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










Publication year range
1.
Blood ; 143(15): 1513-1527, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38096371

ABSTRACT

ABSTRACT: Small molecules that target the menin-KMT2A protein-protein interaction (menin inhibitors) have recently entered clinical trials in lysine methyltransferase 2A (KMT2A or MLL1)-rearranged (KMT2A-r) and nucleophosmin-mutant (NPM1c) acute myeloid leukemia (AML) and are demonstrating encouraging results. However, rationally chosen combination therapy is needed to improve responses and prevent resistance. We have previously identified IKZF1/IKAROS as a target in KMT2A-r AML and shown in preclinical models that IKAROS protein degradation with lenalidomide or iberdomide has modest single-agent activity yet can synergize with menin inhibitors. Recently, the novel IKAROS degrader mezigdomide was developed with greatly enhanced IKAROS protein degradation. In this study, we show that mezigdomide has increased preclinical activity in vitro as a single-agent in KMT2A-r and NPM1c AML cell lines, including sensitivity in cell lines resistant to lenalidomide and iberdomide. Further, we demonstrate that mezigdomide has the greatest capacity to synergize with and induce apoptosis in combination with menin inhibitors, including in MEN1 mutant models. We show that the superior activity of mezigdomide compared with lenalidomide or iberdomide is due to its increased depth, rate, and duration of IKAROS protein degradation. Single-agent mezigdomide was efficacious in 5 patient-derived xenograft models of KMT2A-r and 1 NPM1c AML. The combination of mezigdomide with the menin inhibitor VTP-50469 increased survival and prevented and overcame MEN1 mutations that mediate resistance in patients receiving menin inhibitor monotherapy. These results support prioritization of mezigdomide for early phase clinical trials in KMT2A-r and NPM1c AML, either as a single agent or in combination with menin inhibitors.


Subject(s)
Leukemia, Myeloid, Acute , Morpholines , Myeloid-Lymphoid Leukemia Protein , Phthalimides , Piperidones , Humans , Lenalidomide/therapeutic use , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Transcription Factors/genetics , Mutation
2.
Cell Death Differ ; 31(2): 150-158, 2024 02.
Article in English | MEDLINE | ID: mdl-38097622

ABSTRACT

Whole-genome screens using CRISPR technologies are powerful tools to identify novel tumour suppressors as well as factors that impact responses of malignant cells to anti-cancer agents. Applying this methodology to lymphoma cells, we conducted a genome-wide screen to identify novel inhibitors of tumour expansion that are induced by the tumour suppressor TRP53. We discovered that the absence of Arrestin domain containing 3 (ARRDC3) increases the survival and long-term competitiveness of MYC-driven lymphoma cells when treated with anti-cancer agents that activate TRP53. Deleting Arrdc3 in mice caused perinatal lethality due to various developmental abnormalities, including cardiac defects. Notably, the absence of ARRDC3 markedly accelerated MYC-driven lymphoma development. Thus, ARRDC3 is a new mediator of TRP53-mediated suppression of tumour expansion, and this discovery may open new avenues to harness this process for cancer therapy.


Subject(s)
Lymphoma , Neoplasms , Animals , Mice , Arrestins/genetics , Arrestins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Neoplasms/genetics
4.
Clin Lymphoma Myeloma Leuk ; 22(12): 869-877, 2022 12.
Article in English | MEDLINE | ID: mdl-36030175

ABSTRACT

Higher-risk myelodysplastic syndromes (MDS) carry a dismal prognosis with rapid disease progression, disease-related complications that impact quality of life, high risk of transformation to acute myeloid leukemia (AML), and poor long-term survival. Higher-risk disease is determined by a number of factors including the depth and type of cytopenias, percentage of myeloblasts occupying the bone marrow, cytogenetic abnormalities, and increasingly also by the presence of higher-risk molecular alterations. In addition to disease characteristics, a patient's performance status and degree of co-morbidity strongly influence treatment decisions and clinical outcomes. A critical first step in the management of patients with higher-risk MDS is evaluating eligibility for allogeneic hematopoietic stem cell transplant (HCT), which currently remains the only curative therapy, and is available to an ever-increasing number of patients. Outside of stem cell transplant, treatment with hypomethylating agent chemotherapy, azacitidine or decitabine, remains the cornerstone of therapy with improvements in overall survival and reduced transformation to AML; however, these approaches are palliative in nature and outcomes remain very poor overall. With a deepening understanding of disease pathophysiology has come a burgeoning array of novel targeted therapies that are currently in pre-clinical and early phase clinical trials offering hope for new treatment options for this malignancy.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Myelodysplastic Syndromes/therapy , Myelodysplastic Syndromes/drug therapy , Quality of Life , Azacitidine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy
5.
Nat Cancer ; 3(5): 595-613, 2022 05.
Article in English | MEDLINE | ID: mdl-35534777

ABSTRACT

Acute myeloid leukemia (AML) remains difficult to treat and requires new therapeutic approaches. Potent inhibitors of the chromatin-associated protein MENIN have recently entered human clinical trials, opening new therapeutic opportunities for some genetic subtypes of this disease. Using genome-scale functional genetic screens, we identified IKAROS (encoded by IKZF1) as an essential transcription factor in KMT2A (MLL1)-rearranged (MLL-r) AML that maintains leukemogenic gene expression while also repressing pathways for tumor suppression, immune regulation and cellular differentiation. Furthermore, IKAROS displays an unexpected functional cooperativity and extensive chromatin co-occupancy with mixed lineage leukemia (MLL)1-MENIN and the regulator MEIS1 and an extensive hematopoietic transcriptional complex involving homeobox (HOX)A10, MEIS1 and IKAROS. This dependency could be therapeutically exploited by inducing IKAROS protein degradation with immunomodulatory imide drugs (IMiDs). Finally, we demonstrate that combined IKAROS degradation and MENIN inhibition effectively disrupts leukemogenic transcriptional networks, resulting in synergistic killing of leukemia cells and providing a paradigm for improved drug targeting of transcription and an opportunity for rapid clinical translation.


Subject(s)
Leukemia, Myeloid, Acute , Chromatin , Gene Expression , Humans , Ikaros Transcription Factor/metabolism , Leukemia, Myeloid, Acute/drug therapy , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Transcription Factors/genetics
6.
Cancer Discov ; 12(7): 1804-1823, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35499757

ABSTRACT

Gastrointestinal stromal tumor (GIST) is commonly characterized by activating mutations in the receptor tyrosine kinase KIT. Tyrosine kinase inhibitors are the only approved therapy for GIST, and complementary treatment strategies are urgently needed. As GIST lacks oncogene amplification and relies upon an established network of transcription factors, we hypothesized that unique chromatin-modifying enzymes are essential in orchestrating the GIST epigenome. We identified through genome-scale CRISPR screening that MOZ and Menin-MLL chromatin regulatory complexes are cooperative and unique dependencies in GIST. These complexes were enriched at GIST-relevant genes and regulated their transcription. Inhibition of MOZ and Menin-MLL complexes decreased GIST cell proliferation by disrupting interactions with transcriptional/chromatin regulators, such as DOT1L. MOZ and Menin inhibition caused significant reductions in tumor burden in vivo, with superior effects observed with combined Menin and KIT inhibition. These results define unique chromatin regulatory dependencies in GIST and identify potential therapeutic strategies for clinical application. SIGNIFICANCE: Although many malignancies rely on oncogene amplification, GIST instead depends upon epigenetic regulation of KIT and other essential genes. Utilizing genome-scale CRISPR dependency screens, we identified complementary chromatin-modifying complexes essential to GIST and characterize the consequences of their disruption, elucidating a novel therapeutic approach to this disease. This article is highlighted in the In This Issue feature, p. 1599.


Subject(s)
Gastrointestinal Neoplasms , Gastrointestinal Stromal Tumors , Histone Acetyltransferases/metabolism , Chromatin/genetics , Epigenesis, Genetic , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Humans , Proto-Oncogene Proteins c-kit/genetics , Receptor Protein-Tyrosine Kinases/genetics , Transcription Factors/genetics
8.
Blood ; 137(20): 2721-2735, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33824975

ABSTRACT

Selective targeting of BCL-2 with the BH3-mimetic venetoclax has been a transformative treatment for patients with various leukemias. TP-53 controls apoptosis upstream of where BCL-2 and its prosurvival relatives, such as MCL-1, act. Therefore, targeting these prosurvival proteins could trigger apoptosis across diverse blood cancers, irrespective of TP53 mutation status. Indeed, targeting BCL-2 has produced clinically relevant responses in blood cancers with aberrant TP-53. However, in our study, TP53-mutated or -deficient myeloid and lymphoid leukemias outcompeted isogenic controls with intact TP-53, unless sufficient concentrations of BH3-mimetics targeting BCL-2 or MCL-1 were applied. Strikingly, tumor cells with TP-53 dysfunction escaped and thrived over time if inhibition of BCL-2 or MCL-1 was sublethal, in part because of an increased threshold for BAX/BAK activation in these cells. Our study revealed the key role of TP-53 in shaping long-term responses to BH3-mimetic drugs and reconciled the disparate pattern of initial clinical response to venetoclax, followed by subsequent treatment failure among patients with TP53-mutant chronic lymphocytic leukemia or acute myeloid leukemia. In contrast to BH3-mimetics targeting just BCL-2 or MCL-1 at doses that are individually sublethal, a combined BH3-mimetic approach targeting both prosurvival proteins enhanced lethality and durably suppressed the leukemia burden, regardless of TP53 mutation status. Our findings highlight the importance of using sufficiently lethal treatment strategies to maximize outcomes of patients with TP53-mutant disease. In addition, our findings caution against use of sublethal BH3-mimetic drug regimens that may enhance the risk of disease progression driven by emergent TP53-mutant clones.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Indolizines/pharmacology , Isoquinolines/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Morpholines/pharmacology , Neoplasm Proteins/physiology , Peptide Fragments/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/physiology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Apoptosis Regulatory Proteins/physiology , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , CRISPR-Cas Systems , Cell Line, Tumor , DNA Damage , Genes, p53 , Humans , Indolizines/therapeutic use , Interleukin-2 Receptor alpha Subunit/deficiency , Isoquinolines/therapeutic use , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Morpholines/therapeutic use , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Oxidative Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use , Tumor Suppressor Protein p53/deficiency , Xenograft Model Antitumor Assays
9.
Blood ; 133(16): 1729-1741, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30755422

ABSTRACT

Somatically acquired mutations in PHF6 (plant homeodomain finger 6) frequently occur in hematopoietic malignancies and often coincide with ectopic expression of TLX3. However, there is no functional evidence to demonstrate whether these mutations contribute to tumorigenesis. Similarly, the role of PHF6 in hematopoiesis is unknown. We report here that Phf6 deletion in mice resulted in a reduced number of hematopoietic stem cells (HSCs), an increased number of hematopoietic progenitor cells, and an increased proportion of cycling stem and progenitor cells. Loss of PHF6 caused increased and sustained hematopoietic reconstitution in serial transplantation experiments. Interferon-stimulated gene expression was upregulated in the absence of PHF6 in hematopoietic stem and progenitor cells. The numbers of hematopoietic progenitor cells and cycling hematopoietic stem and progenitor cells were restored to normal by combined loss of PHF6 and the interferon α and ß receptor subunit 1. Ectopic expression of TLX3 alone caused partially penetrant leukemia. TLX3 expression and loss of PHF6 combined caused fully penetrant early-onset leukemia. Our data suggest that PHF6 is a hematopoietic tumor suppressor and is important for fine-tuning hematopoietic stem and progenitor cell homeostasis.


Subject(s)
Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Leukemia/etiology , Repressor Proteins/physiology , Animals , Carcinogenesis , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Receptors, Interferon , Repressor Proteins/genetics , Tumor Suppressor Proteins
10.
Genes Dev ; 32(21-22): 1420-1429, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30366906

ABSTRACT

Mutations in Trp53, prevalent in human cancer, are reported to drive tumorigenesis through dominant-negative effects (DNEs) over wild-type TRP53 function as well as neomorphic gain-of-function (GOF) activity. We show that five TRP53 mutants do not accelerate lymphomagenesis on a TRP53-deficient background but strongly synergize with c-MYC overexpression in a manner that distinguishes the hot spot Trp53 mutations. RNA sequencing revealed that the mutant TRP53 DNE does not globally repress wild-type TRP53 function but disproportionately impacts a subset of wild-type TRP53 target genes. Accordingly, TRP53 mutant proteins impair pathways for DNA repair, proliferation, and metabolism in premalignant cells. This reveals that, in our studies of lymphomagenesis, mutant TRP53 drives tumorigenesis primarily through the DNE, which modulates wild-type TRP53 function in a manner advantageous for neoplastic transformation.


Subject(s)
Carcinogenesis/genetics , Mutation , Tumor Suppressor Protein p53/genetics , Animals , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Suppressor Protein p53/metabolism
11.
Nature ; 560(7717): 253-257, 2018 08.
Article in English | MEDLINE | ID: mdl-30069049

ABSTRACT

Acetylation of histones by lysine acetyltransferases (KATs) is essential for chromatin organization and function1. Among the genes coding for the MYST family of KATs (KAT5-KAT8) are the oncogenes KAT6A (also known as MOZ) and KAT6B (also known as MORF and QKF)2,3. KAT6A has essential roles in normal haematopoietic stem cells4-6 and is the target of recurrent chromosomal translocations, causing acute myeloid leukaemia7,8. Similarly, chromosomal translocations in KAT6B have been identified in diverse cancers8. KAT6A suppresses cellular senescence through the regulation of suppressors of the CDKN2A locus9,10, a function that requires its KAT activity10. Loss of one allele of KAT6A extends the median survival of mice with MYC-induced lymphoma from 105 to 413 days11. These findings suggest that inhibition of KAT6A and KAT6B may provide a therapeutic benefit in cancer. Here we present highly potent, selective inhibitors of KAT6A and KAT6B, denoted WM-8014 and WM-1119. Biochemical and structural studies demonstrate that these compounds are reversible competitors of acetyl coenzyme A and inhibit MYST-catalysed histone acetylation. WM-8014 and WM-1119 induce cell cycle exit and cellular senescence without causing DNA damage. Senescence is INK4A/ARF-dependent and is accompanied by changes in gene expression that are typical of loss of KAT6A function. WM-8014 potentiates oncogene-induced senescence in vitro and in a zebrafish model of hepatocellular carcinoma. WM-1119, which has increased bioavailability, arrests the progression of lymphoma in mice. We anticipate that this class of inhibitors will help to accelerate the development of therapeutics that target gene transcription regulated by histone acetylation.


Subject(s)
Benzenesulfonates/pharmacology , Cellular Senescence/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Hydrazines/pharmacology , Lymphoma/drug therapy , Lymphoma/pathology , Sulfonamides/pharmacology , Acetylation/drug effects , Animals , Benzenesulfonates/therapeutic use , Cell Proliferation/drug effects , Cells, Cultured , Drug Development , Fibroblasts , Gene Expression Regulation, Neoplastic/drug effects , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histones/chemistry , Histones/metabolism , Hydrazines/therapeutic use , Lymphoma/enzymology , Lymphoma/genetics , Lysine/chemistry , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Sulfonamides/therapeutic use
12.
Cell Death Differ ; 25(1): 104-113, 2018 01.
Article in English | MEDLINE | ID: mdl-29149101

ABSTRACT

The tumour suppressor gene TP53 is mutated in ~50% of human cancers. In addition to its function in tumour suppression, p53 also plays a major role in the response of malignant as well as nontransformed cells to many anticancer therapeutics, particularly those that cause DNA damage. P53 forms a homotetrameric transcription factor that is reported to directly regulate ~500 target genes, thereby controlling a broad range of cellular processes, including cell cycle arrest, cell senescence, DNA repair, metabolic adaptation and cell death. For a long time, induction of apoptotic death in nascent neoplastic cells was regarded as the principal mechanism by which p53 prevents tumour development. This concept has, however, recently been challenged by the findings that in striking contrast to Trp53-deficient mice, gene-targeted mice that lack the critical effectors of p53-induced apoptosis do not develop tumours spontaneously. Remarkably, even mice lacking all mediators critical for p53-induced apoptosis, G1/S boundary cell cycle arrest and cell senescence do not develop any tumours spontaneously. In this review we discuss current understanding of the mechanisms by which p53 induces cell death and how this affects p53-mediated tumour suppression and the response of malignant cells to anticancer therapy.


Subject(s)
Apoptosis , Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Humans , Mice , Mutation , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
13.
Cell Death Dis ; 8(7): e2914, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28682312

ABSTRACT

Anaemia is a major global health problem arising from diverse causes and for which improved therapeutic strategies are needed. Erythroid cells can undergo apoptotic cell death and loss of pro-survival BCL-XL is known to trigger apoptosis during late-stage erythroid development. However, the mechanism by which loss or pharmacological blockade of BCL-XL leads to erythroid cell apoptosis remains unclear. Here we sought to identify the precise stage of erythropoiesis that depends on BCL-XL. We also tested whether deficiency of BIM or PUMA, the two main pro-apoptotic antagonists of BCL-XL, could prevent reticulocyte death and anaemia caused by BCL-XL loss. Using an in vivo mouse model of tamoxifen-inducible Bclx gene deletion and in vitro assays with a BCL-XL-selective inhibitor, we interrogated each stage of erythrocyte differentiation for BCL-XL dependency. This revealed that reticulocytes, but not orthochromatic erythroblasts, require BCL-XL for their survival. Surprisingly, concurrent loss of BIM or PUMA had no significant impact on the development of anemia following acute BCL-XL deletion in vivo. However, analysis of mixed bone marrow chimaeric mice revealed that loss of PUMA, but not loss of BIM, partially alleviated impaired erythropoiesis caused by BCL-XL deficiency. Insight into how the network of pro-survival and pro-apoptotic proteins works will assist the development of strategies to mitigate the effects of abnormal cell death during erythropoiesis and prevent anaemia in patients treated with BCL-XL-specific BH3-mimetic drugs.


Subject(s)
Anemia/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Bcl-2-Like Protein 11/genetics , Gene Expression Regulation , Tumor Suppressor Proteins/genetics , bcl-X Protein/genetics , Anemia/metabolism , Anemia/pathology , Animals , Apoptosis Regulatory Proteins/deficiency , Bcl-2-Like Protein 11/deficiency , Benzothiazoles/pharmacology , Cell Survival , Disease Models, Animal , Erythroblasts/metabolism , Erythroblasts/pathology , Erythropoiesis/genetics , Gene Deletion , Humans , Isoquinolines/pharmacology , Mice , Mice, Knockout , Organ Specificity , Protein Domains , Reticulocytes/metabolism , Reticulocytes/pathology , Signal Transduction , Tamoxifen/pharmacology , Tumor Suppressor Proteins/deficiency , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/deficiency
14.
Nature ; 538(7626): 477-482, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27760111

ABSTRACT

Avoidance of apoptosis is critical for the development and sustained growth of tumours. The pro-survival protein myeloid cell leukemia 1 (MCL1) is overexpressed in many cancers, but the development of small molecules targeting this protein that are amenable for clinical testing has been challenging. Here we describe S63845, a small molecule that specifically binds with high affinity to the BH3-binding groove of MCL1. Our mechanistic studies demonstrate that S63845 potently kills MCL1-dependent cancer cells, including multiple myeloma, leukaemia and lymphoma cells, by activating the BAX/BAK-dependent mitochondrial apoptotic pathway. In vivo, S63845 shows potent anti-tumour activity with an acceptable safety margin as a single agent in several cancers. Moreover, MCL1 inhibition, either alone or in combination with other anti-cancer drugs, proved effective against several solid cancer-derived cell lines. These results point towards MCL1 as a target for the treatment of a wide range of tumours.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Models, Biological , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Thiophenes/pharmacology , Thiophenes/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans , Leukemia/drug therapy , Leukemia/metabolism , Leukemia/pathology , Lymphoma/drug therapy , Lymphoma/metabolism , Lymphoma/pathology , Male , Mice , Models, Molecular , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/metabolism , Pyrimidines/administration & dosage , Thiophenes/administration & dosage , Xenograft Model Antitumor Assays , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
15.
J Exp Med ; 213(10): 2039-48, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27621418

ABSTRACT

Neoplastic transformation is driven by oncogenic lesions that facilitate unrestrained cell expansion and resistance to antiproliferative signals. These oncogenic DNA lesions, acquired through errors in DNA replication, gene recombination, or extrinsically imposed damage, are thought to activate multiple tumor suppressive pathways, particularly apoptotic cell death. DNA damage induces apoptosis through well-described p53-mediated induction of PUMA and NOXA. However, loss of both these mediators (even together with defects in p53-mediated induction of cell cycle arrest and cell senescence) does not recapitulate the tumor susceptibility observed in p53(-/-) mice. Thus, potentially oncogenic DNA lesions are likely to also trigger apoptosis through additional, p53-independent processes. We found that loss of the BH3-only protein BIM accelerated lymphoma development in p53-deficient mice. This process was negated by concomitant loss of RAG1/2-mediated antigen receptor gene rearrangement. This demonstrates that BIM is critical for the induction of apoptosis caused by potentially oncogenic DNA lesions elicited by RAG1/2-induced gene rearrangement. Furthermore, this highlights the role of a BIM-mediated tumor suppressor pathway that acts in parallel to the p53 pathway and remains active even in the absence of wild-type p53 function, suggesting this may be exploited in the treatment of p53-deficient cancers.


Subject(s)
Apoptosis , Bcl-2-Like Protein 11/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Tumor Suppressor Protein p53/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/pathology , Heterozygote , Lymphoma/pathology , Mice, Inbred C57BL , Recombinases/metabolism , Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism
16.
Article in English | MEDLINE | ID: mdl-27141080

ABSTRACT

The fundamental biological importance of the Tp53 gene family is highlighted by its evolutionary conservation for more than one billion years dating back to the earliest multicellular organisms. The TP53 protein provides essential functions in the cellular response to diverse stresses and safeguards maintenance of genomic integrity, and this is manifest in its critical role in tumor suppression. The importance of Tp53 in tumor prevention is exemplified in human cancer where it is the most frequently detected genetic alteration. This is confirmed in animal models, in which a defective Tp53 gene leads inexorably to cancer development, whereas reinstatement of TP53 function results in regression of established tumors that had been initiated by loss of TP53. Remarkably, despite extensive investigation, the specific mechanisms by which TP53 acts as a tumor suppressor are yet to be fully defined. We review the history and current standing of efforts to understand these mechanisms and how they complement each other in tumor suppression.


Subject(s)
Neoplasms/therapy , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/therapeutic use , Genes, p53 , Genetic Therapy , Humans , Neoplasms/metabolism , Signal Transduction/drug effects
17.
Cell Rep ; 14(10): 2337-47, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947081

ABSTRACT

MCL-1 is critical for progenitor cell survival during emergency hematopoiesis, but its role in sustaining cells undergoing transformation and in lymphomagenesis is only poorly understood. We investigated the importance of MCL-1 in the survival of B lymphoid progenitors undergoing MYC-driven transformation and its functional interactions with pro-apoptotic BIM and PUMA and the tumor suppressor p53 in lymphoma development. Loss of one Mcl-1 allele almost abrogated MYC-driven-lymphoma development owing to a reduction in lymphoma initiating pre-B cells. Although loss of the p53 target PUMA had minor impact, loss of one p53 allele substantially accelerated lymphoma development when MCL-1 was limiting, most likely because p53 loss also causes defects in non-apoptotic tumor suppressive processes. Remarkably, loss of BIM restored the survival of lymphoma initiating cells and rate of tumor development. Thus, MCL-1 has a major role in lymphoma initiating pro-B cells to oppose BIM, which is upregulated in response to oncogenic stress.


Subject(s)
Apoptosis , Lymphoma/physiopathology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Precursor Cells, B-Lymphoid/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Alleles , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Bone Marrow/metabolism , Bone Marrow/pathology , Flow Cytometry , Genotype , Kaplan-Meier Estimate , Lymphoma/mortality , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Precursor Cells, B-Lymphoid/cytology , Proto-Oncogene Proteins c-myc/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
18.
Cell Rep ; 14(8): 1858-66, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26904937

ABSTRACT

Nutlin3a is a small-molecule antagonist of MDM2 that promotes non-genotoxic activation of p53 through p53 protein stabilization and transactivation of p53 target genes. Nutlin3a is the forerunner of a class of cancer therapeutics that have reached clinical trials. Using transgenic and gene-targeted mouse models lacking the critical p53 target genes, p21, Puma, and Noxa, we found that only loss of PUMA conferred profound protection against Nutlin3a-induced killing in both non-transformed lymphoid cells and Eµ-Myc lymphomas in vitro and in vivo. CRISPR/Cas9-mediated targeting of the PUMA gene rendered human hematopoietic cancer cell lines markedly resistant to Nutlin3a-induced cell death. These results demonstrate that PUMA-mediated apoptosis, but not p21-mediated cell-cycle arrest or senescence, is a critical determinant of the therapeutic response to non-genotoxic p53 activation by Nutlin3a. Importantly, in human cancer, PUMA expression may predict patient responses to treatment with MDM2 antagonists.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Gene Expression Regulation, Neoplastic , Imidazoles/pharmacology , Lymphoma/drug therapy , Piperazines/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , CRISPR-Cas Systems , Cell Cycle Checkpoints , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Humans , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , Tumor Suppressor Protein p53/agonists , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
19.
Cell Rep ; 10(8): 1422-32, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25732831

ABSTRACT

The CRISPR/Cas9 technology enables the introduction of genomic alterations into almost any organism; however, systems for efficient and inducible gene modification have been lacking, especially for deletion of essential genes. Here, we describe a drug-inducible small guide RNA (sgRNA) vector system allowing for ubiquitous and efficient gene deletion in murine and human cells. This system mediates the efficient, temporally controlled deletion of MCL-1, both in vitro and in vivo, in human Burkitt lymphoma cell lines that require this anti-apoptotic BCL-2 protein for sustained survival and growth. Unexpectedly, repeated induction of the same sgRNA generated similar inactivating mutations in the human Mcl-1 gene due to low mutation variability exerted by the accompanying non-homologous end-joining (NHEJ) process. Finally, we were able to generate hematopoietic cell compartment-restricted Trp53-knockout mice, leading to the identification of cancer-promoting mutants of this critical tumor suppressor.


Subject(s)
Genes, Essential/genetics , Genes, Neoplasm/genetics , Lentivirus/genetics , Neoplasms/genetics , RNA, Guide, Kinetoplastida/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Bcl-2-Like Protein 11 , Cell Line , Cell Survival/drug effects , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA End-Joining Repair , Doxycycline/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transplantation, Heterologous , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Genes Dev ; 28(1): 58-70, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24395247

ABSTRACT

The transcriptional regulator c-MYC is abnormally overexpressed in many human cancers. Evasion from apoptosis is critical for cancer development, particularly c-MYC-driven cancers. We explored which anti-apoptotic BCL-2 family member (expressed under endogenous regulation) is essential to sustain c-MYC-driven lymphoma growth to reveal which should be targeted for cancer therapy. Remarkably, inducible Cre-mediated deletion of even a single Mcl-1 allele substantially impaired the growth of c-MYC-driven mouse lymphomas. Mutations in p53 could diminish but not obviate the dependency of c-MYC-driven mouse lymphomas on MCL-1. Importantly, targeting of MCL-1 killed c-MYC-driven human Burkitt lymphoma cells, even those bearing mutations in p53. Given that loss of one allele of Mcl-1 is well tolerated in healthy tissues, our results suggest that therapeutic targeting of MCL-1 would be an attractive therapeutic strategy for MYC-driven cancers.


Subject(s)
Lymphoma/genetics , Lymphoma/therapy , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Gene Expression Regulation, Neoplastic , Humans , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Protein Binding , Proto-Oncogene Proteins c-myc/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...