Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27
Filtrer
1.
Nat Struct Mol Biol ; 30(10): 1456-1467, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37696958

RÉSUMÉ

The extent and efficacy of DNA end resection at DNA double-strand breaks (DSB) determine the repair pathway choice. Here we describe how the 53BP1-associated protein DYNLL1 works in tandem with the Shieldin complex to protect DNA ends. DYNLL1 is recruited to DSBs by 53BP1, where it limits end resection by binding and disrupting the MRE11 dimer. The Shieldin complex is recruited to a fraction of 53BP1-positive DSBs hours after DYNLL1, predominantly in G1 cells. Shieldin localization to DSBs depends on MRE11 activity and is regulated by the interaction of DYNLL1 with MRE11. BRCA1-deficient cells rendered resistant to PARP inhibitors by the loss of Shieldin proteins can be resensitized by the constitutive association of DYNLL1 with MRE11. These results define the temporal and functional dynamics of the 53BP1-centric DNA end resection factors in cells.


Sujet(s)
Protéine BRCA1 , Cassures double-brin de l'ADN , Protéine BRCA1/métabolisme , Protéine-1 liant le suppresseur de tumeur p53/métabolisme , ADN/métabolisme , Réparation de l'ADN par jonction d'extrémités , Noyau de la cellule/métabolisme , Réparation de l'ADN
2.
Cancer Cell ; 40(9): 957-972.e10, 2022 09 12.
Article de Anglais | MEDLINE | ID: mdl-35985342

RÉSUMÉ

Diffuse midline glioma (DMG) is a uniformly fatal pediatric cancer driven by oncohistones that do not readily lend themselves to drug development. To identify druggable targets for DMG, we conducted a genome-wide CRISPR screen that reveals a DMG selective dependency on the de novo pathway for pyrimidine biosynthesis. This metabolic vulnerability reflects an elevated rate of uridine/uracil degradation that depletes DMG cells of substrates for the alternate salvage pyrimidine biosynthesis pathway. A clinical stage inhibitor of DHODH (rate-limiting enzyme in the de novo pathway) diminishes uridine-5'-phosphate (UMP) pools, generates DNA damage, and induces apoptosis through suppression of replication forks-an "on-target" effect, as shown by uridine rescue. Matrix-assisted laser desorption/ionization (MALDI) mass spectroscopy imaging demonstrates that this DHODH inhibitor (BAY2402234) accumulates in the brain at therapeutically relevant concentrations, suppresses de novo pyrimidine biosynthesis in vivo, and prolongs survival of mice bearing intracranial DMG xenografts, highlighting BAY2402234 as a promising therapy against DMGs.


Sujet(s)
Gliome , Pyrimidines , Animaux , Gliome/traitement médicamenteux , Gliome/génétique , Humains , Souris , Uridine/métabolisme , Uridine/pharmacologie
3.
Cancer Res ; 81(10): 2774-2787, 2021 05 15.
Article de Anglais | MEDLINE | ID: mdl-33514515

RÉSUMÉ

Homologous recombination (HR)-deficient cancers are sensitive to poly-ADP ribose polymerase inhibitors (PARPi), which have shown clinical efficacy in the treatment of high-grade serous cancers (HGSC). However, the majority of patients will relapse, and acquired PARPi resistance is emerging as a pressing clinical problem. Here we generated seven single-cell clones with acquired PARPi resistance derived from a PARPi-sensitive TP53 -/- and BRCA1 -/- epithelial cell line generated using CRISPR/Cas9. These clones showed diverse resistance mechanisms, and some clones presented with multiple mechanisms of resistance at the same time. Genomic analysis of the clones revealed unique transcriptional and mutational profiles and increased genomic instability in comparison with a PARPi-sensitive cell line. Clonal evolutionary analyses suggested that acquired PARPi resistance arose via clonal selection from an intrinsically unstable and heterogenous cell population in the sensitive cell line, which contained preexisting drug-tolerant cells. Similarly, clonal and spatial heterogeneity in tumor biopsies from a clinical patient with BRCA1-mutant HGSC with acquired PARPi resistance was observed. In an imaging-based drug screening, the clones showed heterogenous responses to targeted therapeutic agents, indicating that not all PARPi-resistant clones can be targeted with just one therapy. Furthermore, PARPi-resistant clones showed mechanism-dependent vulnerabilities to the selected agents, demonstrating that a deeper understanding on the mechanisms of resistance could lead to improved targeting and biomarkers for HGSC with acquired PARPi resistance. SIGNIFICANCE: This study shows that BRCA1-deficient cells can give rise to multiple genomically and functionally heterogenous PARPi-resistant clones, which are associated with various vulnerabilities that can be targeted in a mechanism-specific manner.


Sujet(s)
Protéine BRCA1/physiologie , Évolution clonale , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Tumeurs de l'ovaire/traitement médicamenteux , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Protéine p53 suppresseur de tumeur/physiologie , Animaux , Apoptose , Prolifération cellulaire , Femelle , Instabilité du génome , Recombinaison homologue , Humains , Souris , Souris knockout , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Transcriptome , Cellules cancéreuses en culture
4.
Proc Natl Acad Sci U S A ; 117(43): 26795-26803, 2020 10 27.
Article de Anglais | MEDLINE | ID: mdl-33051298

RÉSUMÉ

The repair of DNA double strand breaks (DSBs) that arise from external mutagenic agents and routine cellular processes is essential for life. DSBs are repaired by two major pathways, homologous recombination (HR) and classical nonhomologous end joining (C-NHEJ). DSB repair pathway choice is largely dictated at the step of 5'-3' DNA end resection, which is promoted during S phase, in part by BRCA1. Opposing end resection is the 53BP1 protein, which recruits the ssDNA-binding REV7-Shieldin complex to favor C-NHEJ repair. We recently identified TRIP13 as a proresection factor that remodels REV7, causing its dissociation from the Shieldin subunit SHLD3. Here, we identify p31comet, a negative regulator of MAD2 and the spindle assembly checkpoint, as an important mediator of the TRIP13-REV7 interaction. p31comet binds to the REV7-Shieldin complex in cells, promotes REV7 inactivation, and causes PARP inhibitor resistance. p31comet also participates in the extraction of REV7 from the chromatin. Furthermore, p31comet can counteract REV7 function in translesion synthesis (TLS) by releasing it from REV3 in the Pol ζ complex. Finally, p31comet, like TRIP13, is overexpressed in many cancers and this correlates with poor prognosis. Thus, we reveal a key player in the regulation of HR and TLS with significant clinical implications.


Sujet(s)
ATPases associated with diverse cellular activities/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines du cycle cellulaire/métabolisme , Protéines Mad2/métabolisme , Protéines nucléaires/métabolisme , Réparation de l'ADN par recombinaison , Lignée cellulaire tumorale , Cellules HEK293 , Humains , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/mortalité
5.
Cell Rep ; 30(7): 2402-2415.e5, 2020 02 18.
Article de Anglais | MEDLINE | ID: mdl-32075772

RÉSUMÉ

Cells deficient in ataxia telangiectasia mutated (ATM) are hypersensitive to ionizing radiation and other anti-cancer agents that induce double-strand DNA breaks. ATM inhibitors may therefore sensitize cancer cells to these agents. Some cancers may also have underlying genetic defects predisposing them to an ATM inhibitor monotherapy response. We have conducted a genome-wide CRISPR screen to identify genetic vulnerabilities that sensitize lung cancer cells to ATM inhibitors. Knockout of genes in the Fanconi anemia (FA)/BRCA pathway results in hypersensitivity to the ATM inhibitor M3541. Knockdown of either an FA gene or of ATM results in reduced double-strand break end resection, enhanced non-homologous end joining (NHEJ) repair, and decreased homologous recombination repair. Knockout of both the FA/BRCA pathway and ATM strongly inhibits end resection and generates toxic levels of NHEJ, thereby elucidating a mechanism of cellular death by synthetic lethality. ATM inhibitors may therefore be useful for the treatment of tumors with a defective FA/BRCA pathway.


Sujet(s)
Ataxie-télangiectasie/génétique , Clustered regularly interspaced short palindromic repeats/génétique , Cassures double-brin de l'ADN , Protéines de liaison à l'ADN/métabolisme , Anémie de Fanconi/génétique , Humains
6.
Cell Rep ; 17(9): 2367-2381, 2016 11 22.
Article de Anglais | MEDLINE | ID: mdl-27880910

RÉSUMÉ

Although poly(ADP-ribose) polymerase (PARP) inhibitors are active in homologous recombination (HR)-deficient cancers, their utility is limited by acquired resistance after restoration of HR. Here, we report that dinaciclib, an inhibitor of cyclin-dependent kinases (CDKs) 1, 2, 5, and 9, additionally has potent activity against CDK12, a transcriptional regulator of HR. In BRCA-mutated triple-negative breast cancer (TNBC) cells and patient-derived xenografts (PDXs), dinaciclib ablates restored HR and reverses PARP inhibitor resistance. Additionally, we show that de novo resistance to PARP inhibition in BRCA1-mutated cell lines and a PDX derived from a PARP-inhibitor-naive BRCA1 carrier is mediated by residual HR and is reversed by CDK12 inhibition. Finally, dinaciclib augments the degree of response in a PARP-inhibitor-sensitive model, converting tumor growth inhibition to durable regression. These results highlight the significance of HR disruption as a therapeutic strategy and support the broad use of combined CDK12 and PARP inhibition in TNBC.


Sujet(s)
Protéine BRCA1/métabolisme , Kinases cyclines-dépendantes/antagonistes et inhibiteurs , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Mutation/génétique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Tumeurs du sein triple-négatives/enzymologie , Tumeurs du sein triple-négatives/anatomopathologie , Séquence d'acides aminés , Animaux , Protéine BRCA1/génétique , Composés hétérocycliques bicycliques/pharmacologie , Lignée cellulaire tumorale , N-oxydes cycliques , Kinases cyclines-dépendantes/composition chimique , Kinases cyclines-dépendantes/métabolisme , Altération de l'ADN/génétique , Réparation de l'ADN/effets des médicaments et des substances chimiques , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Techniques de knock-out de gènes , Recombinaison homologue/effets des médicaments et des substances chimiques , Humains , Indolizine , Souris , Inhibiteurs de protéines kinases/pharmacologie , Composés de pyridinium/pharmacologie , Petit ARN interférent/métabolisme , Transcription génétique/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
7.
J Clin Invest ; 125(4): 1523-32, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25751062

RÉSUMÉ

The Fanconi anemia/BRCA (FA/BRCA) pathway is a DNA repair pathway that is required for excision of DNA interstrand cross-links. The 17 known FA proteins, along with several FA-associated proteins (FAAPs), cooperate in this pathway to detect, unhook, and excise DNA cross-links and to subsequently repair the double-strand breaks generated in the process. In the current study, we identified a patient with FA with a point mutation in FANCA, which encodes a mutant FANCA protein (FANCAI939S). FANCAI939S failed to bind to the FAAP20 subunit of the FA core complex, leading to decreased stability. Loss of FAAP20 binding exposed a SUMOylation site on FANCA at amino acid residue K921, resulting in E2 SUMO-conjugating enzyme UBC9-mediated SUMOylation, RING finger protein 4-mediated (RNF4-mediated) polyubiquitination, and proteasome-mediated degradation of FANCA. Mutation of the SUMOylation site of FANCA rescued the expression of the mutant protein. Wild-type FANCA was also subject to SUMOylation, RNF4-mediated polyubiquitination, and degradation, suggesting that regulated release of FAAP20 from FANCA is a critical step in the normal FA pathway. Consistent with this model, cells lacking RNF4 exhibited interstrand cross-linker hypersensitivity, and the gene encoding RNF4 was epistatic with the other genes encoding members of the FA/BRCA pathway. Together, the results from our study underscore the importance of analyzing unique patient-derived mutations for dissecting complex DNA repair processes.


Sujet(s)
Protéine BRCA1/physiologie , Réparation de l'ADN/physiologie , Protéine du groupe de complémentation A de l'anémie de Fanconi/physiologie , Anémie de Fanconi/génétique , Protéines nucléaires/physiologie , Facteurs de transcription/physiologie , Adulte , Lignée cellulaire tumorale , Réparation de l'ADN/génétique , Protéine du groupe de complémentation A de l'anémie de Fanconi/génétique , Protéines des groupes de complémentation de l'anémie de Fanconi/antagonistes et inhibiteurs , Protéines des groupes de complémentation de l'anémie de Fanconi/génétique , Protéines des groupes de complémentation de l'anémie de Fanconi/physiologie , Femelle , Gène BRCA1 , Humains , Mutation faux-sens , Pedigree , Phénotype , Mutation ponctuelle , Proteasome endopeptidase complex/métabolisme , Liaison aux protéines , Maturation post-traductionnelle des protéines/physiologie , Protéolyse , Petit ARN interférent/pharmacologie , Transduction du signal/physiologie , Sumoylation , Tumeurs du sein triple-négatives/génétique , Ubiquitination/physiologie
8.
Cancer Discov ; 5(2): 135-42, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25472942

RÉSUMÉ

UNLABELLED: Deficiency in BRCA-dependent DNA interstrand crosslink (ICL) repair is intimately connected to breast cancer susceptibility and to the rare developmental syndrome Fanconi anemia. Bona fide Fanconi anemia proteins, BRCA2 (FANCD1), PALB2 (FANCN), and BRIP1 (FANCJ), interact with BRCA1 during ICL repair. However, the lack of detailed phenotypic and cellular characterization of a patient with biallelic BRCA1 mutations has precluded assignment of BRCA1 as a definitive Fanconi anemia susceptibility gene. Here, we report the presence of biallelic BRCA1 mutations in a woman with multiple congenital anomalies consistent with a Fanconi anemia-like disorder and breast cancer at age 23. Patient cells exhibited deficiency in BRCA1 and RAD51 localization to DNA-damage sites, combined with radial chromosome formation and hypersensitivity to ICL-inducing agents. Restoration of these functions was achieved by ectopic introduction of a BRCA1 transgene. These observations provide evidence in support of BRCA1 as a new Fanconi anemia gene (FANCS). SIGNIFICANCE: We establish that biallelic BRCA1 mutations cause a distinct FA-S, which has implications for risk counselling in families where both parents harbor BRCA1 mutations. The genetic basis of hereditary cancer susceptibility syndromes provides diagnostic information, insights into treatment strategies, and more accurate recurrence risk counseling to families.


Sujet(s)
Tumeurs du sein/génétique , Anémie de Fanconi/génétique , Gène BRCA1 , Mutation , Adulte , Allèles , Protéine BRCA1/génétique , Séquence nucléotidique , Protéines des groupes de complémentation de l'anémie de Fanconi/génétique , Femelle , Prédisposition génétique à une maladie , Humains , Jeune adulte
9.
Proc Natl Acad Sci U S A ; 110(42): 17041-6, 2013 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-24085845

RÉSUMÉ

Breast Cancer Type 1 Susceptibility Protein (BRCA1)-deficient cells have compromised DNA repair and are sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors. Despite initial responses, the development of resistance limits clinical efficacy. Mutations in the BRCA C-terminal (BRCT) domain of BRCA1 frequently create protein products unable to fold that are subject to protease-mediated degradation. Here, we show HSP90-mediated stabilization of a BRCT domain mutant BRCA1 protein under PARP inhibitor selection pressure. The stabilized mutant BRCA1 protein interacted with PALB2-BRCA2-RAD51, was essential for RAD51 focus formation, and conferred PARP inhibitor as well as cisplatin resistance. Treatment of resistant cells with the HSP90 inhibitor 17-dimethylaminoethylamino-17-demethoxygeldanamycin reduced mutant BRCA1 protein levels and restored their sensitivity to PARP inhibition. Resistant cells also acquired a TP53BP1 mutation that facilitated DNA end resection in the absence of a BRCA1 protein capable of binding CtIP. Finally, concomitant increased mutant BRCA1 and decreased 53BP1 protein expression occur in clinical samples of BRCA1-mutated recurrent ovarian carcinomas that have developed resistance to platinum. These results provide evidence for a two-event mechanism by which BRCA1-mutant tumors acquire anticancer therapy resistance.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéine BRCA1/métabolisme , Cisplatine/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Mutation , Tumeurs de l'ovaire/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Protéine BRCA2/métabolisme , Benzoquinones/pharmacologie , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Protéine du groupe de complémentation N de l'anémie de Fanconi , Femelle , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/génétique , Protéines du choc thermique HSP90/métabolisme , Humains , Lactames macrocycliques/pharmacologie , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Platine/pharmacologie , Poly(ADP-ribose) polymerases/génétique , Poly(ADP-ribose) polymerases/métabolisme , Structure tertiaire des protéines , Rad51 Recombinase/génétique , Rad51 Recombinase/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme
10.
Mol Cell Biol ; 33(22): 4360-70, 2013 Nov.
Article de Anglais | MEDLINE | ID: mdl-24001775

RÉSUMÉ

The deubiquitinating enzyme heterodimeric complex USP1-UAF1 regulates the Fanconi anemia (FA) DNA repair pathway. Absence of this complex leads to increased cellular levels of ubiquitinated FANCD2 (FANCD2-Ub) and ubiquitinated PCNA (PCNA-Ub). Mice deficient in the catalytic subunit of the complex, USP1, exhibit an FA-like phenotype and have a cellular deficiency in homologous-recombination (HR) repair. Here, we have characterized mice deficient in the UAF1 subunit. Uaf1(+/-) mice were small at birth and exhibited reduced fertility, thus resembling Usp1(-/-) mice. Unexpectedly, homozygous Uaf1(-/-) embryos died at embryonic day 7.5 (E7.5). These mutant embryos were small and developmentally retarded. As expected, Uaf1 deficiency in mice led to increased levels of cellular Fancd2-Ub and Pcna-Ub. Uaf1(+/-) murine embryonic fibroblasts (MEFs) exhibited profound chromosome instability, genotoxin hypersensitivity, and a significant defect in homologous-recombination repair. Moreover, Uaf1(-/-) mouse embryonic stem cells (mESCs) showed chromosome instability, genotoxin hypersensitivity, and impaired Fancd2 focus assembly. Similar to USP1 knockdown, UAF1 knockdown in tumor cells caused suppression of tumor growth in vivo. Taken together, our data demonstrate the important regulatory role of the USP1-UAF1 complex in HR repair through its regulation of the FANCD2-Ub and PCNA-Ub cellular pools.


Sujet(s)
Perte de l'embryon/génétique , Délétion de gène , Recombinaison homologue , Souris/embryologie , Souris/génétique , Protéines nucléaires/génétique , Animaux , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Cellules cultivées , Instabilité des chromosomes , Réparation de l'ADN , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/métabolisme , Protéine du groupe de complémentation D2 de l'anémie de Fanconi/métabolisme , Femelle , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Homozygote , Humains , Infertilité/génétique , Mâle , Souris de lignée C57BL , Mutagènes/pharmacologie , Protéines nucléaires/métabolisme , Antigène nucléaire de prolifération cellulaire/métabolisme , Ubiquitination
11.
Nat Med ; 18(7): 1118-22, 2012 Jul.
Article de Anglais | MEDLINE | ID: mdl-22683780

RÉSUMÉ

Although the treatment of acute myeloid leukemia (AML) has improved substantially in the past three decades, more than half of all patients develop disease that is refractory to intensive chemotherapy. Functional genomics approaches offer a means to discover specific molecules mediating the aberrant growth and survival of cancer cells. Thus, using a loss-of-function RNA interference genomic screen, we identified the aberrant expression of hepatocyte growth factor (HGF) as a crucial element in AML pathogenesis. We found HGF expression leading to autocrine activation of its receptor tyrosine kinase, MET, in nearly half of the AML cell lines and clinical samples we studied. Genetic depletion of HGF or MET potently inhibited the growth and survival of HGF-expressing AML cells. However, leukemic cells treated with the specific MET kinase inhibitor crizotinib developed resistance resulting from compensatory upregulation of HGF expression, leading to the restoration of MET signaling. In cases of AML where MET is coactivated with other tyrosine kinases, such as fibroblast growth factor receptor 1 (FGFR1), concomitant inhibition of FGFR1 and MET blocked this compensatory HGF upregulation, resulting in sustained logarithmic cell killing both in vitro and in xenograft models in vivo. Our results show a widespread dependence of AML cells on autocrine activation of MET, as well as the key role of compensatory upregulation of HGF expression in maintaining leukemogenic signaling by this receptor. We anticipate that these findings will lead to the design of additional strategies to block adaptive cellular responses that drive compensatory ligand expression as an essential component of the targeted inhibition of oncogenic receptors in human cancers.


Sujet(s)
Communication autocrine , Leucémie aigüe myéloïde/enzymologie , Protéines proto-oncogènes c-met/métabolisme , Animaux , Communication autocrine/effets des médicaments et des substances chimiques , Communication autocrine/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Crizotinib , Activation enzymatique/effets des médicaments et des substances chimiques , Femelle , Régulation de l'expression des gènes dans la leucémie/effets des médicaments et des substances chimiques , Facteur de croissance des hépatocytes/génétique , Facteur de croissance des hépatocytes/métabolisme , Humains , Immunohistochimie , Cinétique , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Mesures de luminescence , Mâle , Souris , Souris de lignée C57BL , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/génétique , Pyrazoles/pharmacologie , Pyridines/pharmacologie , Récepteur FGFR1/métabolisme , Induction de rémission
12.
Blood ; 119(23): 5449-57, 2012 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-22371882

RÉSUMÉ

Fanconi anemia (FA) is a recessive syndrome characterized by progressive fatal BM failure and chromosomal instability. FA cells have inactivating mutations in a signaling pathway that is critical for maintaining genomic integrity and protecting cells from the DNA damage caused by cross-linking agents. Transgenic expression of the implicated genes corrects the phenotype of hematopoietic cells, but previous attempts at gene therapy have failed largely because of inadequate numbers of hematopoietic stem cells available for gene correction. Induced pluripotent stem cells (iPSCs) constitute an alternate source of autologous cells that are amenable to ex vivo expansion, genetic correction, and molecular characterization. In the present study, we demonstrate that reprogramming leads to activation of the FA pathway, increased DNA double-strand breaks, and senescence. We also demonstrate that defects in the FA DNA-repair pathway decrease the reprogramming efficiency of murine and human primary cells. FA pathway complementation reduces senescence and restores the reprogramming efficiency of somatic FA cells to normal levels. Disease-specific iPSCs derived in this fashion maintain a normal karyotype and are capable of hematopoietic differentiation. These data define the role of the FA pathway in reprogramming and provide a strategy for future translational applications of patient-specific FA iPSCs.


Sujet(s)
Anémie de Fanconi/génétique , Thérapie génétique/méthodes , Hématopoïèse , Cellules souches pluripotentes induites/cytologie , Animaux , Cellules cultivées , Altération de l'ADN , Anémie de Fanconi/métabolisme , Anémie de Fanconi/thérapie , Protéine du groupe de complémentation A de l'anémie de Fanconi/génétique , Fibroblastes/cytologie , Fibroblastes/métabolisme , Délétion de gène , Humains , Cellules souches pluripotentes induites/métabolisme , Caryotype , Souris , Souris de lignée C57BL , Souris knockout , Transduction du signal
13.
Mol Cancer Res ; 10(3): 369-77, 2012 Mar.
Article de Anglais | MEDLINE | ID: mdl-22219386

RÉSUMÉ

The Fanconi anemia pathway is required for repair of DNA interstrand cross-links (ICL). Fanconi anemia pathway-deficient cells are hypersensitive to DNA ICL-inducing drugs such as cisplatin. Conversely, hyperactivation of the Fanconi anemia pathway is a mechanism that may underlie cellular resistance to DNA ICL agents. Modulating FANCD2 monoubiquitination, a key step in the Fanconi anemia pathway, may be an effective therapeutic approach to conferring cellular sensitivity to ICL agents. Here, we show that inhibition of the Nedd8 conjugation system increases cellular sensitivity to DNA ICL-inducing agents. Mechanistically, the Nedd8 inhibition, either by siRNA-mediated knockdown of Nedd8-conjugating enzymes or treatment with a Nedd8-activating enzyme inhibitor MLN4924, suppressed DNA damage-induced FANCD2 monoubiquitination and CHK1 phosphorylation. Our data indicate that inhibition of the Fanconi anemia pathway is largely responsible for the heightened cellular sensitivity to DNA ICLs upon Nedd8 inhibition. These results suggest that a combination of Nedd8 inhibition with ICL-inducing agents may be an effective strategy for sensitizing a subset of drug-resistant cancer cells.


Sujet(s)
Réactifs réticulants/pharmacologie , Altération de l'ADN , ADN/métabolisme , Ubiquitines/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Checkpoint kinase 1 , Cyclopentanes/pharmacologie , Tests de criblage d'agents antitumoraux , Anémie de Fanconi/métabolisme , Anémie de Fanconi/anatomopathologie , Protéine du groupe de complémentation D2 de l'anémie de Fanconi/métabolisme , Techniques de knock-down de gènes , Humains , Modèles biologiques , Protéine NEDD8 , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteurs de poly(ADP-ribose) polymérases , Poly(ADP-ribose) polymerases/métabolisme , Protein kinases/métabolisme , Pyrimidines/pharmacologie , Ubiquitination/effets des médicaments et des substances chimiques , Ubiquitines/métabolisme
14.
DNA Repair (Amst) ; 10(12): 1203-12, 2011 Dec 10.
Article de Anglais | MEDLINE | ID: mdl-21975120

RÉSUMÉ

The Fanconi Anemia (FA) pathway encodes a DNA damage response activated by DNA damage-stalled replication forks. Current evidence suggests that the FA pathway initiates with DNA damage recognition by the FANCM complex (FANCM/FAAP24/MHF). However, genetic inactivation of FANCM in mouse and DT40 cells causes only a partial defect in the FA pathway activation, suggesting the existence of redundant DNA damage sensors. Here we show that the MutS homologs function in this capacity. A RNAi screen revealed that MSH2 silencing caused defective FA pathway activation, as assessed by damage-induced FANCD2 mono-ubiquitination. A similar FA pathway defect was observed with MSH3 or MSH6 silencing. MSH2 depletion caused cellular phenotypes associated with defective FA pathway, including mitomycin C hypersensitivity and chromosomal instability. Further, silencing of FANCM in MSH2 deficient HEC59 cells caused a more severe FA defect relative to comparable silencing in MSH2 complemented HEC59+Chr2 cells, suggesting redundant functions between MSH2 and FANCM. Consistent with this hypothesis, depletion of MSH2 resulted in defective chromatin localization of the FA core complex upon DNA damage. Further, MSH2 was co-purified and co-immunoprecipitated with FA core complex components. Taken together, our results suggest that human MutS homologs and FANCM complexes function as redundant DNA damage sensors of the FA pathway.


Sujet(s)
Altération de l'ADN , Helicase/métabolisme , Protéines de liaison à l'ADN/métabolisme , Anémie de Fanconi/génétique , Anémie de Fanconi/métabolisme , Animaux , Lignée cellulaire , Chromatine/métabolisme , Protéines de liaison à l'ADN/déficit , Protéines de liaison à l'ADN/génétique , Anémie de Fanconi/enzymologie , Extinction de l'expression des gènes , Humains , Souris
15.
Nat Med ; 17(7): 875-82, 2011 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-21706030

RÉSUMÉ

Cells that are deficient in homologous recombination, such as those that lack functional breast cancer-associated 1 (BRCA1) or BRCA2, are hypersensitive to inhibition of poly(ADP-ribose) polymerase (PARP). However, BRCA-deficient tumors represent only a small fraction of adult cancers, which might restrict the therapeutic utility of PARP inhibitor monotherapy. Cyclin-dependent kinase 1 (Cdk1) phosphorylates BRCA1, and this is essential for efficient formation of BRCA1 foci. Here we show that depletion or inhibition of Cdk1 compromises the ability of cells to repair DNA by homologous recombination. Combined inhibition of Cdk1 and PARP in BRCA-wild-type cancer cells resulted in reduced colony formation, delayed growth of human tumor xenografts and tumor regression with prolonged survival in a mouse model of lung adenocarcinoma. Inhibition of Cdk1 did not sensitize nontransformed cells or tissues to inhibition of PARP. Because reduced Cdk1 activity impaired BRCA1 function and consequently, repair by homologous recombination, inhibition of Cdk1 represents a plausible strategy for expanding the utility of PARP inhibitors to BRCA-proficient cancers.


Sujet(s)
Protéine BRCA1/physiologie , Tumeurs du sein/traitement médicamenteux , Protéine-kinase CDC2/physiologie , Inhibiteurs de poly(ADP-ribose) polymérases , Animaux , Protéine BRCA1/métabolisme , Benzimidazoles/pharmacologie , Technique de Western , Tumeurs du sein/métabolisme , Tumeurs du sein/physiopathologie , Protéine-kinase CDC2/antagonistes et inhibiteurs , Protéine-kinase CDC2/métabolisme , Protéines de transport/métabolisme , Protéines de transport/physiologie , Mort cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Altération de l'ADN/effets des médicaments et des substances chimiques , Altération de l'ADN/physiologie , Protéines de liaison à l'ADN , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/physiologie , Humains , Indazoles/pharmacologie , Indoles/pharmacologie , Mâle , Souris , Transplantation tumorale , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/physiopathologie , Protéines nucléaires/métabolisme , Protéines nucléaires/physiologie , Phosphorylation , Protéines de liaison à l'ARN
16.
Am J Med Genet A ; 155A(7): 1673-9, 2011 Jul.
Article de Anglais | MEDLINE | ID: mdl-21626672

RÉSUMÉ

We describe a newborn female with a de novo interstitial deletion of chromosome 21q21.1-22.12 including the RUNX1 gene who had developmental delay, multiple congenital anomalies, tetralogy of Fallot, anemia, and chronic thromobocytopenia requiring frequent platelet transfusions from birth. Because of her physical and hematologic abnormalities, she was tested for Fanconi anemia (FA). Lymphocytes and fibroblasts from this patient demonstrated increased chromosome breakage with exposure to the clastogen mitomycin C, but not, in contrast to most FA patients, to diepoxybutane. Further testing by Western analysis and complementation testing did not show a defect in the function of known Fanconi proteins. Her constitutional deletion was later found to span 13.2 Mb by chromosome microarray analysis, encompassing the RUNX1 gene that has been implicated in thrombocytopenia and predisposition to acute myelogenous leukemia (AML) when in the haploinsufficient state. We compare her phenotype to other individuals with similar 21q deletions and thrombocytopenia, as well as those with FA. We suggest that deletion of RUNX1 or another critical gene within the deleted region may result in chromosomal instability similar to that seen in FA.


Sujet(s)
Délétion de segment de chromosome , Chromosomes humains de la paire 21/génétique , Sous-unité alpha 2 du facteur CBF/génétique , Anémie de Fanconi/génétique , Cassure de chromosome/effets des médicaments et des substances chimiques , Anémie de Fanconi/diagnostic , Issue fatale , Femelle , Humains , Nourrisson , Nouveau-né , Caryotypage , Mutagènes/pharmacologie , Phénotype
17.
Nat Cell Biol ; 11(11): 1376-82, 2009 Nov.
Article de Anglais | MEDLINE | ID: mdl-19783983

RÉSUMÉ

DNA double-strand break (DSB) repair involves complex interactions between chromatin and repair proteins, including Tip60, a tumour suppressor. Tip60 is an acetyltransferase that acetylates both histones and ATM (ataxia telangiectasia mutated) kinase. Inactivation of Tip60 leads to defective DNA repair and increased cancer risk. However, how DNA damage activates the acetyltransferase activity of Tip60 is not known. Here, we show that direct interaction between the chromodomain of Tip60 and histone H3 trimethylated on lysine 9 (H3K9me3) at DSBs activates the acetyltransferase activity of Tip60. Depletion of intracellular H3K9me3 blocks activation of the acetyltransferase activity of Tip60, resulting in defective ATM activation and widespread defects in DSB repair. In addition, the ability of Tip60 to access H3K9me3 is dependent on the DNA damage-induced displacement of HP1beta (heterochromatin protein 1beta) from H3K9me3. Finally, we demonstrate that the Mre11-Rad50-Nbs1 (MRN) complex targets Tip60 to H3K9me3, and is required to activate the acetyltransferase activity of Tip60. These results reveal a new function for H3K9me3 in coordinating activation of Tip60-dependent DNA repair pathways, and imply that aberrant patterns of histone methylation may contribute to cancer by altering the efficiency of DSB repair.


Sujet(s)
Altération de l'ADN , Méthylation de l'ADN , Gènes suppresseurs de tumeur , Histone acetyltransferases/génétique , Histone/métabolisme , Homologue-5 de la protéine chromobox , Cellules HeLa , Humains , Lysine acetyltransferase 5
18.
Mol Cell Biol ; 29(21): 5911-22, 2009 Nov.
Article de Anglais | MEDLINE | ID: mdl-19704007

RÉSUMÉ

A growing body of evidence indicates that early mitotic inhibitor 1 (Emi1) is essential for genomic stability, but how this function relates to embryonic development and cancer pathogenesis remains unclear. We have identified a zebrafish mutant line in which deficient emi1 gene expression results in multilineage hematopoietic defects and widespread developmental defects that are p53 independent. Cell cycle analyses of Emi1-depleted zebrafish or human cells showed chromosomal rereplication, and metaphase preparations from mutant zebrafish embryos revealed rereplicated, unsegregated chromosomes and polyploidy. Furthermore, EMI1-depleted mammalian cells relied on topoisomerase II alpha-dependent mitotic decatenation to progress through metaphase. Interestingly, the loss of a single emi1 allele in the absence of p53 enhanced the susceptibility of adult fish to neural sheath tumorigenesis. Our results cast Emi1 as a critical regulator of genomic fidelity during embryogenesis and suggest that the factor may act as a tumor suppressor.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Développement embryonnaire/génétique , Génome/génétique , Tumeurs/anatomopathologie , Protéine p53 suppresseur de tumeur/métabolisme , Protéines de poisson-zèbre/métabolisme , Danio zébré/embryologie , Danio zébré/génétique , Animaux , Apoptose , Cycle cellulaire , Taille de la cellule , Altération de l'ADN , Embryon non mammalien/malformations , Embryon non mammalien/anatomopathologie , Hématopoïèse , Mutation/génétique , Cellules myéloïdes/anatomopathologie , Phénotype
19.
Blood ; 114(3): 647-50, 2009 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-19458356

RÉSUMÉ

To more comprehensively assess the pathogenic contribution of the PTEN-PI3K-AKT pathway to T-cell acute lymphoblastic leukemia (T-ALL), we examined diagnostic DNA samples from children with T-ALL using array comparative genomic hybridization and sequence analysis. Alterations of PTEN, PI3K, or AKT were identified in 47.7% of 44 cases. There was a striking clustering of PTEN mutations in exon 7 in 12 cases, all of which were predicted to truncate the C2 domain without disrupting the phosphatase domain of PTEN. Induction chemotherapy failed to induce remission in 3 of the 4 patients whose lymphoblasts harbored PTEN deletions at the time of diagnosis, compared with none of the 12 patients with mutations of PTEN exon 7 (P = .007), suggesting that PTEN deletion has more adverse therapeutic consequences than mutational disruptions that preserve the phosphatase domain. These findings add significant support to the rationale for the development of therapies targeting the PTEN-PI3K-AKT pathway in T-ALL.


Sujet(s)
Mutation , Phosphohydrolase PTEN/génétique , Phosphatidylinositol 3-kinases/génétique , Leucémie-lymphome lymphoblastique à précurseurs T/génétique , Protéines proto-oncogènes c-akt/génétique , Enfant , Hybridation génomique comparative , Résistance aux médicaments antinéoplasiques/génétique , Humains , Transduction du signal
20.
Mol Cell Biol ; 28(20): 6223-33, 2008 Oct.
Article de Anglais | MEDLINE | ID: mdl-18710953

RÉSUMÉ

There is a growing appreciation of the role that epigenetic alterations can play in oncogenesis. However, given the large number of genetic anomalies present in most cancers, it has been difficult to evaluate the extent to which epigenetic changes contribute to cancer. SNF5 (INI1/SMARCB1/BAF47) is a tumor suppressor that regulates the epigenome as a core member of the SWI/SNF chromatin remodeling complex. While the SWI/SNF complex displays potent tumor suppressor activity, it is unknown whether this activity is exerted genetically via maintenance of genome integrity or epigenetically via transcriptional regulation. Here we show that Snf5-deficient primary cells do not show altered sensitivity to DNA damaging agents, defects in gamma-H2AX induction, or an abrogated DNA damage checkpoint. Further, the aggressive malignancies that arise following SNF5 loss are diploid and genomically stable. Remarkably, we demonstrate that most human SNF5-deficient cancers lack genomic amplifications/deletions and, aside from SNF5 loss, are indistinguishable from normal cells on single-nucleotide polymorphism arrays. Finally, we show that epigenetically based changes in transcription that occur following SNF5 loss correlate with the tumor phenotype. Collectively, our results provide novel insight into the mechanisms of oncogenesis by demonstrating that disruption of a chromatin remodeling complex can largely, if not completely, substitute for genomic instability in the genesis of aggressive cancer.


Sujet(s)
Protéines chromosomiques nonhistones/déficit , Protéines de liaison à l'ADN/déficit , Épigenèse génétique , Instabilité du génome , Tumeurs/génétique , Facteurs de transcription/déficit , Animaux , Lignée cellulaire tumorale , Protéines chromosomiques nonhistones/métabolisme , Cisplatine/pharmacologie , Cycline D1/métabolisme , Altération de l'ADN , Réparation de l'ADN/effets des médicaments et des substances chimiques , Protéines de liaison à l'ADN/métabolisme , Diploïdie , Épigenèse génétique/effets des médicaments et des substances chimiques , Étoposide/pharmacologie , Phase G2/effets des médicaments et des substances chimiques , Instabilité du génome/effets des médicaments et des substances chimiques , Histone/métabolisme , Humains , Souris , Phénotype , Polymorphisme de nucléotide simple/génétique , Transport des protéines/effets des médicaments et des substances chimiques , Tumeur rhabdoïde/génétique , Protéine SMARCB1 , Facteurs de transcription/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...