynamics at both DSBs and deprotected telomeres. Mechanistically, we propose that 53BP1 topology is shaped by two processes: (1) chromatin mobility driven by the SLFN5-LINC-microtubule axis and (2) the assembly of 53BP1 oligomers mediated by SLFN5. In mammals, SLFN5 deficiency disrupts the DSB repair topology and impairs non-homologous end joining, telomere fusions, class switch recombination, and sensitivity to poly (ADP-ribose) polymerase inhibitor. We establish a molecular mechanism that shapes higher-order chromatin topologies to safeguard genomic stability.


Subject(s)
Chromatin , DNA Repair , Animals , Chromatin/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Mammals/metabolism , Telomere-Binding Proteins/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Cell Cycle Proteins/metabolism
4.
Mol Cell ; 82(13): 2363-2369, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35568026

ABSTRACT

Defects in DNA double-strand break repair are thought to render BRCA1 or BRCA2 (BRCA) mutant tumors selectively sensitive to PARP inhibitors (PARPis). Challenging this framework, BRCA and PARP1 share functions in DNA synthesis on the lagging strand. Thus, BRCA deficiency or "BRCAness" could reflect an inherent lagging strand problem that is vulnerable to drugs such as PARPi that also target the lagging strand, a combination that generates a toxic accumulation of replication gaps.


Subject(s)
BRCA1 Protein , BRCA2 Protein , DNA Breaks, Double-Stranded , DNA Repair , Neoplasms , BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA , DNA Repair/genetics , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
5.
Mol Cell ; 82(20): 3932-3942.e6, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36130596

ABSTRACT

The DNA-PKcs kinase mediates the repair of DNA double-strand breaks via classical non-homologous end joining (NHEJ). DNA-PKcs is also recruited to active replication forks, although a role for DNA-PKcs in the control of fork dynamics is unclear. Here, we identify a crucial role for DNA-PKcs in promoting fork reversal, a process that stabilizes stressed replication forks and protects genome integrity. DNA-PKcs promotes fork reversal and slowing in response to several replication stress-inducing agents in a manner independent of its role in NHEJ. Cells lacking DNA-PKcs activity show increased DNA damage during S-phase and cellular sensitivity to replication stress. Notably, prevention of fork slowing and reversal via DNA-PKcs inhibition efficiently restores chemotherapy sensitivity in BRCA2-deficient mammary tumors with acquired PARPi resistance. Together, our data uncover a new key regulator of fork reversal and show how DNA-PKcs signaling can be manipulated to alter fork dynamics and drug resistance in cancer.


Subject(s)
DNA Breaks, Double-Stranded , Drug Resistance, Neoplasm , Drug Resistance, Neoplasm/genetics , DNA Damage , DNA End-Joining Repair , DNA/genetics , DNA Replication , DNA Repair
6.
Mol Cell ; 82(7): 1297-1312.e8, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35219381

ABSTRACT

Synthetic lethality through combinatorial targeting DNA damage response (DDR) pathways provides exciting anticancer therapeutic benefit. Currently, the long noncoding RNAs (lncRNAs) have been implicated in tumor drug resistance; however, their potential significance in DDR is still largely unknown. Here, we report that a human lncRNA, CTD-2256P15.2, encodes a micropeptide, named PAR-amplifying and CtIP-maintaining micropeptide (PACMP), with a dual function to maintain CtIP abundance and promote poly(ADP-ribosyl)ation. PACMP not only prevents CtIP from ubiquitination through inhibiting the CtIP-KLHL15 association but also directly binds DNA damage-induced poly(ADP-ribose) chains to enhance PARP1-dependent poly(ADP-ribosyl)ation. Targeting PACMP alone inhibits tumor growth by causing a synthetic lethal interaction between CtIP and PARP inhibitions and confers sensitivity to PARP/ATR/CDK4/6 inhibitors, ionizing radiation, epirubicin, and camptothecin. Our findings reveal that a lncRNA-derived micropeptide regulates cancer progression and drug resistance by modulating DDR, whose inhibition could be employed to augment the existing anticancer therapeutic strategies.


Subject(s)
Endodeoxyribonucleases , Neoplasms , Peptides , Poly ADP Ribosylation , RNA, Long Noncoding , DNA Repair , Endodeoxyribonucleases/metabolism , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Peptides/pharmacology , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
7.
Mol Cell ; 82(12): 2315-2334, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35271815

ABSTRACT

ADP-ribosylation (ADPRylation) is a post-translational modification of proteins catalyzed by ADP-ribosyl transferase (ART) enzymes, including nuclear PARPs (e.g., PARP1 and PARP2). Historically, studies of ADPRylation and PARPs have focused on DNA damage responses in cancers, but more recent studies elucidate diverse roles in a broader array of biological processes. Here, we summarize the expanding array of molecular mechanisms underlying the biological functions of nuclear PARPs with a focus on PARP1, the founding member of the family. This includes roles in DNA repair, chromatin regulation, gene expression, ribosome biogenesis, and RNA biology. We also present new concepts in PARP1-dependent regulation, including PAR-dependent post-translational modifications, "ADPR spray," and PAR-mediated biomolecular condensate formation. Moreover, we review advances in the therapeutic mechanisms of PARP inhibitors (PARPi) as well as the progress on the mechanisms of PARPi resistance. Collectively, the recent progress in the field has yielded new insights into the expanding universe of PARP1-mediated molecular and therapeutic mechanisms in a variety of biological processes.


Subject(s)
ADP-Ribosylation , DNA Repair , Chromatin/genetics , DNA Damage , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Processing, Post-Translational , RNA/metabolism
8.
Mol Cell ; 81(12): 2640-2655.e8, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34019811

ABSTRACT

ARH3/ADPRHL2 and PARG are the primary enzymes reversing ADP-ribosylation in vertebrates, yet their functions in vivo remain unclear. ARH3 is the only hydrolase able to remove serine-linked mono(ADP-ribose) (MAR) but is much less efficient than PARG against poly(ADP-ribose) (PAR) chains in vitro. Here, by using ARH3-deficient cells, we demonstrate that endogenous MARylation persists on chromatin throughout the cell cycle, including mitosis, and is surprisingly well tolerated. Conversely, persistent PARylation is highly toxic and has distinct physiological effects, in particular on active transcription histone marks such as H3K9ac and H3K27ac. Furthermore, we reveal a synthetic lethal interaction between ARH3 and PARG and identify loss of ARH3 as a mechanism of PARP inhibitor resistance, both of which can be exploited in cancer therapy. Finally, we extend our findings to neurodegeneration, suggesting that patients with inherited ARH3 deficiency suffer from stress-induced pathogenic increase in PARylation that can be mitigated by PARP inhibition.


Subject(s)
Glycoside Hydrolases/metabolism , Poly ADP Ribosylation/physiology , ADP-Ribosylation , Adenosine Diphosphate Ribose/metabolism , Cell Line, Tumor , Chromatin , DNA , DNA Damage , Fibroblasts/metabolism , Glycoside Hydrolases/genetics , Glycoside Hydrolases/physiology , HEK293 Cells , HeLa Cells , Humans , Poly Adenosine Diphosphate Ribose/metabolism , Primary Cell Culture
9.
Mol Cell ; 81(2): 340-354.e5, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33450210

ABSTRACT

In addition to its role as an electron transporter, mitochondrial nicotinamide adenine dinucleotide (NAD+) is an important co-factor for enzymatic reactions, including ADP-ribosylation. Although mitochondria harbor the most intra-cellular NAD+, mitochondrial ADP-ribosylation remains poorly understood. Here we provide evidence for mitochondrial ADP-ribosylation, which was identified using various methodologies including immunofluorescence, western blot, and mass spectrometry. We show that mitochondrial ADP-ribosylation reversibly increases in response to respiratory chain inhibition. Conversely, H2O2-induced oxidative stress reciprocally induces nuclear and reduces mitochondrial ADP-ribosylation. Elevated mitochondrial ADP-ribosylation, in turn, dampens H2O2-triggered nuclear ADP-ribosylation and increases MMS-induced ARTD1 chromatin retention. Interestingly, co-treatment of cells with the mitochondrial uncoupler FCCP decreases PARP inhibitor efficacy. Together, our results suggest that mitochondrial ADP-ribosylation is a dynamic cellular process that impacts nuclear ADP-ribosylation and provide evidence for a NAD+-mediated mitochondrial-nuclear crosstalk.


Subject(s)
ADP-Ribosylation , Cell Nucleus/enzymology , Mitochondria/enzymology , NAD/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , ADP-Ribosylation/drug effects , Animals , Antimycin A/analogs & derivatives , Antimycin A/pharmacology , Cell Line , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Chromatin/chemistry , Chromatin/metabolism , Electron Transport/drug effects , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Methacrylates/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/enzymology , Oligomycins/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/enzymology , Poly (ADP-Ribose) Polymerase-1/genetics , Rotenone/pharmacology , Thiazoles/pharmacology
10.
Mol Cell ; 81(15): 3128-3144.e7, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34216544

ABSTRACT

Mutations in BRCA1 or BRCA2 (BRCA) is synthetic lethal with poly(ADP-ribose) polymerase inhibitors (PARPi). Lethality is thought to derive from DNA double-stranded breaks (DSBs) necessitating BRCA function in homologous recombination (HR) and/or fork protection (FP). Here, we report instead that toxicity derives from replication gaps. BRCA1- or FANCJ-deficient cells, with common repair defects but distinct PARPi responses, reveal gaps as a distinguishing factor. We further uncouple HR, FP, and fork speed from PARPi response. Instead, gaps characterize BRCA-deficient cells, are diminished upon resistance, restored upon resensitization, and, when exposed, augment PARPi toxicity. Unchallenged BRCA1-deficient cells have elevated poly(ADP-ribose) and chromatin-associated PARP1, but aberrantly low XRCC1 consistent with defects in backup Okazaki fragment processing (OFP). 53BP1 loss resuscitates OFP by restoring XRCC1-LIG3 that suppresses the sensitivity of BRCA1-deficient cells to drugs targeting OFP or generating gaps. We highlight gaps as a determinant of PARPi toxicity changing the paradigm for synthetic lethal interactions.


Subject(s)
BRCA1 Protein/genetics , DNA Replication/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Animals , Cell Line , Cisplatin/pharmacology , DNA/genetics , DNA/metabolism , DNA, Single-Stranded/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Homologous Recombination/drug effects , Humans , Mice, Inbred NOD , RNA Helicases/genetics , Rad51 Recombinase/genetics , Replication Protein A/genetics , Tumor Suppressor p53-Binding Protein 1/genetics
11.
Mol Cell ; 81(22): 4692-4708.e9, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34555355

ABSTRACT

Inhibitors of poly(ADP-ribose) (PAR) polymerase (PARPi) have entered the clinic for the treatment of homologous recombination (HR)-deficient cancers. Despite the success of this approach, preclinical and clinical research with PARPi has revealed multiple resistance mechanisms, highlighting the need for identification of novel functional biomarkers and combination treatment strategies. Functional genetic screens performed in cells and organoids that acquired resistance to PARPi by loss of 53BP1 identified loss of LIG3 as an enhancer of PARPi toxicity in BRCA1-deficient cells. Enhancement of PARPi toxicity by LIG3 depletion is dependent on BRCA1 deficiency but independent of the loss of 53BP1 pathway. Mechanistically, we show that LIG3 loss promotes formation of MRE11-mediated post-replicative ssDNA gaps in BRCA1-deficient and BRCA1/53BP1 double-deficient cells exposed to PARPi, leading to an accumulation of chromosomal abnormalities. LIG3 depletion also enhances efficacy of PARPi against BRCA1-deficient mammary tumors in mice, suggesting LIG3 as a potential therapeutic target.


Subject(s)
BRCA1 Protein/genetics , DNA Ligase ATP/genetics , DNA, Single-Stranded , MRE11 Homologue Protein/genetics , Ovarian Neoplasms/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly-ADP-Ribose Binding Proteins/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Animals , Biopsy , CRISPR-Cas Systems , Cell Line , Cell Nucleus/metabolism , Cell Proliferation , Chromosome Aberrations , DNA Damage , DNA Ligase ATP/metabolism , Female , Humans , Lentivirus/genetics , Mammary Neoplasms, Animal , Mice , Mutation , Poly-ADP-Ribose Binding Proteins/metabolism , RNA, Small Interfering/metabolism , Transgenes
12.
Genes Dev ; 35(17-18): 1271-1289, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34385259

ABSTRACT

PARP inhibitor (PARPi) is widely used to treat BRCA1/2-deficient tumors, but why PARPi is more effective than other DNA-damaging drugs is unclear. Here, we show that PARPi generates DNA double-strand breaks (DSBs) predominantly in a trans cell cycle manner. During the first S phase after PARPi exposure, PARPi induces single-stranded DNA (ssDNA) gaps behind DNA replication forks. By trapping PARP on DNA, PARPi prevents the completion of gap repair until the next S phase, leading to collisions of replication forks with ssDNA gaps and a surge of DSBs. In the second S phase, BRCA1/2-deficient cells are unable to suppress origin firing through ATR, resulting in continuous DNA synthesis and more DSBs. Furthermore, BRCA1/2-deficient cells cannot recruit RAD51 to repair collapsed forks. Thus, PARPi induces DSBs progressively through trans cell cycle ssDNA gaps, and BRCA1/2-deficient cells fail to slow down and repair DSBs over multiple cell cycles, explaining the unique efficacy of PARPi in BRCA1/2-deficient cells.


Subject(s)
BRCA2 Protein , Poly(ADP-ribose) Polymerase Inhibitors , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Cycle/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA Replication , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
13.
Mol Cell ; 78(6): 1070-1085, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32459988

ABSTRACT

Anti-cancer drugs targeting the DNA damage response (DDR) exploit genetic or functional defects in this pathway through synthetic lethal mechanisms. For example, defects in homologous recombination (HR) repair arise in cancer cells through inherited or acquired mutations in BRCA1, BRCA2, or other genes in the Fanconi anemia/BRCA pathway, and these tumors have been shown to be particularly sensitive to inhibitors of the base excision repair (BER) protein poly (ADP-ribose) polymerase (PARP). Recent work has identified additional genomic and functional assays of DNA repair that provide new predictive and pharmacodynamic biomarkers for these targeted therapies. Here, we examine the development of selective agents targeting DNA repair, including PARP inhibitors; inhibitors of the DNA damage kinases ataxia-telangiectasia and Rad3 related (ATR), CHK1, WEE1, and ataxia-telangiectasia mutated (ATM); and inhibitors of classical non-homologous end joining (cNHEJ) and alternative end joining (Alt EJ). We also review the biomarkers that guide the use of these agents and current clinical trials with these therapies.


Subject(s)
DNA Repair/drug effects , DNA Repair/physiology , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Pharmacological , DNA Damage/drug effects , DNA End-Joining Repair/drug effects , DNA Repair/genetics , Genes, BRCA1/drug effects , Homologous Recombination , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism
14.
Genes Dev ; 34(5-6): 360-394, 2020 03 01.
Article in English | MEDLINE | ID: mdl-32029455

ABSTRACT

Oxidative and replication stress underlie genomic instability of cancer cells. Amplifying genomic instability through radiotherapy and chemotherapy has been a powerful but nonselective means of killing cancer cells. Precision medicine has revolutionized cancer therapy by putting forth the concept of selective targeting of cancer cells. Poly(ADP-ribose) polymerase (PARP) inhibitors represent a successful example of precision medicine as the first drugs targeting DNA damage response to have entered the clinic. PARP inhibitors act through synthetic lethality with mutations in DNA repair genes and were approved for the treatment of BRCA mutated ovarian and breast cancer. PARP inhibitors destabilize replication forks through PARP DNA entrapment and induce cell death through replication stress-induced mitotic catastrophe. Inhibitors of poly(ADP-ribose) glycohydrolase (PARG) exploit and exacerbate replication deficiencies of cancer cells and may complement PARP inhibitors in targeting a broad range of cancer types with different sources of genomic instability. Here I provide an overview of the molecular mechanisms and cellular consequences of PARP and PARG inhibition. I highlight clinical performance of four PARP inhibitors used in cancer therapy (olaparib, rucaparib, niraparib, and talazoparib) and discuss the predictive biomarkers of inhibitor sensitivity, mechanisms of resistance as well as the means of overcoming them through combination therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Glycoside Hydrolases/antagonists & inhibitors , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Genomic Instability , Glycoside Hydrolases/metabolism , Humans , Neoplasms/enzymology , Poly(ADP-ribose) Polymerases/metabolism
15.
Mol Cell ; 75(6): 1270-1285.e14, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31351877

ABSTRACT

PARP inhibitors (PARPi) prevent cancer cell growth by inducing synthetic lethality with DNA repair defects (e.g., in BRCA1/2 mutant cells). We have identified an alternative pathway for PARPi-mediated growth control in BRCA1/2-intact breast cancer cells involving rDNA transcription and ribosome biogenesis. PARP-1 binds to snoRNAs, which stimulate PARP-1 catalytic activity in the nucleolus independent of DNA damage. Activated PARP-1 ADP-ribosylates DDX21, an RNA helicase that localizes to nucleoli and promotes rDNA transcription when ADP-ribosylated. Treatment with PARPi or mutation of the ADP-ribosylation sites reduces DDX21 nucleolar localization, rDNA transcription, ribosome biogenesis, protein translation, and cell growth. The salient features of this pathway are evident in xenografts in mice and human breast cancer patient samples. Elevated levels of PARP-1 and nucleolar DDX21 are associated with cancer-related outcomes. Our studies provide a mechanistic rationale for efficacy of PARPi in cancer cells lacking defects in DNA repair whose growth is inhibited by PARPi.


Subject(s)
Breast Neoplasms/metabolism , DEAD-box RNA Helicases/metabolism , Neoplasm Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA, Neoplasm/metabolism , RNA, Small Nucleolar/metabolism , Ribosomes/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DEAD-box RNA Helicases/genetics , DNA Repair , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , RNA, Neoplasm/genetics , RNA, Small Nucleolar/genetics , Ribosomes/genetics
16.
Proc Natl Acad Sci U S A ; 120(31): e2301972120, 2023 08.
Article in English | MEDLINE | ID: mdl-37487079

ABSTRACT

PARP1 (poly-ADP ribose polymerase 1) is recruited and activated by DNA strand breaks, catalyzing the generation of poly-ADP-ribose (PAR) chains from NAD+. PAR relaxes chromatin and recruits other DNA repair factors, including XRCC1 and DNA Ligase 3, to maintain genomic stability. Here we show that, in contrast to the normal development of Parp1-null mice, heterozygous expression of catalytically inactive Parp1 (E988A, Parp1+/A) acts in a dominant-negative manner to disrupt murine embryogenesis. As such, all the surviving F1 Parp1+/A mice are chimeras with mixed Parp1+/AN (neoR retention) cells that act similarly to Parp1+/-. Pure F2 Parp1+/A embryos were found at Mendelian ratios at the E3.5 blastocyst stage but died before E9.5. Compared to Parp1-/- cells, genotype and expression-validated pure Parp1+/A cells retain significant ADP-ribosylation and PARylation activities but accumulate markedly higher levels of sister chromatid exchange and mitotic bridges. Despite proficiency for homologous recombination and nonhomologous end-joining measured by reporter assays and supported by normal lymphocyte and germ cell development, Parp1+/A cells are hypersensitive to base damages, radiation, and Topoisomerase I and II inhibition. The sensitivity of Parp1+/A cells to base damages and Topo inhibitors exceed Parp1-/- controls. The findings show that the enzymatically inactive PARP1 dominant negatively blocks DNA repair in selective pathways beyond wild-type PARP1 and establishes a crucial physiological difference between PARP1 inactivation vs. deletion. As a result, the expression of enzymatically inactive PARP1 from one allele is sufficient to abrogate murine embryonic development, providing a mechanism for the on-target side effect of PARP inhibitors used for cancer therapy.


Subject(s)
ADP-Ribosylation , Genomic Instability , Female , Pregnancy , Animals , Mice , Causality , Alleles , Genotype
17.
J Virol ; 98(7): e0057224, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38860782

ABSTRACT

Treatment options for Epstein-Barr virus (EBV)-cancers are limited, underscoring the need for new therapeutic approaches. We have previously shown that EBV-transformed cells and cancers lack homologous recombination (HR) repair, a prominent error-free pathway that repairs double-stranded DNA breaks; instead, EBV-transformed cells demonstrate genome-wide scars of the error-prone microhomology-mediated end joining (MMEJ) repair pathway. This suggests that EBV-cancers are vulnerable to synthetic lethal therapeutic approaches that target MMEJ repair. Indeed, we have previously found that targeting PARP, an enzyme that contributes to MMEJ, results in the death of EBV-lymphoma cells. With the emergence of clinical resistance to PARP inhibitors and the recent discovery of inhibitors of Polymerase theta (POLθ), the polymerase essential for MMEJ, we investigated the role of POLθ in EBV-lymphoma cells. We report that EBV-transformed cell lines, EBV-lymphoma cell lines, and EBV-lymphomas in AIDS patients demonstrate greater abundance of POLθ, driven by the EBV protein EBNA1, compared to EBV-uninfected primary lymphocytes and EBV-negative lymphomas from AIDS patients (a group that also abundantly expresses POLθ). We also find POLθ enriched at cellular DNA replication forks and exposure to the POLθ inhibitor Novobiocin impedes replication fork progress, impairs MMEJ-mediated repair of DNA double-stranded breaks, and kills EBV-lymphoma cells. Notably, cell killing is not due to Novobiocin-induced activation of the lytic/replicative phase of EBV. These findings support a role for POLθ not just in DNA repair but also DNA replication and as a therapeutic target in EBV-lymphomas and potentially other EBV-cancers as EBNA1 is expressed in all EBV-cancers.IMPORTANCEEpstein-Barr virus (EBV) contributes to ~2% of the global cancer burden. With a recent estimate of >200,000 deaths a year, identifying molecular vulnerabilities will be key to the management of these frequently aggressive and treatment-resistant cancers. Building on our earlier work demonstrating reliance of EBV-cancers on microhomology-mediated end-joining repair, we now report that EBV lymphomas and transformed B cell lines abundantly express the MMEJ enzyme POLθ that likely protects cellular replication forks and repairs replication-related cellular DNA breaks. Importantly also, we show that a newly identified POLθ inhibitor kills EBV-cancer cells, revealing a novel strategy to block DNA replication and repair of these aggressive cancers.


Subject(s)
DNA Polymerase theta , DNA-Directed DNA Polymerase , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Humans , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Epstein-Barr Virus Infections/virology , Cell Line, Tumor , DNA End-Joining Repair , Lymphoma/virology , Lymphoma/drug therapy , Lymphoma/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Epstein-Barr Virus Nuclear Antigens/genetics , DNA Breaks, Double-Stranded , Synthetic Lethal Mutations , DNA Replication/drug effects
18.
Exp Cell Res ; 438(1): 114036, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38614421

ABSTRACT

Ovarian cancer is the leading cause of gynecologic cancer death. Among the most innovative anti-cancer approaches, the genetic concept of synthetic lethality is that mutations in multiple genes work synergistically to effect cell death. Previous studies found that although vaccinia-related kinase-1 (VRK1) associates with DNA damage repair proteins, its underlying mechanisms remain unclear. Here, we found high VRK1 expression in ovarian tumors, and that VRK1 depletion can significantly promote apoptosis and cell cycle arrest. The effect of VRK1 knockdown on apoptosis was manifested by increased DNA damage, genomic instability, and apoptosis, and also blocked non-homologous end joining (NHEJ) by destabilizing DNA-PK. Further, we verified that VRK1 depletion enhanced sensitivity to a PARP inhibitor (PARPi), olaparib, promoting apoptosis through DNA damage, especially in ovarian cancer cell lines with high VRK1 expression. Proteins implicated in DNA damage responses are suitable targets for the development of new anti-cancer therapeutic strategies, and their combination could represent an alternative form of synthetic lethality. Therefore, normal protective DNA damage responses are impaired by combining olaparib with elimination of VRK1 and could be used to reduce drug dose and its associated toxicity. In summary, VRK1 represents both a potential biomarker for PARPi sensitivity, and a new DDR-associated therapeutic target, in ovarian cancer.


Subject(s)
DNA Damage , DNA-Activated Protein Kinase , Intracellular Signaling Peptides and Proteins , Ovarian Neoplasms , Protein Serine-Threonine Kinases , Female , Humans , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage/drug effects , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genomic Instability/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
19.
Mol Cell ; 65(2): 260-271, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28107648

ABSTRACT

Poly(ADP-ribosyl)ation (PARylation) is a post-translational modification of proteins mediated by PARP family members, such as PARP-1. Although PARylation has been studied extensively, few examples of definitive biological roles for site-specific PARylation have been reported. Here we show that C/EBPß, a key pro-adipogenic transcription factor, is PARylated by PARP-1 on three amino acids in a conserved regulatory domain. PARylation at these sites inhibits C/EBPß's DNA binding and transcriptional activities and attenuates adipogenesis in various genetic and cell-based models. Interestingly, PARP-1 catalytic activity drops precipitously during the first 48 hr of differentiation, corresponding to a release of C/EBPß from PARylation-mediated inhibition. This promotes the binding of C/EBPß at enhancers controlling the expression of adipogenic target genes and continued differentiation. Depletion or chemical inhibition of PARP-1, or mutation of the PARylation sites on C/EBPß, enhances these early adipogenic events. Collectively, our results provide a clear example of how site-specific PARylation drives biological outcomes.


Subject(s)
Adipocytes/enzymology , Adipogenesis , CCAAT-Enhancer-Binding Protein-beta/metabolism , Embryonic Stem Cells/enzymology , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Protein Processing, Post-Translational , Transcription, Genetic , 3T3-L1 Cells , Adipocytes/drug effects , Adipogenesis/drug effects , Animals , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/genetics , DNA/genetics , DNA/metabolism , Embryonic Stem Cells/drug effects , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , NIH 3T3 Cells , Phenotype , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/deficiency , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Binding , Protein Domains , RNA Interference , Signal Transduction , Time Factors , Transcription, Genetic/drug effects , Transcriptional Activation , Transfection
20.
Mol Cell ; 67(3): 374-386.e5, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28735897

ABSTRACT

RAD51 promotes homology-directed repair (HDR), replication fork reversal, and stalled fork protection. Defects in these functions cause genomic instability and tumorigenesis but also generate hypersensitivity to cancer therapeutics. Here we describe the identification of RADX as an RPA-like, single-strand DNA binding protein. RADX is recruited to replication forks, where it prevents fork collapse by regulating RAD51. When RADX is inactivated, excessive RAD51 activity slows replication elongation and causes double-strand breaks. In cancer cells lacking BRCA2, RADX deletion restores fork protection without restoring HDR. Furthermore, RADX inactivation confers chemotherapy and PARP inhibitor resistance to cancer cells with reduced BRCA2/RAD51 pathway function. By antagonizing RAD51 at forks, RADX allows cells to maintain a high capacity for HDR while ensuring that replication functions of RAD51 are properly regulated. Thus, RADX is essential to achieve the proper balance of RAD51 activity to maintain genome stability.


Subject(s)
DNA, Neoplasm/biosynthesis , Drug Resistance, Neoplasm , Genomic Instability , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rad51 Recombinase/metabolism , Replication Origin , A549 Cells , Animals , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , CRISPR-Cas Systems , DNA Breaks, Double-Stranded , DNA Repair , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Models, Molecular , Mutation , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , RNA Interference , Rad51 Recombinase/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL