32 research outputs found

    A genome-scale in vivo loss-of-function screen identifies Phf6 as a lineage-specific regulator of leukemia cell growth

    Get PDF
    We performed a genome-scale shRNA screen for modulators of B-cell leukemia progression in vivo. Results from this work revealed dramatic distinctions between the relative effects of shRNAs on the growth of tumor cells in culture versus in their native microenvironment. Specifically, we identified many “context-specific” regulators of leukemia development. These included the gene encoding the zinc finger protein Phf6. While inactivating mutations in PHF6 are commonly observed in human myeloid and T-cell malignancies, we found that Phf6 suppression in B-cell malignancies impairs tumor progression. Thus, Phf6 is a “lineage-specific” cancer gene that plays opposing roles in developmentally distinct hematopoietic malignancies.Massachusetts Institute of Technology. Department of Biology (Training Grant)National Cancer Institute (U.S.). Integrative Cancer Biology Program (U54-CA112967-06)National Institutes of Health (U.S.) (RO1-CA128803-05

    The creatine kinase pathway is a metabolic vulnerability in EVI1-positive acute myeloid leukemia

    Get PDF
    Expression of the MECOM (also known as EVI1) proto-oncogene is deregulated by chromosomal translocations in some cases of acute myeloid leukemia (AML) and is associated with poor clinical outcome. Here, through transcriptomic and metabolomic profiling of hematopoietic cells, we reveal that EVI1 overexpression alters cellular metabolism. A screen using pooled short hairpin RNAs (shRNAs) identified the ATP-buffering, mitochondrial creatine kinase CKMT1 as necessary for survival of EVI1-expressing cells in subjects with EVI1-positive AML. EVI1 promotes CKMT1 expression by repressing the myeloid differentiation regulator RUNX1. Suppression of arginine-creatine metabolism by CKMT1-directed shRNAs or by the small molecule cyclocreatine selectively decreased the viability, promoted the cell cycle arrest and apoptosis of human EVI1-positive cell lines, and prolonged survival in both orthotopic xenograft models and mouse models of primary AML. CKMT1 inhibition altered mitochondrial respiration and ATP production, an effect that was abrogated by phosphocreatine-mediated reactivation of the arginine-creatine pathway. Targeting CKMT1 is thus a promising therapeutic strategy for this EVI1-driven AML subtype that is highly resistant to current treatment regimens. Keywords: AML; RUNX1; CKMT1; cyclocreatine; arginine metabolismNational Cancer Institute (U.S.) (NIH 1R35 CA210030-01)Stand Up To CancerBridge ProjectNational Cancer Institute (U.S.) (David H. Koch Institute for Integrative Cancer Research at MIT. Grant P30-CA14051

    BCL-B (BCL2L10) is overexpressed in patients suffering from multiple myeloma (MM) and drives an MM-like disease in transgenic mice

    No full text
    Multiple myeloma (MM) evolves from a premalignant condition known as monoclonal gammopathy of undetermined significance (MGUS). However, the factors underlying the malignant transformation of plasmocytes in MM are not fully characterized. We report here that Eµ-directed expression of the antiapoptotic Bcl-B protein in mice drives an MM phenotype that reproduces accurately the human disease. Indeed, with age, Eµ-bcl-b transgenic mice develop the characteristic features of human MM, including bone malignant plasma cell infiltration, a monoclonal immunoglobulin peak, immunoglobulin deposit in renal tubules, and highly characteristic bone lytic lesions. In addition, the tumors are serially transplantable in irradiated wild-type mice, underlying the tumoral origin of the disease. Eµ-bcl-b plasmocytes show increased expression of a panel of genes known to be dysregulated in human MM pathogenesis. Treatment of Eµ-bcl-b mice with drugs currently used to treat patients such as melphalan and VELCADE efficiently kills malignant plasmocytes in vivo. Finally, we find that Bcl-B is overexpressed in plasmocytes from MM patients but neither in MGUS patients nor in healthy individuals, suggesting that Bcl-B may drive MM. These findings suggest that Bcl-B could be an important factor in MM disease and pinpoint Eµ-bcl-b mice as a pertinent model to validate new therapies in MM.Ligue nationale contre le cancer (France) (Equipe Labellisée Grant R08001AA)Fondation de France (Grant R08080AA)Fondation ARC pour la Recherche sur le Cancer (Grant PGA120140200777)Fondation ARC pour la Recherche sur le Cancer (Project 2015–2016)French Research National Agency (Grant ANR-11-LABX-0028-01

    Calpain 2-dependent IκBα degradation mediates CPT-11 secondary resistance in colorectal cancer xenografts

    Get PDF
    International audienceCPT-11 (irinotecan), the first-line chemotherapy for advanced stage colorectal cancer, remains inactive in about half of patients (primary chemoresistance) and almost all initial responders develop secondary resistance after several courses of treatment (8 months on average). Nude mice bearing HT-29 colon cancer xenografts were treated with CPT-11 and/or an NF-κB inhibitor for two courses. We confirm that NF-κB inhibition potentiated CPT-11 anti-tumoural effect after the first course of treatment. However, tumours grew again at the end of the second course of treatment, generating resistant tumours. We observed an increase in the basal NF-κB activation in resistant tumours and in two resistant sublines, either obtained from resistant HT-29 tumours (HT-29R cells) or generated in vitro (RSN cells). The decrease of NF-κB activation in HT-29R and RSN cells by stable transfections with the super-repressor form of IκBα augmented their sensitivity to CPT-11. Comparing gene expression profiles of HT-29 and HT-29R cells, we identified the S100A10/Annexin A2 complex and calpain 2 as over-expressed potential NF-κB inducers. SiRNA silencing of calpain 2 but not of S100A10 and/or annexin A2, resulted in a decrease in NF-κB activation, an increase in cellular levels of IκBα and a partial restoration of the CPT-11 sensitivity in both HT-29R and RSN cells, suggesting that calpain 2-dependent IκBα degradation mediates CPT-11 secondary resistance. Thus, targeted therapies directed against calpain 2 may represent a novel strategy to enhance the anti-cancer efficacy of CPT-11.</p

    Spleen tyrosine kinase functions as a tumor suppressor in melanoma cells by inducing senescence-like growth arrest.: Syk activates p53 signaling pathway

    Get PDF
    International audienceLoss of tumor-suppressive pathways that control cellular senescence is a crucial step in malignant transformation. Spleen tyrosine kinase (Syk) is a cytoplasmic tyrosine kinase that has been recently implicated in tumor suppression of melanoma, a deadly skin cancer derived from pigment-producing melanocytes. However, the mechanism by which Syk suppresses melanoma growth remains unclear. Here, we report that reexpression of Syk in melanoma cells induces a p53-dependent expression of the cyclin-dependent kinase (cdk) inhibitor p21 and a senescence program. We first observed that Syk expression is lost in a subset of melanoma cell lines, primarily by DNA methylation-mediated gene silencing and restored after treatment with the demethylating agent 5-aza-2-deoxycytidine. We analyzed the significance of epigenetic inactivation of Syk and found that reintroduction of Syk in melanoma cells dramatically reduces clonogenic survival and three-dimensional tumor spheroid growth and invasion. Remarkably, melanoma cells reexpressing Syk display hallmarks of senescent cells, including reduction of proliferative activity and DNA synthesis, large and flattened morphology, senescence-associated beta-galactosidase activity, and heterochromatic foci. This phenotype is accompanied by hypophosphorylated retinoblastoma protein (Rb) and accumulation of p21, which depends on functional p53. Our results highlight a new role for Syk tyrosine kinase in regulating cellular senescence and identify Syk-mediated senescence as a novel tumor suppressor pathway the inactivation of which may contribute to melanoma tumorigenicity

    Targeting MTHFD2 in acute myeloid leukemia

    Get PDF
    Drugs targeting metabolism have formed the backbone of therapy for some cancers. We sought to identify new such targets in acute myeloid leukemia (AML). The one-carbon folate pathway, specifically methylenetetrahydrofolate dehydrogenase-cyclohydrolase 2 (MTHFD2), emerged as a top candidate in our analyses. MTHFD2 is the most differentially expressed metabolic enzyme in cancer versus normal cells. Knockdown of MTHFD2 in AML cells decreased growth, induced differentiation, and impaired colony formation in primary AML blasts. In human xenograft and MLL-AF9 mouse leukemia models, MTHFD2 suppression decreased leukemia burden and prolonged survival. Based upon primary patient AML data and functional genomic screening, we determined that FLT3-ITD is a biomarker of response to MTHFD2 suppression. Mechanistically, MYC regulates the expression of MTHFD2, and MTHFD2 knockdown suppresses the TCA cycle. This study supports the therapeutic targeting of MTHFD2 in AML. It has been known for decades that cancer cells have an altered metabolism. As early as the 1920s, Otto Warburg observed that tumor cells consume glucose at a high rate and undergo fermentation even in the presence of oxygen (Warburg et al., 1927). Since then, drugs targeting metabolism have transformed the treatment of certain cancers. In the 1940s, the discovery and application of aminopterin, which was later found to target dihydrofolate reductase (DHFR), a cytoplasmic enzyme involved in one-carbon folate metabolism, yielded the first remission in a child with acute lymphoblastic leukemia (Farber et al., 1948). Other folate derivatives, such as methotrexate, were later developed. More recently, drugs such as 5-fluorouracil and pemetrexed that target thymidylate synthetase, another enzyme involved in one-carbon folate metabolism, were found to be effective therapies for some cancers (Locasale, 2013). The discovery of germline and somatic mutations that alter metabolic proteins in cancer further supports the role of altered metabolism in cancer pathogenesis. Mutations in genes of the succinate dehydrogenase complex, critical for both the tricarboxylic acid (TCA) cycle and electron transport chain, have been implicated in the pathogenesis of hereditary paragangliomas (Baysal et al., 2000; Niemann and Müller, 2000), pheochromocytomas (Astuti et al., 2001), renal cell cancer (Vanharanta et al., 2004), and gastrointestinal stromal tumors (Janeway et al., 2011; Pantaleo et al., 2011). In addition, mutations in isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) have been found in subsets of gliomas (Yan et al., 2009; Brennan et al., 2013) and acute myeloid leukemia (AML; Paschka et al., 2010; Cancer Genome Atlas Research Network, 2013), among other malignancies. Drugs targeting these mutant proteins have entered the clinic with some successes in early phase trials (Stein et al. 2014. 56th Annual American Hematoligical Society Annual Meeting and Exposition. Abstract 115.). Moreover, as understanding of the metabolic derangements necessary to promote and maintain the malignant state continues to expand, so does the list of potential drug targets. For example, aerobic glycolysis is thought to enable the generation of the nucleotides, proteins, and lipids necessary to maintain the malignant proliferative state, in part through regulation of the glycolytic enzyme pyruvate kinase (Vander Heiden et al., 2010). Additionally, the discovery of the critical importance of glycine and serine in cancer metabolism has led to a resurgence in interest in better understanding the mechanistic relevance of one-carbon folate metabolism (Jain et al., 2012; Zhang et al., 2012; Labuschagne et al., 2014; Ye et al., 2014; Kim et al., 2015; Maddocks et al., 2016). Although drugs targeting metabolism, such as methotrexate and asparaginase (a drug that reduces the availability of asparagine and glutamine), have been critical for the treatment of acute lymphoblastic leukemia, they are not used in therapy for AML, a hematopoietic malignancy where cure rates are still quite poor despite high-dose cytotoxic chemotherapy, including stem cell transplantation. This is especially true for patients with subtypes of AML characterized by high-risk features, such as the presence of FLT3-ITD mutations. New therapies are urgently needed for the treatment of these patients. In this study, we set out to define common mechanisms critical to the maintenance of AML cells to nominate novel, potentially targetable metabolic pathways for the treatment of this disease. We integrated gene expression signatures generated from the treatment of AML cells with multiple small molecules known to promote AML differentiation and death. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), an NAD+-dependent enzyme with dehydrogenase and cyclohydrolase activity, which plays an essential role in mitochondrial one-carbon folate metabolism, was prioritized as a target relevant to AML cell growth and differentiation. Suppression of MTHFD2 impaired AML growth and induced differentiation in vitro and impaired disease progression in multiple mouse models of AML. Additionally, FLT3-ITD mutations are a biomarker of response to MTHFD2 suppression. Mechanistically, MYC directly regulates MTHFD2 expression, and suppression of MTHFD2 leads to marked alteration of the TCA cycle.National Cancer Institute (U.S.) (R01 CA140292)National Cancer Institute (U.S.) (R21 CA198028)National Heart, Lung, and Blood Institute (5T32 HL07574-32)Eunice Kennedy Shriver National Institute of Child Health and Human Development (U.S.) (5K12HD052896-09)Library of Integrated Cellular Signatures (U54HG006093)Library of Integrated Cellular Signatures (U54HL127366
    corecore