Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Curr Oncol ; 29(2): 497-509, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35200545

ABSTRACT

Background: 5-FU-based chemoradiotherapy (CRT) could be associated with severe treatment-related toxicities in patients harboring at-risk DPYD polymorphisms. Methods: The studied population included consecutive patients with locoregionally advanced oropharyngeal carcinoma treated with carboplatin and 5-FU-based CRT one year before and after the implementation of upfront DPYD*2A genotyping. We aimed to determine the effect of DPYD genotyping on grade ≥3 toxicities. Results: 181 patients were analyzed (87 patients before and 94 patients following DPYD*2A screening). Of the patients, 91% (n = 86) were prospectively genotyped for the DPYD*2A allele. Of those screened, 2% (n = 2/87) demonstrated a heterozygous DPYD*2A mutation. Extended genotyping of DPYD*2A-negative patients later allowed for the retrospective identification of six additional patients with alternative DPYD variants (two c.2846A>T and four c.1236G>A mutations). Grade ≥3 toxicities occurred in 71% of the patients before DPYD*2A screening versus 62% following upfront genotyping (p = 0.18). When retrospectively analyzing additional non-DPYD*2A variants, the relative risks for mucositis (RR 2.36 [1.39-2.13], p = 0.0063), dysphagia (RR 2.89 [1.20-5.11], p = 0.019), and aspiration pneumonia (RR 13 [2.42-61.5)], p = 0.00065) were all significantly increased. Conclusion: The DPYD*2A, c.2846A>T, and c.1236G>A polymorphisms are associated with an increased risk of grade ≥3 toxicity to 5-FU. Upfront DPYD genotyping can identify patients in whom 5-FU-related toxicity should be avoided.


Subject(s)
Carcinoma , Dihydrouracil Dehydrogenase (NADP) , Chemoradiotherapy/adverse effects , Dihydrouracil Dehydrogenase (NADP)/genetics , Genotype , Humans , Retrospective Studies
2.
J Immunol ; 207(6): 1599-1615, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34408010

ABSTRACT

GFI1 is a DNA-binding transcription factor that regulates hematopoiesis by repressing target genes through its association with complexes containing histone demethylases such as KDM1A (LSD1) and histone deacetylases (HDACs). To study the consequences of the disruption of the complex between GFI1 and histone-modifying enzymes, we have used knock-in mice harboring a P2A mutation in GFI1 coding region that renders it unable to bind LSD1 and associated histone-modifying enzymes such as HDACs. GFI1P2A mice die prematurely and show increased numbers of memory effector and regulatory T cells in the spleen accompanied by a severe systemic inflammation with high serum levels of IL-6, TNF-α, and IL-1ß and overexpression of the gene encoding the cytokine oncostatin M (OSM). We identified lung alveolar macrophages, CD8 T cell from the spleen and thymic eosinophils, and monocytes as the sources of these cytokines in GFI1P2A mice. Chromatin immunoprecipitation showed that GFI1/LSD1 complexes occupy sites at the Osm promoter and an intragenic region of the Tnfα gene and that a GFI1P2A mutant still remains bound at these sites even without LSD1. Methylation and acetylation of histone H3 at these sites were enriched in cells from GFI1P2A mice, the H3K27 acetylation being the most significant. These data suggest that the histone modification facilitated by GFI1 is critical to control inflammatory pathways in different cell types, including monocytes and eosinophils, and that a disruption of GFI1-associated complexes can lead to systemic inflammation with fatal consequences.


Subject(s)
DNA-Binding Proteins/deficiency , Histone Demethylases/metabolism , Mutant Proteins/metabolism , Signal Transduction/genetics , Systemic Inflammatory Response Syndrome/blood , Transcription Factors/deficiency , Animals , Bone Marrow Transplantation/methods , CD8-Positive T-Lymphocytes/immunology , Cytokines/blood , Cytokines/genetics , DNA-Binding Proteins/genetics , Female , Gene Expression , Gene Knock-In Techniques , Histones/metabolism , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Protein Binding , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/immunology , Transcription Factors/genetics
3.
Oncologist ; 26(4): e597-e602, 2021 04.
Article in English | MEDLINE | ID: mdl-33274825

ABSTRACT

BACKGROUND: Fluoropyrimidines are used in chemotherapy combinations for multiple cancers. Deficient dihydropyrimidine dehydrogenase activity can lead to severe life-threatening toxicities. DPYD*2A polymorphism is one of the most studied variants. The study objective was to document the impact of implementing this test in routine clinical practice. METHODS: We retrospectively performed chart reviews of all patients who tested positive for a heterozygous or homozygous DPYD*2A mutation in samples obtained from patients throughout the province of Quebec, Canada. RESULTS: During a period of 17 months, 2,617 patients were tested: 25 patients tested positive. All were White. Twenty-four of the 25 patients were heterozygous (0.92%), and one was homozygous (0.038%). Data were available for 20 patients: 15 were tested upfront, whereas five were identified after severe toxicities. Of the five patients confirmed after toxicities, all had grade 4 cytopenias, 80% grade ≥3 mucositis, 20% grade 3 rash, and 20% grade 3 diarrhea. Eight patients identified with DPYD*2A mutation prior to treatment received fluoropyrimidine-based chemotherapy at reduced initial doses. The average fluoropyrimidine dose intensity during chemotherapy was 50%. No grade ≥3 toxicities were observed. DPYD*2A test results were available in an average of 6 days, causing no significant delays in treatment initiation. CONCLUSION: Upfront genotyping before fluoropyrimidine-based treatment is feasible in clinical practice and can prevent severe toxicities and hospitalizations without delaying treatment initiation. The administration of chemotherapy at reduced doses appears to be safe in patients heterozygous for DPYD*2A. IMPLICATIONS FOR PRACTICE: Fluoropyrimidines are part of chemotherapy combinations for multiple cancers. Deficient dihydropyrimidine dehydrogenase activity can lead to severe life-threatening toxicities. This retrospective analysis demonstrates that upfront genotyping of DPYD before fluoropyrimidine-based treatment is feasible in clinical practice and can prevent severe toxicities and hospitalizations without delaying treatment initiation. This approach was reported previously, but insufficient data concerning its application in real practice are available. This is likely the first reported experience of systematic DPYD genotyping all over Canada and North America as well.


Subject(s)
Dihydrouracil Dehydrogenase (NADP) , Fluorouracil , Antimetabolites, Antineoplastic , Canada , Capecitabine/adverse effects , Dihydrouracil Dehydrogenase (NADP)/genetics , Genotype , Humans , Quebec/epidemiology , Retrospective Studies
4.
Cancer Res ; 79(16): 4184-4195, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31273062

ABSTRACT

Acute lymphoblastic leukemia (ALL) is an aggressive blood cancer that mainly affects children. Relapse rates are high and toxic chemotherapies that block DNA replication and induce DNA damage lead to health problems later in life, underlining the need for improved therapies. MYC is a transcription factor that is hyperactive in a large proportion of cancers including leukemia but is difficult to target in therapy. We show that ablation of the function of the BTB/POZ domain factor Zbtb17 (Miz-1), an important cofactor of c-Myc, significantly delayed T- and B-ALL/lymphoma in mice and interfered with the oncogenic transcriptional activity of c-Myc. Leukemic cells that still emerged in this system activated DNA replication pathways that could be targeted by current chemotherapeutic drugs such as cytarabine. Acute ablation of the Miz-1 POZ domain enhanced the effect of cytarabine treatment. The combined treatment was effective in both Eµ-Myc and Notch ICN-driven leukemia models and prolonged survival of tumor-bearing animals by accelerating apoptosis of leukemic cells. These observations suggest that targeting MIZ-1 could render current ALL chemotherapies more effective, with a better outcome for patients. SIGNIFICANCE: Ablation of the POZ domain of Miz-1 perturbs its interaction with c-MYC and delays the generation of T- and B-cell leukemias and lymphomas.


Subject(s)
Cytarabine/pharmacology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Inhibitors of Activated STAT/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Antimetabolites, Antineoplastic/pharmacology , Female , Gene Expression Regulation, Neoplastic , Genes, myc , Mice, Transgenic , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Domains , Protein Inhibitors of Activated STAT/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
Sci Rep ; 9(1): 6304, 2019 04 19.
Article in English | MEDLINE | ID: mdl-31004086

ABSTRACT

Here we demonstrate a mode of reciprocal regulation between GFI1 and p53 that controls the induction of apoptosis in T cells. We show that GFI1 prevents induction of p53 dependent apoptosis by recruiting LSD1 to p53, which leads to the demethylation of its C-terminal domain. This is accompanied by a decrease of the acetylation of lysine 117 within the core domain of the murine p53 protein, which is required for transcriptional induction of apoptosis. Our results support a model in which the effect of GFI1's regulation of methylation at the c-terminus of p53 is ultimately mediated through control of acetylation at lysine 117 of p53. We propose that GFI1 acts prior to the occurrence of DNA damage to affect the post-translational modification state and limit the subsequent activation of p53. Once activated, p53 then transcriptionally activates GFI1, presumably in order to re-establish the homeostatic balance of p53 activity. These findings have implications for the activity level of p53 in various disease contexts where levels of GFI1 are either increased or decreased.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , T-Lymphocytes/cytology , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics
6.
Nat Commun ; 10(1): 1270, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894540

ABSTRACT

Gfi1b is a transcriptional repressor expressed in hematopoietic stem cells (HSCs) and megakaryocytes (MKs). Gfi1b deficiency leads to expansion of both cell types and abrogates the ability of MKs to respond to integrin. Here we show that Gfi1b forms complexes with ß-catenin, its co-factors Pontin52, CHD8, TLE3 and CtBP1 and regulates Wnt/ß-catenin-dependent gene expression. In reporter assays, Gfi1b can activate TCF-dependent transcription and Wnt3a treatment enhances this activation. This requires interaction between Gfi1b and LSD1 and suggests that a tripartite ß-catenin/Gfi1b/LSD1 complex exists, which regulates Wnt/ß-catenin target genes. Consistently, numerous canonical Wnt/ß-catenin target genes, co-occupied by Gfi1b, ß-catenin and LSD1, have their expression deregulated in Gfi1b-deficient cells. When Gfi1b-deficient cells are treated with Wnt3a, their normal cellularity is restored and Gfi1b-deficient MKs regained their ability to spread on integrin substrates. This indicates that Gfi1b controls both the cellularity and functional integrity of HSCs and MKs by regulating Wnt/ß-catenin signaling pathway.


Subject(s)
Hematopoietic Stem Cells/metabolism , Megakaryocytes/metabolism , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Wnt Signaling Pathway , Wnt3A Protein/genetics , beta Catenin/genetics , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Gene Ontology , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Hematopoietic Stem Cells/cytology , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , K562 Cells , Megakaryocytes/cytology , Mice , Mice, Knockout , Molecular Sequence Annotation , Primary Cell Culture , Proto-Oncogene Proteins/deficiency , Repressor Proteins/deficiency , Tamoxifen , Wnt3A Protein/metabolism , beta Catenin/metabolism
7.
Leukemia ; 33(1): 110-121, 2019 01.
Article in English | MEDLINE | ID: mdl-29925903

ABSTRACT

Growth factor independent 1 (Gfi1) controls myeloid differentiation by regulating gene expression and limits the activation of p53 by facilitating its de-methylation at Lysine 372. In human myeloid leukemia, low GFI1 levels correlate with an inferior prognosis. Here, we show that knockdown (KD) of Gfi1 in mice causes a fatal myeloproliferative disease (MPN) that could progress to leukemia after additional mutations. Both KO and KD mice accumulate myeloid cells that show signs of metabolic stress and high levels of reactive oxygen species. However, only KO cells have elevated levels of Lysine 372 methylated p53. This suggests that in contrast to absence of GFI1, KD of GFI1 leads to the accumulation of myeloid cells because sufficient amount of GFI1 is present to impede p53-mediated cell death, leading to a fatal MPN. The combination of myeloid accumulation and the ability to counteract p53 activity under metabolic stress could explain the role of reduced GF1 expression in human myeloid leukemia.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/physiology , Leukemia, Myeloid/pathology , Myeloid Cells/pathology , Myeloproliferative Disorders/pathology , Transcription Factors/physiology , Animals , Leukemia, Myeloid/etiology , Leukemia, Myeloid/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/metabolism , Oxidative Stress , TRPC Cation Channels/physiology
8.
Nat Commun ; 9(1): 1418, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29651020

ABSTRACT

GFI1 is a transcriptional regulator expressed in lymphoid cells, and an "oncorequisite" factor required for development and maintenance of T-lymphoid leukemia. GFI1 deletion causes hypersensitivity to ionizing radiation, for which the molecular mechanism remains unknown. Here, we demonstrate that GFI1 is required in T cells for the regulation of key DNA damage signaling and repair proteins. Specifically, GFI1 interacts with the arginine methyltransferase PRMT1 and its substrates MRE11 and 53BP1. We demonstrate that GFI1 enables PRMT1 to bind and methylate MRE11 and 53BP1, which is necessary for their function in the DNA damage response. Thus, our results provide evidence that GFI1 can adopt non-transcriptional roles, mediating the post-translational modification of proteins involved in DNA repair. These findings have direct implications for treatment responses in tumors overexpressing GFI1 and suggest that GFI1's activity may be a therapeutic target in these malignancies.


Subject(s)
CD4-Positive T-Lymphocytes/radiation effects , DNA Repair , DNA-Binding Proteins/metabolism , MRE11 Homologue Protein/metabolism , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , DNA Damage , DNA-Binding Proteins/genetics , Gamma Rays , Humans , Jurkat Cells , MRE11 Homologue Protein/genetics , Methylation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor p53-Binding Protein 1/genetics
9.
Cell Stress ; 2(8): 213-215, 2018 Jul 24.
Article in English | MEDLINE | ID: mdl-31225488

ABSTRACT

Despite recent advances in cancer treatment through personalized and precision medicine and new avenues such as immunotherapy and chimeric antibodies, the induction of DNA damage either through irradiation or specific compounds remains the primary approach to kill tumour cells. Improvements in our understanding of how tumour cells respond to DNA damage, and especially how this response differs from that of normal cells, are crucial to the development of better and more efficient therapies. We have recently shown that the activity of the oncogenic transcription factor GFI1, which is required for the development and maintenance of T and B cell leukemia, increases the ability of tumour cells to repair their DNA following damage (Vadnais et al. Nat Commun 9(1):1418). GFI1 accomplishes this by regulating the post-translational modifications (PTM) of key DNA repair proteins, including MRE11 and 53BP1, by the methyltransferase PRMT1. Here, GFI1 acts as an accessory protein required for the interaction between the enzyme and its substrates. This has implications for the treatment response of tumour cells overexpressing GFI1, which includes T cell leukemia, neuroendocrine lung carcinomas and aggressive subtypes of medulloblastoma, and suggests that targeting GFI1's activity and with this its capacity to aid DNA repair may open avenues for new therapeutic approaches.

10.
Haematologica ; 102(3): 484-497, 2017 03.
Article in English | MEDLINE | ID: mdl-28082345

ABSTRACT

Mutations in GFI1B are associated with inherited bleeding disorders called GFI1B-related thrombocytopenias. We show here that mice with a megakaryocyte-specific Gfi1b deletion exhibit a macrothrombocytopenic phenotype along a megakaryocytic dysplasia reminiscent of GFI1B-related thrombocytopenia. GFI1B deficiency increases megakaryocyte proliferation and affects their ploidy, but also abrogates their responsiveness towards integrin signaling and their ability to spread and reorganize their cytoskeleton. Gfi1b-null megakaryocytes are also unable to form proplatelets, a process independent of integrin signaling. GFI1B-deficient megakaryocytes exhibit aberrant expression of several components of both the actin and microtubule cytoskeleton, with a dramatic reduction of α-tubulin. Inhibition of FAK or ROCK, both important for actin cytoskeleton organization and integrin signaling, only partially restored their response to integrin ligands, but the inhibition of PAK, a regulator of the actin cytoskeleton, completely rescued the responsiveness of Gfi1b-null megakaryocytes to ligands, but not their ability to form proplatelets. We conclude that Gfi1b controls major functions of megakaryocytes such as integrin-dependent cytoskeleton organization, spreading and migration through the regulation of PAK activity whereas the proplatelet formation defect in GFI1B-deficient megakaryocytes is due, at least partially, to an insufficient α-tubulin content.


Subject(s)
Cytoskeleton/metabolism , Integrins/metabolism , Megakaryocytes/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Actins/chemistry , Actins/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Profiling , Genetic Association Studies , Megakaryocytes/drug effects , Megakaryocytes/pathology , Megakaryocytes/ultrastructure , Mice , Mice, Knockout , Microtubules/metabolism , Phenotype , Platelet Count , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Thrombocytopenia/blood , Thrombocytopenia/diagnosis , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Transcriptome
11.
PLoS One ; 11(7): e0160344, 2016.
Article in English | MEDLINE | ID: mdl-27467586

ABSTRACT

A regulatory circuit that controls myeloid versus B lymphoid cell fate in hematopoietic progenitors has been proposed, in which a network of the transcription factors Egr1/2, Nab, Gfi1 and PU.1 forms the core element. Here we show that a direct link between Gfi1, the transcription factor E2A and its inhibitor Id1 is a critical element of this regulatory circuit. Our data suggest that a certain threshold of Gfi1 is required to gauge E2A activity by adjusting levels of Id1 in multipotent progenitors, which are the first bipotential myeloid/lymphoid-restricted progeny of hematopoietic stem cells. If Gfi1 levels are high, Id1 is repressed enabling E2A to activate a specific set of B lineage genes by binding to regulatory elements for example the IL7 receptor gene. If Gfi1 levels fall below a threshold, Id1 expression increases and renders E2A unable to function, which prevents hematopoietic progenitors from engaging along the B lymphoid lineage.


Subject(s)
B-Lymphocytes/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Transcription Factors/metabolism , Animals , B-Lymphocytes/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Gene Expression Profiling , Mice , Mice, Transgenic
12.
Sci Rep ; 6: 27379, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27271479

ABSTRACT

The proliferation and survival of hematopoietic stem cells (HSCs) has to be strictly coordinated to ensure the timely production of all blood cells. Here we report that the splice factor and RNA binding protein hnRNP L (heterogeneous nuclear ribonucleoprotein L) is required for hematopoiesis, since its genetic ablation in mice reduces almost all blood cell lineages and causes premature death of the animals. In agreement with this, we observed that hnRNP L deficient HSCs lack both the ability to self-renew and foster hematopoietic differentiation in transplanted hosts. They also display mitochondrial dysfunction, elevated levels of γH2AX, are Annexin V positive and incorporate propidium iodide indicating that they undergo cell death. Lin(-)c-Kit(+) fetal liver cells from hnRNP L deficient mice show high p53 protein levels and up-regulation of p53 target genes. In addition, cells lacking hnRNP L up-regulated the expression of the death receptors TrailR2 and CD95/Fas and show Caspase-3, Caspase-8 and Parp cleavage. Treatment with the pan-caspase inhibitor Z-VAD-fmk, but not the deletion of p53, restored cell survival in hnRNP L deficient cells. Our data suggest that hnRNP L is critical for the survival and functional integrity of HSCs by restricting the activation of caspase-dependent death receptor pathways.


Subject(s)
Cell Survival/physiology , Hematopoietic Stem Cells/cytology , Heterogeneous-Nuclear Ribonucleoprotein L/physiology , Animals , Apoptosis/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Stress, Physiological , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Exp Hematol ; 44(8): 713-726.e14, 2016 08.
Article in English | MEDLINE | ID: mdl-27216773

ABSTRACT

Epigenetic changes can contribute to development of acute myeloid leukemia (AML), a malignant disease of the bone marrow. A single-nucleotide polymorphism of transcription factor growth factor independence 1 (GFI1) generates a protein with an asparagine at position 36 (GFI1(36N)) instead of a serine at position 36 (GFI1(36S)), which is associated with de novo AML in humans. However, how GFI1(36N) predisposes to AML is poorly understood. To explore the mechanism, we used knock-in mouse strains expressing GFI1(36N) or GFI1(36S). Presence of GFI1(36N) shortened the latency and increased the incidence of AML in different murine models of myelodysplastic syndrome/AML. On a molecular level, GFI1(36N) induced genomewide epigenetic changes, leading to expression of AML-associated genes. On a therapeutic level, use of histone acetyltransferase inhibitors specifically impeded growth of GFI1(36N)-expressing human and murine AML cells in vitro and in vivo. These results establish, as a proof of principle, how epigenetic changes in GFI1(36N)-induced AML can be targeted.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Leukemia, Myeloid, Acute/genetics , Mutation , Transcription Factors/genetics , Amino Acid Substitution , Animals , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/genetics , Codon , Disease Models, Animal , Disease Progression , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Leukemic , Genetic Predisposition to Disease , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Transgenic , Models, Biological , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/mortality
14.
Oncotarget ; 6(6): 3613-26, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25682875

ABSTRACT

Despite having long telomeres, mouse embryo fibroblasts (MEFs) senesce more rapidly than human diploid fibroblasts because of the accumulation of oxidative DNA damage. The CUX1 homeodomain protein was recently found to prevent senescence in RAS-driven cancer cells that produce elevated levels of reactive-oxygen species. Here we show that Cux1-/- MEFs are unable to proliferate in atmospheric (20%) oxygen although they can proliferate normally in physiological (3%) oxygen levels. CUX1 contains three domains called Cut repeats. Structure/function analysis established that a single Cut repeat domain can stimulate the DNA binding, Schiff-base formation, glycosylase and AP-lyase activities of 8-oxoguanine DNA glycosylase 1, OGG1. Strikingly and in contrast to previous reports, OGG1 exhibits efficient AP-lyase activity in the presence of a Cut repeat. Repair of oxidative DNA damage and proliferation in 20% oxygen were both rescued in Cux1-/- MEFs by ectopic expression of CUX1 or of a recombinant Cut repeat protein that stimulates OGG1 but is devoid of transcription activation potential. These findings reinforce the causal link between oxidative DNA damage and cellular senescence and suggest that the role of CUX1 as an accessory factor in DNA repair will be critical in physiological situations that generate higher levels of reactive oxygen species.


Subject(s)
Cellular Senescence/genetics , DNA Repair , Fibroblasts/cytology , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Repressor Proteins/metabolism , Animals , DNA Damage , Fibroblasts/metabolism , HCT116 Cells , Homeodomain Proteins/genetics , Humans , Mice , Nuclear Proteins/genetics , Oxidation-Reduction , Oxidative Stress/genetics , Repressor Proteins/genetics , Transcription Factors
15.
Proc Natl Acad Sci U S A ; 111(50): E5411-9, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25468973

ABSTRACT

To be effective, the adaptive immune response requires a large repertoire of antigen receptors, which are generated through V(D)J recombination in lymphoid precursors. These precursors must be protected from DNA damage-induced cell death, however, because V(D)J recombination generates double-strand breaks and may activate p53. Here we show that the BTB/POZ domain protein Miz-1 restricts p53-dependent induction of apoptosis in both pro-B and DN3a pre-T cells that actively rearrange antigen receptor genes. Miz-1 exerts this function by directly activating the gene for ribosomal protein L22 (Rpl22), which binds to p53 mRNA and negatively regulates its translation. This mechanism limits p53 expression levels and thus contains its apoptosis-inducing functions in lymphocytes, precisely at differentiation stages in which V(D)J recombination occurs.


Subject(s)
Gene Expression Regulation/physiology , Lymphoid Progenitor Cells/physiology , Nuclear Proteins/metabolism , Protein Biosynthesis/physiology , Protein Inhibitors of Activated STAT/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , V(D)J Recombination/genetics , Analysis of Variance , Animals , Cell Death/physiology , Chromatin Immunoprecipitation , Flow Cytometry , Gene Expression Regulation/genetics , Genetic Vectors/genetics , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Protein Biosynthesis/genetics , Protein Inhibitors of Activated STAT/genetics , Real-Time Polymerase Chain Reaction , Ubiquitin-Protein Ligases , V(D)J Recombination/physiology
16.
Biol Open ; 3(10): 937-46, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25217618

ABSTRACT

Autocrine activation of the Wnt/ß-catenin pathway occurs in several cancers, notably in breast tumors, and is associated with higher expression of various Wnt ligands. Using various inhibitors of the FZD/LRP receptor complex, we demonstrate that some adenosquamous carcinomas that develop in MMTV-CUX1 transgenic mice represent a model for autocrine activation of the Wnt/ß-catenin pathway. By comparing expression profiles of laser-capture microdissected mammary tumors, we identify Glis1 as a transcription factor that is highly expressed in the subset of tumors with elevated Wnt gene expression. Analysis of human cancer datasets confirms that elevated WNT gene expression is associated with high levels of CUX1 and GLIS1 and correlates with genes of the epithelial-to-mesenchymal transition (EMT) signature: VIM, SNAI1 and TWIST1 are elevated whereas CDH1 and OCLN are decreased. Co-expression experiments demonstrate that CUX1 and GLIS1 cooperate to stimulate TCF/ß-catenin transcriptional activity and to enhance cell migration and invasion. Altogether, these results provide additional evidence for the role of GLIS1 in reprogramming gene expression and suggest a hierarchical model for transcriptional regulation of the Wnt/ß-catenin pathway and the epithelial-to-mesenchymal transition.

17.
PLoS Biol ; 12(3): e1001807, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24618719

ABSTRACT

The Cut homeobox 1 (CUX1) gene is a target of loss-of-heterozygosity in many cancers, yet elevated CUX1 expression is frequently observed and is associated with shorter disease-free survival. The dual role of CUX1 in cancer is illustrated by the fact that most cell lines with CUX1 LOH display amplification of the remaining allele, suggesting that decreased CUX1 expression facilitates tumor development while increased CUX1 expression is needed in tumorigenic cells. Indeed, CUX1 was found in a genome-wide RNAi screen to identify synthetic lethal interactions with oncogenic RAS. Here we show that CUX1 functions in base excision repair as an ancillary factor for the 8-oxoG-DNA glycosylase, OGG1. Single cell gel electrophoresis (comet assay) reveals that Cux1⁺/⁻ MEFs are haploinsufficient for the repair of oxidative DNA damage, whereas elevated CUX1 levels accelerate DNA repair. In vitro base excision repair assays with purified components demonstrate that CUX1 directly stimulates OGG1's enzymatic activity. Elevated reactive oxygen species (ROS) levels in cells with sustained RAS pathway activation can cause cellular senescence. We show that elevated expression of either CUX1 or OGG1 prevents RAS-induced senescence in primary cells, and that CUX1 knockdown is synthetic lethal with oncogenic RAS in human cancer cells. Elevated CUX1 expression in a transgenic mouse model enables the emergence of mammary tumors with spontaneous activating Kras mutations. We confirmed cooperation between Kras(G12V) and CUX1 in a lung tumor model. Cancer cells can overcome the antiproliferative effects of excessive DNA damage by inactivating a DNA damage response pathway such as ATM or p53 signaling. Our findings reveal an alternate mechanism to allow sustained proliferation in RAS-transformed cells through increased DNA base excision repair capability. The heightened dependency of RAS-transformed cells on base excision repair may provide a therapeutic window that could be exploited with drugs that specifically target this pathway.


Subject(s)
DNA Damage , DNA Repair/physiology , Genes, ras/physiology , Homeodomain Proteins/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cellular Senescence/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mammary Neoplasms, Experimental/genetics , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidative Stress , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors
18.
BMC Genomics ; 14: 258, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23590133

ABSTRACT

BACKGROUND: Overexpression of the Cut homeobox 1 gene, CUX1, inversely correlates with patient survival in breast cancers. Cell-based assays and molecular studies have revealed that transcriptional regulation by CUX1 involves mostly the proteolytically processed p110 isoform. As there is no antibody specific to p110 CUX1 only, an alternate strategy must be employed to identify its targets. RESULTS: We expressed physiological levels of a tagged-p110 CUX1 protein and performed chromatin affinity purification followed by hybridization on ENCODE and promoter arrays. Targets were validated by chromatin immunoprecipitation and transcriptional regulation by CUX1 was analyzed in expression profiling and RT-qPCR assays following CUX1 knockdown or p110 CUX1 overexpression. Approximately 47% and 14% of CUX1 binding sites were respectively mapped less than 4 Kbp, or more than 40 Kbp, away from a transcription start site. More genes exhibited changes in expression following CUX1 knockdown than p110 CUX1 overexpression. CUX1 directly activated or repressed 7.4% and 8.4% of putative targets identified on the ENCODE and promoter arrays respectively. This proportion increased to 11.2% for targets with 2 binding sites or more. Transcriptional repression was observed in a slightly higher proportion of target genes. The CUX1 consensus binding motif, ATCRAT, was found at 47.2% of the CUX1 binding sites, yet only 8.3% of the CUX1 consensus motifs present on the array were bound in vivo. The presence of a consensus binding motif did not have an impact on whether a target gene was repressed or activated. Interestingly, the distance between a binding site and a transcription start site did not significantly reduced the ability of CUX1 to regulate a target gene. Moreover, CUX1 not only was able to regulate the next adjacent gene, but also regulated the gene located beyond this one as well as the gene located further away in the opposite direction. CONCLUSION: Our results demonstrate that p110 CUX1 can activate or repress transcription when bound at a distance and can regulate more than one gene on certain genomic loci.


Subject(s)
Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Binding Sites/genetics , Chromatin Immunoprecipitation , Female , Gene Knockdown Techniques , Humans , Protein Isoforms/metabolism , Tissue Array Analysis , Transcription Factors , Transcription Initiation Site , Transcriptional Activation/physiology
19.
Nucleic Acids Res ; 40(10): 4483-95, 2012 May.
Article in English | MEDLINE | ID: mdl-22319212

ABSTRACT

The p110 Cut homeobox 1 (CUX1) transcription factor regulates genes involved in DNA replication and chromosome segregation. Using a genome-wide-approach, we now demonstrate that CUX1 also modulates the constitutive expression of DNA damage response genes, including ones encoding ATM and ATR, as well as proteins involved in DNA damage-induced activation of, and signaling through, these kinases. Consistently, RNAi knockdown or genetic inactivation of CUX1 reduced ATM/ATR expression and negatively impacted hallmark protective responses mediated by ATM and ATR following exposure to ionizing radiation (IR) and UV, respectively. Specifically, abrogation of CUX1 strongly reduced ATM autophosphorylation after IR, in turn causing substantial decreases in (i) levels of phospho-Chk2 and p53, (ii) γ-H2AX and Rad51 DNA damage foci and (iii) the efficiency of DNA strand break repair. Similarly remarkable reductions in ATR-dependent responses, including phosphorylation of Chk1 and H2AX, were observed post-UV. Finally, multiple cell cycle checkpoints and clonogenic survival were compromised in CUX1 knockdown cells. Our results indicate that CUX1 regulates a transcriptional program that is necessary to mount an efficient response to mutagenic insult. Thus, CUX1 ensures not only the proper duplication and segregation of the genetic material, but also the preservation of its integrity.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Homeodomain Proteins/physiology , Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/physiology , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Checkpoints , Cell Survival , Cells, Cultured , DNA Breaks , DNA Repair/genetics , Gene Expression Regulation , Genomic Instability , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Mice , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Interference , Rad51 Recombinase/analysis , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Transcription, Genetic
20.
Nat Commun ; 2: 338, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21654638

ABSTRACT

α-fetoprotein (AFP) is not only a widely used biomarker in hepatocellular carcinoma (HCC) surveillance, but is also clinically recognized as linked with aggressive tumour behaviour. Here we show that deregulation of microRNA122, a liver-specific microRNA, is a cause of both AFP elevation and a more biologically aggressive phenotype in HCC. We identify CUX1, a direct target of microRNA122, as a common central mediator of these two effects. Using liver tissues from transgenic mice in which microRNA122 is functionally silenced, an orthotopic xenograft tumour model, and human clinical samples, we further demonstrate that a microRNA122/CUX1/microRNA214/ZBTB20 pathway regulates AFP expression. We also show that the microRNA122/CUX1/RhoA pathway regulates the aggressive characteristics of tumours. We conclude that microRNA122 and associated signalling proteins may represent viable therapeutic targets, and that serum AFP levels in HCC patients may be a surrogate marker for deregulated intracellular microRNA122 signalling pathways in HCC tissues.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic/physiology , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Signal Transduction/genetics , alpha-Fetoproteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Mice , Mice, Transgenic , MicroRNAs/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL