Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
1.
Cancer Res ; 81(19): 5089-5101, 2021 10 01.
Article de Anglais | MEDLINE | ID: mdl-34215619

RÉSUMÉ

Somatic variants in TET2 and DNMT3A are founding mutations in hematological malignancies that affect the epigenetic regulation of DNA methylation. Mutations in both genes often co-occur with activating mutations in genes encoding oncogenic tyrosine kinases such as FLT3ITD, BCR-ABL1, JAK2V617F , and MPLW515L , or with mutations affecting related signaling pathways such as NRASG12D and CALRdel52 . Here, we show that TET2 and DNMT3A mutations exert divergent roles in regulating DNA repair activities in leukemia cells expressing these oncogenes. Malignant TET2-deficient cells displayed downregulation of BRCA1 and LIG4, resulting in reduced activity of BRCA1/2-mediated homologous recombination (HR) and DNA-PK-mediated non-homologous end-joining (D-NHEJ), respectively. TET2-deficient cells relied on PARP1-mediated alternative NHEJ (Alt-NHEJ) for protection from the toxic effects of spontaneous and drug-induced DNA double-strand breaks. Conversely, DNMT3A-deficient cells favored HR/D-NHEJ owing to downregulation of PARP1 and reduction of Alt-NHEJ. Consequently, malignant TET2-deficient cells were sensitive to PARP inhibitor (PARPi) treatment in vitro and in vivo, whereas DNMT3A-deficient cells were resistant. Disruption of TET2 dioxygenase activity or TET2-Wilms' tumor 1 (WT1)-binding ability was responsible for DNA repair defects and sensitivity to PARPi associated with TET2 deficiency. Moreover, mutation or deletion of WT1 mimicked the effect of TET2 mutation on DSB repair activity and sensitivity to PARPi. Collectively, these findings reveal that TET2 and WT1 mutations may serve as biomarkers of synthetic lethality triggered by PARPi, which should be explored therapeutically. SIGNIFICANCE: TET2 and DNMT3A mutations affect distinct DNA repair mechanisms and govern the differential sensitivities of oncogenic tyrosine kinase-positive malignant hematopoietic cells to PARP inhibitors.


Sujet(s)
DNA methyltransferase 3A/génétique , Réparation de l'ADN , Protéines de liaison à l'ADN/génétique , Dioxygenases/génétique , Résistance aux médicaments antinéoplasiques/génétique , Mutation , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Animaux , Systèmes CRISPR-Cas , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Techniques de knock-down de gènes , Génotype , Humains , Leucémies , Souris , Souris transgéniques , Modèles biologiques , Cellules souches tumorales , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Cell Rep ; 33(1): 108221, 2020 10 06.
Article de Anglais | MEDLINE | ID: mdl-33027668

RÉSUMÉ

Synthetic lethality triggered by PARP inhibitor (PARPi) yields promising therapeutic results. Unfortunately, tumor cells acquire PARPi resistance, which is usually associated with the restoration of homologous recombination, loss of PARP1 expression, and/or loss of DNA double-strand break (DSB) end resection regulation. Here, we identify a constitutive mechanism of resistance to PARPi. We report that the bone marrow microenvironment (BMM) facilitates DSB repair activity in leukemia cells to protect them against PARPi-mediated synthetic lethality. This effect depends on the hypoxia-induced overexpression of transforming growth factor beta receptor (TGFßR) kinase on malignant cells, which is activated by bone marrow stromal cells-derived transforming growth factor beta 1 (TGF-ß1). Genetic and/or pharmacological targeting of the TGF-ß1-TGFßR kinase axis results in the restoration of the sensitivity of malignant cells to PARPi in BMM and prolongs the survival of leukemia-bearing mice. Our finding may lead to the therapeutic application of the TGFßR inhibitor in patients receiving PARPis.


Sujet(s)
Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Récepteurs TGF-bêta/métabolisme , Protéine Smad-3/métabolisme , Animaux , Humains , Souris , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Microenvironnement tumoral
3.
Leuk Lymphoma ; 60(1): 262-264, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-29932782
5.
Blood ; 132(1): 67-77, 2018 07 05.
Article de Anglais | MEDLINE | ID: mdl-29784639

RÉSUMÉ

Mutations in FMS-like tyrosine kinase 3 (FLT3), such as internal tandem duplications (ITDs), can be found in up to 23% of patients with acute myeloid leukemia (AML) and confer a poor prognosis. Current treatment options for FLT3(ITD)-positive AMLs include genotoxic therapy and FLT3 inhibitors (FLT3i's), which are rarely curative. PARP1 inhibitors (PARP1i's) have been successfully applied to induce synthetic lethality in tumors harboring BRCA1/2 mutations and displaying homologous recombination (HR) deficiency. We show here that inhibition of FLT3(ITD) activity by the FLT3i AC220 caused downregulation of DNA repair proteins BRCA1, BRCA2, PALB2, RAD51, and LIG4, resulting in inhibition of 2 major DNA double-strand break (DSB) repair pathways, HR, and nonhomologous end-joining. PARP1i, olaparib, and BMN673 caused accumulation of lethal DSBs and cell death in AC220-treated FLT3(ITD)-positive leukemia cells, thus mimicking synthetic lethality. Moreover, the combination of FLT3i and PARP1i eliminated FLT3(ITD)-positive quiescent and proliferating leukemia stem cells, as well as leukemic progenitors, from human and mouse leukemia samples. Notably, the combination of AC220 and BMN673 significantly delayed disease onset and effectively reduced leukemia-initiating cells in an FLT3(ITD)-positive primary AML xenograft mouse model. In conclusion, we postulate that FLT3i-induced deficiencies in DSB repair pathways sensitize FLT3(ITD)-positive AML cells to synthetic lethality triggered by PARP1i's. Therefore, FLT3(ITD) could be used as a precision medicine marker for identifying AML patients that may benefit from a therapeutic regimen combining FLT3 and PARP1i's.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Réparation de l'ADN/effets des médicaments et des substances chimiques , Leucémie aigüe myéloïde , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Tyrosine kinase-3 de type fms/métabolisme , Animaux , Protéine BRCA1/génétique , Protéine BRCA1/métabolisme , Protéine BRCA2/génétique , Protéine BRCA2/métabolisme , Benzothiazoles/pharmacologie , Lignée cellulaire tumorale , DNA ligase ATP/génétique , DNA ligase ATP/métabolisme , Protéine du groupe de complémentation N de l'anémie de Fanconi/génétique , Protéine du groupe de complémentation N de l'anémie de Fanconi/métabolisme , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Souris , Mutation , Phénylurées/pharmacologie , Phtalazines/pharmacologie , Pipérazines/pharmacologie , Poly (ADP-Ribose) polymerase-1/génétique , Poly (ADP-Ribose) polymerase-1/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Rad51 Recombinase/génétique , Rad51 Recombinase/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Tyrosine kinase-3 de type fms/génétique
6.
Oncotarget ; 8(41): 69281-69294, 2017 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-29050203

RÉSUMÉ

There is substantial evidence that early growth response-1 (Egr1) gene, a zinc-finger transcription factor, behaves as a tumor suppressor in leukemia. This includes reports from this laboratory that constitutive Egr1 overrides leukemia conferred by deregulated c-Myc or E2F-1 in the M1 myeloid leukemic cell line by promoting differentiation. To investigate the effect of Egr1 on the initiation and progression of Chronic Myelogenous Leukemia (CML), lethally irradiated syngeneic wild type mice were reconstituted with bone marrow (BM) from either wild type or Egr1 null mice transduced with a 210-kD BCR-ABL-expressing MSCV-retrovirus (bone marrow transplantation {BMT}). Loss of Egr1 was observed to accelerate the development of BCR-ABL driven leukemia in recipient mice, resulting in the development of a more aggressive disease, a significantly shortened median survival time, and increased BCR-ABL expressing leukemic stem/progenitor cells (GFP+Lin-cKit+Sca+). Egr1 deficient progenitors expressing BCR-ABL exhibited decreased apoptosis, and increased cell viability and proliferation relative to WT counterparts. Secondary BMT of BCR-ABL BM revealed that loss of Egr1 resulted in enrichment of LSCs, consistent with shorter survival time and more aggressive disease of these mice compared to WT counterparts. Furthermore, serial re-plating colony assays indicated that loss of Egr1 increased self-renewal ability of BCR-ABL expressing BM. These novel findings on the tumor suppressor role of Egr1 in CML provide the impetus to study the effect of altering Egr1 expression in AML, where the overall five year survival rate remains low. The effect of loss of Egr1 in CML could reflect its established functions in normal hematopoiesis, maintaining quiescence of HSCs and driving terminal differentiation to the monocyte/macrophage lineage. Gain of function studies should validate these conclusions and provide further rationale for increased Egr1 as a therapeutic target in AML.

7.
Blood ; 130(26): 2848-2859, 2017 12 28.
Article de Anglais | MEDLINE | ID: mdl-29042365

RÉSUMÉ

Myeloproliferative neoplasms (MPNs) often carry JAK2(V617F), MPL(W515L), or CALR(del52) mutations. Current treatment options for MPNs include cytoreduction by hydroxyurea and JAK1/2 inhibition by ruxolitinib, both of which are not curative. We show here that cell lines expressing JAK2(V617F), MPL(W515L), or CALR(del52) accumulated reactive oxygen species-induced DNA double-strand breaks (DSBs) and were modestly sensitive to poly-ADP-ribose polymerase (PARP) inhibitors olaparib and BMN673. At the same time, primary MPN cell samples from individual patients displayed a high degree of variability in sensitivity to these drugs. Ruxolitinib inhibited 2 major DSB repair mechanisms, BRCA-mediated homologous recombination and DNA-dependent protein kinase-mediated nonhomologous end-joining, and, when combined with olaparib, caused abundant accumulation of toxic DSBs resulting in enhanced elimination of MPN primary cells, including the disease-initiating cells from the majority of patients. Moreover, the combination of BMN673, ruxolitinib, and hydroxyurea was highly effective in vivo against JAK2(V617F)+ murine MPN-like disease and also against JAK2(V617F)+, CALR(del52)+, and MPL(W515L)+ primary MPN xenografts. In conclusion, we postulate that ruxolitinib-induced deficiencies in DSB repair pathways sensitized MPN cells to synthetic lethality triggered by PARP inhibitors.


Sujet(s)
Réparation de l'ADN/effets des médicaments et des substances chimiques , Syndromes myéloprolifératifs/traitement médicamenteux , Tumeurs/traitement médicamenteux , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Pyrazoles/pharmacologie , Animaux , Calréticuline/génétique , Lignée cellulaire , Synergie des médicaments , Hétérogreffes , Humains , Kinase Janus-2/génétique , Souris , Syndromes myéloprolifératifs/génétique , Tumeurs/génétique , Nitriles , Phtalazines/pharmacologie , Pipérazines/pharmacologie , Pyrimidines , Récepteurs à la thrombopoïétine/génétique , Cellules cancéreuses en culture
8.
Mol Cancer Res ; 15(8): 967-972, 2017 08.
Article de Anglais | MEDLINE | ID: mdl-28634224

RÉSUMÉ

Burkitt lymphoma/leukemia cells carry t(8;14)(q24;q32) chromosomal translocation encoding IGH/MYC, which results in the constitutive expression of the MYC oncogene. Here, it is demonstrated that untreated and cytarabine (AraC)-treated IGH/MYC-positive Burkitt lymphoma cells accumulate a high number of potentially lethal DNA double-strand breaks (DSB) and display low levels of the BRCA2 tumor suppressor protein, which is a key element of homologous recombination (HR)-mediated DSB repair. BRCA2 deficiency in IGH/MYC-positive cells was associated with diminished HR activity and hypersensitivity to PARP1 inhibitors (olaparib, talazoparib) used alone or in combination with cytarabine in vitro Moreover, talazoparib exerted a therapeutic effect in NGS mice bearing primary Burkitt lymphoma xenografts. In conclusion, IGH/MYC-positive Burkitt lymphoma/leukemia cells have decreased BRCA2 and are sensitive to PARP1 inhibition alone or in combination with other chemotherapies.Implications: This study postulates that IGH/MYC-induced BRCA2 deficiency may predispose Burkitt lymphoma cells to synthetic lethality triggered by PARP1 inhibitors.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/15/8/967/F1.large.jpgMol Cancer Res; 15(8); 967-72. ©2017 AACR.


Sujet(s)
Protéine BRCA2/génétique , Lymphome de Burkitt/traitement médicamenteux , Cassures double-brin de l'ADN/effets des médicaments et des substances chimiques , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Animaux , Protéine BRCA2/déficit , Lymphome de Burkitt/génétique , Cytarabine/administration et posologie , Réparation de l'ADN/effets des médicaments et des substances chimiques , Gènes myc/génétique , Recombinaison homologue/effets des médicaments et des substances chimiques , Humains , Souris , Phtalazines/administration et posologie , Pipérazines/administration et posologie , Poly (ADP-Ribose) polymerase-1/génétique , Mutations synthétiques létales/génétique , Translocation génétique/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
9.
J Clin Invest ; 127(6): 2392-2406, 2017 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-28481221

RÉSUMÉ

Quiescent and proliferating leukemia cells accumulate highly lethal DNA double-strand breaks that are repaired by 2 major mechanisms: BRCA-dependent homologous recombination and DNA-dependent protein kinase-mediated (DNA-PK-mediated) nonhomologous end-joining, whereas DNA repair pathways mediated by poly(ADP)ribose polymerase 1 (PARP1) serve as backups. Here we have designed a personalized medicine approach called gene expression and mutation analysis (GEMA) to identify BRCA- and DNA-PK-deficient leukemias either directly, using reverse transcription-quantitative PCR, microarrays, and flow cytometry, or indirectly, by the presence of oncogenes such as BCR-ABL1. DNA-PK-deficient quiescent leukemia cells and BRCA/DNA-PK-deficient proliferating leukemia cells were sensitive to PARP1 inhibitors that were administered alone or in combination with current antileukemic drugs. In conclusion, GEMA-guided targeting of PARP1 resulted in dual cellular synthetic lethality in quiescent and proliferating immature leukemia cells, and is thus a potential approach to eradicate leukemia stem and progenitor cells that are responsible for initiation and manifestation of the disease. Further, an analysis of The Cancer Genome Atlas database indicated that this personalized medicine approach could also be applied to treat numerous solid tumors from individual patients.


Sujet(s)
Antinéoplasiques/pharmacologie , Prolifération cellulaire , Leucémies/génétique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Animaux , Lignée cellulaire tumorale , Transformation cellulaire néoplasique , Cricetinae , Cassures double-brin de l'ADN , Réparation de l'ADN par jonction d'extrémités , Gène BRCA1 , Gène BRCA2 , Gènes létaux , Gènes abl , Humains , Leucémies/traitement médicamenteux , Souris , Souris de lignée NOD , Souris knockout , Souris SCID , Cellules souches embryonnaires de souris/physiologie , Phtalazines/pharmacologie , Pipérazines/pharmacologie , Transcriptome , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Oncotarget ; 8(7): 10809-10821, 2017 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-28086219

RÉSUMÉ

The Gadd45a stress sensor gene is a member in the Gadd45 family of genes that includes Gadd45b & Gadd45g. To investigate the effect of GADD45A in the development of CML, syngeneic wild type lethally irradiated mice were reconstituted with either wild type or Gadd45a null myeloid progenitors transduced with a retroviral vector expressing the 210-kD BCR-ABL fusion oncoprotein. Loss of Gadd45a was observed to accelerate BCR-ABL driven CML resulting in the development of a more aggressive disease, a significantly shortened median mice survival time, and increased BCR-ABL expressing leukemic stem/progenitor cells (GFP+Lin- cKit+Sca+). GADD45A deficient progenitors expressing BCR-ABL exhibited increased proliferation and decreased apoptosis relative to WT counterparts, which was associated with enhanced PI3K-AKT-mTOR-4E-BP1 signaling, upregulation of p30C/EBPα expression, and hyper-activation of p38 and Stat5. Furthermore, Gadd45a expression in samples obtained from CML patients was upregulated in more indolent chronic phase CML samples and down regulated in aggressive accelerated phase CML and blast crisis CML. These results provide novel evidence that Gadd45a functions as a suppressor of BCR/ABL driven leukemia and may provide a unique prognostic marker of CML progression.


Sujet(s)
Protéines du cycle cellulaire/génétique , Protéines de fusion bcr-abl/génétique , Régulation de l'expression des gènes dans la leucémie , Leucémie myéloïde chronique BCR-ABL positive/génétique , Protéines nucléaires/génétique , Animaux , Apoptose/génétique , Crise blastique/génétique , Crise blastique/métabolisme , Transplantation de moelle osseuse/méthodes , Protéines du cycle cellulaire/déficit , Protéines du cycle cellulaire/métabolisme , Prolifération cellulaire/génétique , Cellules cultivées , Cytométrie en flux , Protéines de fusion bcr-abl/métabolisme , Humains , Immunotransfert , Cellules K562 , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Souris de souche-129 , Souris knockout , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Protéines nucléaires/déficit , Protéines nucléaires/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , RT-PCR , Transduction du signal
11.
Blood Adv ; 1(19): 1467-1472, 2017 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-29296788

RÉSUMÉ

PARP1 is required for the maintenance of MLL-AF9 leukemias.PARP1 inhibitors enhance the therapeutic effect of cytotoxic drugs against MLL-AF9 leukemias.

12.
Oncotarget ; 7(19): 26935-48, 2016 May 10.
Article de Anglais | MEDLINE | ID: mdl-27105496

RÉSUMÉ

The GADD45 family of proteins functions as stress sensors in response to various physiological and environmental stressors. Here we show that primary mouse embryo fibroblasts (MEFs) from Gadd45b null mice proliferate slowly, accumulate increased levels of DNA damage, and senesce prematurely. The impaired proliferation and increased senescence in Gadd45b null MEFs is partially reversed by culturing at physiological oxygen levels, indicating that Gadd45b deficiency leads to decreased ability to cope with oxidative stress. Interestingly, Gadd45b null MEFs arrest at the G2/M phase of cell cycle, in contrast to other senescent MEFs, which arrest at G1. FACS analysis of phospho-histone H3 staining showed that Gadd45b null MEFs are arrested in G2 phase rather than M phase. H2O2 and UV irradiation, known to increase oxidative stress, also triggered increased senescence in Gadd45b null MEFs compared to wild type MEFs. In vivo evidence for increased senescence in Gadd45b null mice includes the observation that embryos from Gadd45b null mice exhibit increased senescence staining compared to wild type embryos. Furthermore, it is shown that Gadd45b deficiency promotes senescence and aging phenotypes in mouse skin. Together, these results highlight a novel role for Gadd45b in stress-induced senescence and in tissue aging.


Sujet(s)
Antigènes de différenciation/génétique , Vieillissement de la cellule/génétique , Fibroblastes/métabolisme , Vieillissement de la peau/génétique , Animaux , Antigènes de différenciation/métabolisme , Prolifération cellulaire/génétique , Cellules cultivées , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des radiations , Embryon de mammifère/cytologie , Embryon de mammifère/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/effets des radiations , Points de contrôle de la phase G2 du cycle cellulaire/génétique , Peroxyde d'hydrogène/pharmacologie , Souris knockout , Oxydants/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Stress oxydatif/effets des radiations , Oxygène/métabolisme , Rayons ultraviolets
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...