Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 169(6): 979-981, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28575676

ABSTRACT

Humans with inherited heterozygous BRCA2 mutations have an increased risk of developing cancer; however, what triggers carcinogenesis in these individuals is unclear. Tan et al. find that environmental and metabolic aldehydes pose a threat to these individuals by promoting degradation of wild-type BRCA2 protein, thereby predisposing them to genomic instability and perhaps to cancer.


Subject(s)
Aldehydes , Mutation , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Genes, BRCA1 , Genomic Instability , Germ-Line Mutation , Heterozygote , Humans
2.
Cell ; 168(5): 856-866.e12, 2017 Feb 23.
Article in English | MEDLINE | ID: mdl-28215707

ABSTRACT

HSP90 acts as a protein-folding buffer that shapes the manifestations of genetic variation in model organisms. Whether HSP90 influences the consequences of mutations in humans, potentially modifying the clinical course of genetic diseases, remains unknown. By mining data for >1,500 disease-causing mutants, we found a strong correlation between reduced phenotypic severity and a dominant (HSP90 ≥ HSP70) increase in mutant engagement by HSP90. Examining the cancer predisposition syndrome Fanconi anemia in depth revealed that mutant FANCA proteins engaged predominantly by HSP70 had severely compromised function. In contrast, the function of less severe mutants was preserved by a dominant increase in HSP90 binding. Reducing HSP90's buffering capacity with inhibitors or febrile temperatures destabilized HSP90-buffered mutants, exacerbating FA-related chemosensitivities. Strikingly, a compensatory FANCA somatic mutation from an "experiment of nature" in monozygotic twins both prevented anemia and reduced HSP90 binding. These findings provide one plausible mechanism for the variable expressivity and environmental sensitivity of genetic diseases.


Subject(s)
Fanconi Anemia/genetics , Fanconi Anemia/pathology , HSP90 Heat-Shock Proteins/genetics , Protein Folding , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/chemistry , Fanconi Anemia Complementation Group A Protein/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Mutation, Missense , Protein Interaction Domains and Motifs , Stress, Physiological , Twins, Monozygotic
3.
Mol Cell ; 84(14): 2618-2633.e10, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39025073

ABSTRACT

The twenty-three Fanconi anemia (FA) proteins cooperate in the FA/BRCA pathway to repair DNA interstrand cross-links (ICLs). The cell division cycle and apoptosis regulator 1 (CCAR1) protein is also a regulator of ICL repair, though its possible function in the FA/BRCA pathway remains unknown. Here, we demonstrate that CCAR1 plays a unique upstream role in the FA/BRCA pathway and is required for FANCA protein expression in human cells. Interestingly, CCAR1 co-immunoprecipitates with FANCA pre-mRNA and is required for FANCA mRNA processing. Loss of CCAR1 results in retention of a poison exon in the FANCA transcript, thereby leading to reduced FANCA protein expression. A unique domain of CCAR1, the EF hand domain, is required for interaction with the U2AF heterodimer of the spliceosome and for excision of the poison exon. Taken together, CCAR1 is a splicing modulator required for normal splicing of the FANCA mRNA and other mRNAs involved in various cellular pathways.


Subject(s)
Apoptosis Regulatory Proteins , Cell Cycle Proteins , Fanconi Anemia Complementation Group A Protein , Fanconi Anemia , RNA Splicing , Splicing Factor U2AF , Humans , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , DNA Repair , Endodeoxyribonucleases , Exons , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , HEK293 Cells , HeLa Cells , Protein Binding , RNA Precursors/metabolism , RNA Precursors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Spliceosomes/metabolism , Spliceosomes/genetics , Splicing Factor U2AF/metabolism , Splicing Factor U2AF/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism
4.
Mol Cell ; 83(20): 3590-3592, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37863027

ABSTRACT

In this issue of Molecular Cell, Brunner et al.1 reveal that eliminating FANCD2 from stalled forks via FBXL12-mediated degradation enables cells to tolerate oncogene-induced replication stress, making FBXL12 a promising target for cancer treatment.


Subject(s)
DNA Replication , DNA-Binding Proteins , DNA-Binding Proteins/metabolism
5.
Nature ; 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232161

ABSTRACT

CDK1 has been known to be the sole cyclin-dependent kinase (CDK) partner of cyclin B1 to drive mitotic progression1. Here we demonstrate that CDK5 is active during mitosis and is necessary for maintaining mitotic fidelity. CDK5 is an atypical CDK owing to its high expression in post-mitotic neurons and activation by non-cyclin proteins p35 and p392. Here, using independent chemical genetic approaches, we specifically abrogated CDK5 activity during mitosis, and observed mitotic defects, nuclear atypia and substantial alterations in the mitotic phosphoproteome. Notably, cyclin B1 is a mitotic co-factor of CDK5. Computational modelling, comparison with experimentally derived structures of CDK-cyclin complexes and validation with mutational analysis indicate that CDK5-cyclin B1 can form a functional complex. Disruption of the CDK5-cyclin B1 complex phenocopies CDK5 abrogation in mitosis. Together, our results demonstrate that cyclin B1 partners with both CDK5 and CDK1, and CDK5-cyclin B1 functions as a canonical CDK-cyclin complex to ensure mitotic fidelity.

6.
Mol Cell ; 81(21): 4440-4456.e7, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34597596

ABSTRACT

Protection of stalled replication forks is critical to genomic stability. Using genetic and proteomic analyses, we discovered the Protexin complex containing the ssDNA binding protein SCAI and the DNA polymerase REV3. Protexin is required specifically for protecting forks stalled by nucleotide depletion, fork barriers, fragile sites, and DNA inter-strand crosslinks (ICLs), where it promotes homologous recombination and repair. Protexin loss leads to ssDNA accumulation and profound genomic instability in response to ICLs. Protexin interacts with RNA POL2, and both oppose EXO1's resection of DNA on forks remodeled by the FANCM translocase activity. This pathway acts independently of BRCA/RAD51-mediated fork stabilization, and cells with BRCA2 mutations were dependent on SCAI for survival. These data suggest that Protexin and its associated factors establish a new fork protection pathway that counteracts fork resection in part through a REV3 polymerase-dependent resynthesis mechanism of excised DNA, particularly at ICL stalled forks.


Subject(s)
BRCA2 Protein/metabolism , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/chemistry , DNA-Directed DNA Polymerase/chemistry , Exodeoxyribonucleases/metabolism , Transcription Factors/chemistry , Animals , CRISPR-Cas Systems , Cell Line, Tumor , DNA Repair , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , HeLa Cells , Humans , Mevalonic Acid , Mice , Multiprotein Complexes , Mutation , Protein Binding , Protein Conformation , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Interfering/metabolism , Recombination, Genetic
7.
Nat Rev Mol Cell Biol ; 17(6): 337-49, 2016 06.
Article in English | MEDLINE | ID: mdl-27145721

ABSTRACT

The Fanconi anaemia pathway repairs DNA interstrand crosslinks (ICLs) in the genome. Our understanding of this complex pathway is still evolving, as new components continue to be identified and new biochemical systems are used to elucidate the molecular steps of repair. The Fanconi anaemia pathway uses components of other known DNA repair processes to achieve proper repair of ICLs. Moreover, Fanconi anaemia proteins have functions in genome maintenance beyond their canonical roles of repairing ICLs. Such functions include the stabilization of replication forks and the regulation of cytokinesis. Thus, Fanconi anaemia proteins are emerging as master regulators of genomic integrity that coordinate several repair processes. Here, we summarize our current understanding of the functions of the Fanconi anaemia pathway in ICL repair, together with an overview of its connections with other repair pathways and its emerging roles in genome maintenance.


Subject(s)
DNA Repair , Fanconi Anemia Complementation Group Proteins/physiology , Fanconi Anemia/genetics , Animals , DNA Damage , DNA Replication , Humans
8.
Cell ; 152(6): 1344-54, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23498941

ABSTRACT

DNA double-strand breaks (DSBs) can arise from multiple sources, including exposure to ionizing radiation. The repair of DSBs involves both posttranslational modification of nucleosomes and concentration of DNA-repair proteins at the site of damage. Consequently, nucleosome packing and chromatin architecture surrounding the DSB may limit the ability of the DNA-damage response to access and repair the break. Here, we review early chromatin-based events that promote the formation of open, relaxed chromatin structures at DSBs and that allow the DNA-repair machinery to access the spatially confined region surrounding the DSB, thereby facilitating mammalian DSB repair.


Subject(s)
Chromatin Assembly and Disassembly , DNA Breaks, Double-Stranded , DNA Repair , Animals , Genomic Instability , Histones/metabolism , Humans , Neoplasms/genetics
9.
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
10.
Mol Cell ; 80(3): 410-422.e6, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33108758

ABSTRACT

While effective anti-cancer drugs targeting the CHK1 kinase are advancing in the clinic, drug resistance is rapidly emerging. Here, we demonstrate that CRISPR-mediated knockout of the little-known gene FAM122A/PABIR1 confers cellular resistance to CHK1 inhibitors (CHK1is) and cross-resistance to ATR inhibitors. Knockout of FAM122A results in activation of PP2A-B55α, a phosphatase that dephosphorylates the WEE1 protein and rescues WEE1 from ubiquitin-mediated degradation. The resulting increase in WEE1 protein expression reduces replication stress, activates the G2/M checkpoint, and confers cellular resistance to CHK1is. Interestingly, in tumor cells with oncogene-driven replication stress, CHK1 can directly phosphorylate FAM122A, leading to activation of the PP2A-B55α phosphatase and increased WEE1 expression. A combination of a CHK1i plus a WEE1 inhibitor can overcome CHK1i resistance of these tumor cells, thereby enhancing anti-cancer activity. The FAM122A expression level in a tumor cell can serve as a useful biomarker for predicting CHK1i sensitivity or resistance.


Subject(s)
Checkpoint Kinase 1/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Pyrazines/pharmacology , Pyrazoles/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/metabolism , DNA Damage/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Intracellular Signaling Peptides and Proteins/physiology , Nuclear Proteins/metabolism , Phosphoproteins/physiology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/genetics , Pyrazines/metabolism , Pyrazoles/metabolism , Signal Transduction/drug effects
11.
Mol Cell ; 73(5): 885-899.e6, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30686591

ABSTRACT

BRCA1 or BRCA2 inactivation drives breast and ovarian cancer but also creates vulnerability to poly(ADP-ribose) polymerase (PARP) inhibitors. To search for additional targets whose inhibition is synthetically lethal in BRCA2-deficient backgrounds, we screened two pairs of BRCA2 isogenic cell lines with DNA-repair-focused small hairpin RNA (shRNA) and CRISPR (clustered regularly interspaced short palindromic repeats)-based libraries. We found that BRCA2-deficient cells are selectively dependent on multiple pathways including base excision repair, ATR signaling, and splicing. We identified APEX2 and FEN1 as synthetic lethal genes with both BRCA1 and BRCA2 loss of function. BRCA2-deficient cells require the apurinic endonuclease activity and the PCNA-binding domain of Ape2 (APEX2), but not Ape1 (APEX1). Furthermore, BRCA2-deficient cells require the 5' flap endonuclease but not the 5'-3' exonuclease activity of Fen1, and chemically inhibiting Fen1 selectively targets BRCA-deficient cells. Finally, we developed a microhomology-mediated end-joining (MMEJ) reporter and showed that Fen1 participates in MMEJ, underscoring the importance of MMEJ as a collateral repair pathway in the context of homologous recombination (HR) deficiency.


Subject(s)
BRCA2 Protein/genetics , CRISPR-Cas Systems , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Flap Endonucleases/genetics , Genes, Lethal , Neoplasms/genetics , RNA Interference , Synthetic Lethal Mutations , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Cell Death , Cell Line, Tumor , DNA Damage , DNA End-Joining Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Endonucleases , Flap Endonucleases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Multifunctional Enzymes , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/pathology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Protein Interaction Domains and Motifs , RNA, Small Interfering/genetics
12.
Mol Cell ; 72(6): 925-941.e4, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30576655

ABSTRACT

BRCA1-deficient tumor cells have defects in homologous-recombination repair and replication fork stability, resulting in PARP inhibitor sensitivity. Here, we demonstrate that a deubiquitinase, USP1, is upregulated in tumors with mutations in BRCA1. Knockdown or inhibition of USP1 resulted in replication fork destabilization and decreased viability of BRCA1-deficient cells, revealing a synthetic lethal relationship. USP1 binds to and is stimulated by fork DNA. A truncated form of USP1, lacking its DNA-binding region, was not stimulated by DNA and failed to localize and protect replication forks. Persistence of monoubiquitinated PCNA at the replication fork was the mechanism of cell death in the absence of USP1. Taken together, USP1 exhibits DNA-mediated activation at the replication fork, protects the fork, and promotes survival in BRCA1-deficient cells. Inhibition of USP1 may be a useful treatment for a subset of PARP-inhibitor-resistant BRCA1-deficient tumors with acquired replication fork stabilization.


Subject(s)
BRCA1 Protein/deficiency , Breast Neoplasms/enzymology , DNA Replication , DNA, Neoplasm/biosynthesis , Ubiquitin-Specific Proteases/metabolism , Uterine Cervical Neoplasms/enzymology , Animals , BRCA1 Protein/genetics , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Cell Survival , DNA, Neoplasm/genetics , Drug Resistance , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Mice, Nude , Mutation , Nucleic Acid Denaturation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
Mol Cell ; 72(3): 568-582.e6, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30344097

ABSTRACT

Protecting stalled DNA replication forks from degradation by promiscuous nucleases is essential to prevent genomic instability, a major driving force of tumorigenesis. Several proteins commonly associated with the repair of DNA double-strand breaks (DSBs) by homologous recombination (HR) have been implicated in the stabilization of stalled forks. Human CtIP, in conjunction with the MRE11 nuclease complex, plays an important role in HR by promoting DSB resection. Here, we report an unanticipated function for CtIP in protecting reversed forks from degradation. Unlike BRCA proteins, which defend nascent DNA strands from nucleolytic attack by MRE11, we find that CtIP protects perturbed forks from erroneous over-resection by DNA2. Finally, we uncover functionally synergistic effects between CtIP and BRCA1 in mitigating replication-stress-induced genomic instability. Collectively, our findings reveal a DSB-resection- and MRE11-independent role for CtIP in preserving fork integrity that contributes to the survival of BRCA1-deficient cells.


Subject(s)
Carrier Proteins/metabolism , Carrier Proteins/physiology , DNA Replication/physiology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , BRCA1 Protein , BRCA2 Protein , Cell Line , DNA Breaks, Double-Stranded , DNA Helicases/physiology , DNA Repair , DNA-Binding Proteins , Deoxyribonucleases , Endodeoxyribonucleases , Genomic Instability/physiology , Homologous Recombination/genetics , Humans , MRE11 Homologue Protein/metabolism , Protein Binding
14.
Nucleic Acids Res ; 51(18): 9920-9937, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37665033

ABSTRACT

Polymerase theta (Polθ) acts in DNA replication and repair, and its inhibition is synthetic lethal in BRCA1 and BRCA2-deficient tumor cells. Novobiocin (NVB) is a first-in-class inhibitor of the Polθ ATPase activity, and it is currently being tested in clinical trials as an anti-cancer drug. Here, we investigated the molecular mechanism of NVB-mediated Polθ inhibition. Using hydrogen deuterium exchange-mass spectrometry (HX-MS), biophysical, biochemical, computational and cellular assays, we found NVB is a non-competitive inhibitor of ATP hydrolysis. NVB sugar group deletion resulted in decreased potency and reduced HX-MS interactions, supporting a specific NVB binding orientation. Collective results revealed that NVB binds to an allosteric site to block DNA binding, both in vitro and in cells. Comparisons of The Cancer Genome Atlas (TCGA) tumors and matched controls implied that POLQ upregulation in tumors stems from its role in replication stress responses to increased cell proliferation: this can now be tested in fifteen tumor types by NVB blocking ssDNA-stimulation of ATPase activity, required for Polθ function at replication forks and DNA damage sites. Structural and functional insights provided in this study suggest a path for developing NVB derivatives with improved potency for Polθ inhibition by targeting ssDNA binding with entropically constrained small molecules.


Subject(s)
Adenosine Triphosphatases , DNA Polymerase theta , Neoplasms , Novobiocin , Humans , Adenosine Triphosphatases/metabolism , DNA Replication , DNA, Single-Stranded , DNA-Directed DNA Polymerase/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Novobiocin/pharmacology
15.
Proc Natl Acad Sci U S A ; 119(49): e2214935119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36442094

ABSTRACT

The 53BP1-RIF1 pathway restricts the resection of DNA double-strand breaks (DSBs) and promotes blunt end-ligation by non-homologous end joining (NHEJ) repair. The Shieldin complex is a downstream effector of the 53BP1-RIF1 pathway. Here, we identify a component of this pathway, CCAR2/DBC1, which is also required for restriction of DNA end-resection. CCAR2 co-immunoprecipitates with the Shieldin complex, and knockout of CCAR2 in a BRCA1-deficient cell line results in elevated DSB end-resection, RAD51 loading, and PARP inhibitor (PARPi) resistance. Knockout of CCAR2 is epistatic with knockout of other Shieldin proteins. The S1-like RNA-binding domain of CCAR2 is required for its interaction with the Shieldin complex and for suppression of DSB end-resection. CCAR2 functions downstream of the Shieldin complex, and CCAR2 knockout cells have delayed resolution of Shieldin complex foci. Forkhead-associated (FHA)-dependent targeting of CCAR2 to DSB sites re-sensitized BRCA1-/-SHLD2-/- cells to PARPi. Taken together, CCAR2 is a functional component of the 53BP1-RIF1 pathway, promotes the refill of resected DSBs, and suppresses homologous recombination.


Subject(s)
DNA Breaks, Double-Stranded , Poly(ADP-ribose) Polymerase Inhibitors , DNA End-Joining Repair , Homologous Recombination , DNA
16.
PLoS Genet ; 18(11): e1010459, 2022 11.
Article in English | MEDLINE | ID: mdl-36441774

ABSTRACT

Overexpression of the TGFß pathway impairs the proliferation of the hematopoietic stem and progenitor cells (HSPCs) pool in Fanconi anemia (FA). TGFß promotes the expression of NHEJ genes, known to function in a low-fidelity DNA repair pathway, and pharmacological inhibition of TGFß signaling rescues FA HSPCs. Here, we demonstrate that genetic disruption of Smad3, a transducer of the canonical TGFß pathway, modifies the phenotype of FA mouse models deficient for Fancd2. We observed that the TGFß and NHEJ pathway genes are overexpressed during the embryogenesis of Fancd2-/- mice and that the Fancd2-/-Smad3-/- double knockout (DKO) mice undergo high levels of embryonic lethality due to loss of the TGFß-NHEJ axis. Fancd2-deficient embryos acquire extensive genomic instability during gestation which is not reversed by Smad3 inactivation. Strikingly, the few DKO survivors have activated the non-canonical TGFß-ERK pathway, ensuring expression of NHEJ genes during embryogenesis and improved survival. Activation of the TGFß-NHEJ axis was critical for the survival of the few Fancd2-/-Smad3-/- DKO newborn mice but had detrimental consequences for these surviving mice, such as enhanced genomic instability and ineffective hematopoiesis.


Subject(s)
Fanconi Anemia , Mice , Animals , Fanconi Anemia/genetics , Transforming Growth Factor beta/genetics
17.
Cell ; 139(7): 1222-4, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20064367

ABSTRACT

Left unrepaired, DNA interstrand crosslinks represent impassable hurdles for DNA replication, and their removal is a complicated stepwise process involving a variety of enzymes. In a recent paper in Science, Knipscheer et al. (2009) demonstrate that the Fanconi Anemia protein FANCD2 promotes multiple steps of the crosslink repair process.


Subject(s)
DNA Repair , Fanconi Anemia Complementation Group D2 Protein/metabolism , Animals , Humans
18.
Nature ; 563(7732): 522-526, 2018 11.
Article in English | MEDLINE | ID: mdl-30464262

ABSTRACT

Limited DNA end resection is the key to impaired homologous recombination in BRCA1-mutant cancer cells. Here, using a loss-of-function CRISPR screen, we identify DYNLL1 as an inhibitor of DNA end resection. The loss of DYNLL1 enables DNA end resection and restores homologous recombination in BRCA1-mutant cells, thereby inducing resistance to platinum drugs and inhibitors of poly(ADP-ribose) polymerase. Low BRCA1 expression correlates with increased chromosomal aberrations in primary ovarian carcinomas, and the junction sequences of somatic structural variants indicate diminished homologous recombination. Concurrent decreases in DYNLL1 expression in carcinomas with low BRCA1 expression reduced genomic alterations and increased homology at lesions. In cells, DYNLL1 limits nucleolytic degradation of DNA ends by associating with the DNA end-resection machinery (MRN complex, BLM helicase and DNA2 endonuclease). In vitro, DYNLL1 binds directly to MRE11 to limit its end-resection activity. Therefore, we infer that DYNLL1 is an important anti-resection factor that influences genomic stability and responses to DNA-damaging chemotherapy.


Subject(s)
BRCA1 Protein/deficiency , Cytoplasmic Dyneins/metabolism , DNA/metabolism , Genes, BRCA1 , MRE11 Homologue Protein/metabolism , Recombinational DNA Repair , BRCA1 Protein/genetics , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Chromosome Aberrations , DNA Damage/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Gene Editing , Genomic Instability/drug effects , Homologous Recombination/drug effects , Humans , Mutation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Platinum/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Binding , Recombinational DNA Repair/drug effects , Transcription Factors/metabolism
19.
Mol Cell ; 63(2): 249-260, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27373336

ABSTRACT

Ubiquitin-specific proteases (USPs) constitute the largest family of deubiquitinating enzymes, whose catalytic competency is often modulated by their binding partners through unknown mechanisms. Here we report on a series of crystallographic and biochemical analyses of an evolutionarily conserved deubiquitinase, USP12, which is activated by two ß-propeller proteins, UAF1 and WDR20. Our structures reveal that UAF1 and WDR20 interact with USP12 at two distinct sites far from its catalytic center. Without increasing the substrate affinity of USP12, the two ß-propeller proteins potentiate the enzyme through different allosteric mechanisms. UAF1 docks at the distal end of the USP12 Fingers domain and induces a cascade of structural changes that reach a critical ubiquitin-contacting loop adjacent to the catalytic cleft. By contrast, WDR20 anchors at the base of this loop and remotely modulates the catalytic center of the enzyme. Our results provide a mechanistic example for allosteric activation of USPs by their regulatory partners.


Subject(s)
Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Allosteric Regulation , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Coumarins/metabolism , HEK293 Cells , Humans , Hydrolysis , Kinetics , Models, Molecular , Multiprotein Complexes , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Binding , Protein Conformation , Structure-Activity Relationship , Substrate Specificity , Transfection , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/genetics , Ubiquitination , Ubiquitins/metabolism
20.
Cancer Treat Res ; 186: 285-298, 2023.
Article in English | MEDLINE | ID: mdl-37978141

ABSTRACT

Polymerase theta (POLθ) is the critical multi-domain enzyme in microhomology-mediated end-joining DNA double-stranded break repair. POLθ is expressed at low levels in normal tissue but is often overexpressed in cancers, especially in DNA repair deficient cancers, such as homologous-recombination cancers, rendering them exquisitely sensitive to POLθ inhibition secondary to synthetic lethality. Development of POLθ inhibitors is an active area of investigation with inhibitors of the N-terminal helicase domain or the C-terminal polymerase domain currently in clinical trial. Here, we review POLθ-mediated microhomology-mediated end-joining, the development of POLθ inhibitors, and the potential clinical uses of POLθ inhibitors.


Subject(s)
DNA-Directed DNA Polymerase , Neoplasms , Humans , DNA-Directed DNA Polymerase/genetics , DNA Breaks, Double-Stranded , Neoplasms/drug therapy , Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL