Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Genes Dev ; 33(17-18): 1252-1264, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31395740

ABSTRACT

Although MAX is regarded as an obligate dimerization partner for MYC, its function in normal development and neoplasia is poorly defined. We show that B-cell-specific deletion of Max has a modest effect on B-cell development but completely abrogates Eµ-Myc-driven lymphomagenesis. While Max loss affects only a few hundred genes in normal B cells, it leads to the global down-regulation of Myc-activated genes in premalignant Eµ-Myc cells. We show that the balance between MYC-MAX and MNT-MAX interactions in B cells shifts in premalignant B cells toward a MYC-driven transcriptional program. Moreover, we found that MAX loss leads to a significant reduction in MYC protein levels and down-regulation of direct transcriptional targets, including regulators of MYC stability. This phenomenon is also observed in multiple cell lines treated with MYC-MAX dimerization inhibitors. Our work uncovers a layer of Myc autoregulation critical for lymphomagenesis yet partly dispensable for normal development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Lymphoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Active Transport, Cell Nucleus , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Kynurenine/genetics , Kynurenine/metabolism , Lymphoma/physiopathology , Mice , Organoids/growth & development , Organoids/physiopathology , Oximes/pharmacology , Sulfonamides/pharmacology
2.
PLoS Biol ; 19(10): e3001085, 2021 10.
Article in English | MEDLINE | ID: mdl-34669700

ABSTRACT

Male germ cell (GC) production is a metabolically driven and apoptosis-prone process. Here, we show that the glucose-sensing transcription factor (TF) MAX-Like protein X (MLX) and its binding partner MondoA are both required for male fertility in the mouse, as well as survival of human tumor cells derived from the male germ line. Loss of Mlx results in altered metabolism as well as activation of multiple stress pathways and GC apoptosis in the testes. This is concomitant with dysregulation of the expression of male-specific GC transcripts and proteins. Our genomic and functional analyses identify loci directly bound by MLX involved in these processes, including metabolic targets, obligate components of male-specific GC development, and apoptotic effectors. These in vivo and in vitro studies implicate MLX and other members of the proximal MYC network, such as MNT, in regulation of metabolism and differentiation, as well as in suppression of intrinsic and extrinsic death signaling pathways in both spermatogenesis and male germ cell tumors (MGCTs).


Subject(s)
Apoptosis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glucose/metabolism , Spermatogenesis , Stress, Physiological , Animals , Base Sequence , Cell Survival , Exons/genetics , Fertility , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation , Gene Targeting , Lipid Metabolism , Male , Mice, Knockout , Models, Biological , Neoplasms, Germ Cell and Embryonal/pathology , Principal Component Analysis , RNA/genetics , RNA/metabolism , Repressor Proteins/metabolism , Reproduction , Sertoli Cells/metabolism , Spermatogenesis/genetics , Spermatozoa/metabolism , Testicular Neoplasms/pathology , Testis/metabolism , Transcription Factors/metabolism , Transcription, Genetic
3.
Genes Dev ; 30(23): 2637-2648, 2016 12 01.
Article in English | MEDLINE | ID: mdl-28007786

ABSTRACT

Myc plays critical roles in the self-renewal division of various stem cell types. In spermatogonial stem cells (SSCs), Myc controls SSC fate decisions because Myc overexpression induces enhanced self-renewal division, while depletion of Max, a Myc-binding partner, leads to meiotic induction. However, the mechanism by which Myc acts on SSC fate is unclear. Here we demonstrate a critical link between Myc/Mycn gene activity and glycolysis in SSC self-renewal. In SSCs, Myc/Mycn are regulated by Foxo1, whose deficiency impairs SSC self-renewal. Myc/Mycn-deficient SSCs not only undergo limited self-renewal division but also display diminished glycolytic activity. While inhibition of glycolysis decreased SSC activity, chemical stimulation of glycolysis or transfection of active Akt1 or Pdpk1 (phosphoinositide-dependent protein kinase 1 ) augmented self-renewal division, and long-term SSC cultures were derived from a nonpermissive strain that showed limited self-renewal division. These results suggested that Myc-mediated glycolysis is an important factor that increases the frequency of SSC self-renewal division.


Subject(s)
Cell Self Renewal/genetics , Gene Expression Regulation, Developmental/genetics , Glycolysis/genetics , N-Myc Proto-Oncogene Protein/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Spermatogonia/cytology , Stem Cells/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Animals , Cell Division/genetics , Cell Proliferation/genetics , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , N-Myc Proto-Oncogene Protein/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA Splicing Factors/metabolism , Stem Cells/enzymology
4.
Genes Dev ; 30(11): 1289-99, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27298335

ABSTRACT

Small cell lung cancer (SCLC) is a devastating neuroendocrine carcinoma. MYCL (L-Myc) is frequently amplified in human SCLC, but its roles in SCLC progression are poorly understood. We isolated preneoplastic neuroendocrine cells from a mouse model of SCLC and found that ectopic expression of L-Myc, c-Myc, or N-Myc conferred tumor-forming capacity. We focused on L-Myc, which promoted pre-rRNA synthesis and transcriptional programs associated with ribosomal biogenesis. Deletion of Mycl in two genetically engineered models of SCLC resulted in strong suppression of SCLC. The high degree of suppression suggested that L-Myc may constitute a therapeutic target for a broad subset of SCLC. We then used an RNA polymerase I inhibitor to target rRNA synthesis in an autochthonous Rb/p53-deleted mouse SCLC model and found significant tumor inhibition. These data reveal that activation of RNA polymerase I by L-Myc and other MYC family proteins provides an axis of vulnerability for this recalcitrant cancer.


Subject(s)
Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Polymerase I/metabolism , Small Cell Lung Carcinoma/enzymology , Small Cell Lung Carcinoma/genetics , Animals , Animals, Genetically Modified , Benzothiazoles/pharmacology , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Silencing , Lung Neoplasms/physiopathology , Mice , Naphthyridines/pharmacology , Proto-Oncogene Proteins c-myc/genetics , RNA Polymerase I/antagonists & inhibitors , Ribosomes/metabolism , Small Cell Lung Carcinoma/physiopathology , Tumor Burden/drug effects , Tumor Cells, Cultured
5.
Genes Dev ; 29(23): 2475-89, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26584623

ABSTRACT

Metabolic stress and changes in nutrient levels modulate many aspects of skeletal muscle function during aging and disease. Growth factors and cytokines secreted by skeletal muscle, known as myokines, are important signaling factors, but it is largely unknown whether they modulate muscle growth and differentiation in response to nutrients. Here, we found that changes in glucose levels increase the activity of the glucose-responsive transcription factor MLX (Max-like protein X), which promotes and is necessary for myoblast fusion. MLX promotes myogenesis not via an adjustment of glucose metabolism but rather by inducing the expression of several myokines, including insulin-like growth factor 2 (IGF2), whereas RNAi and dominant-negative MLX reduce IGF2 expression and block myogenesis. This phenotype is rescued by conditioned medium from control muscle cells and by recombinant IGF2, which activates the myogenic kinase Akt. Importantly, MLX-null mice display decreased IGF2 induction and diminished muscle regeneration in response to injury, indicating that the myogenic function of MLX is manifested in vivo. Thus, glucose is a signaling molecule that regulates myogenesis and muscle regeneration via MLX/IGF2/Akt signaling.


Subject(s)
Muscle Development/genetics , Muscle, Skeletal/cytology , Nuclear Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Acetylation , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Line , Gene Expression Regulation, Developmental/genetics , Glucose/metabolism , Histones/metabolism , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/physiology , Regeneration
6.
Elife ; 102021 07 08.
Article in English | MEDLINE | ID: mdl-34236315

ABSTRACT

MGA, a transcription factor and member of the MYC network, is mutated or deleted in a broad spectrum of malignancies. As a critical test of a tumor suppressive role, we inactivated Mga in two mouse models of non-small cell lung cancer using a CRISPR-based approach. MGA loss significantly accelerated tumor growth in both models and led to de-repression of non-canonical Polycomb ncPRC1.6 targets, including genes involved in metastasis and meiosis. Moreover, MGA deletion in human lung adenocarcinoma lines augmented invasive capabilities. We further show that MGA-MAX, E2F6, and L3MBTL2 co-occupy thousands of promoters and that MGA stabilizes these ncPRC1.6 subunits. Lastly, we report that MGA loss also induces a pro-growth effect in human colon organoids. Our studies establish MGA as a bona fide tumor suppressor in vivo and suggest a tumor suppressive mechanism in adenocarcinomas resulting from widespread transcriptional attenuation of MYC and E2F target genes mediated by MGA-MAX associated with a non-canonical Polycomb complex.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Epigenetic Repression , Polycomb-Group Proteins/genetics , Adenocarcinoma of Lung/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Disease Progression , Female , Humans , Male , Mice , Neoplasm Invasiveness/genetics , Polycomb-Group Proteins/metabolism
7.
Cancer Cell ; 38(1): 97-114.e7, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32470392

ABSTRACT

Small cell lung cancer (SCLC) is a highly aggressive and lethal neoplasm. To identify candidate tumor suppressors we applied CRISPR/Cas9 gene inactivation screens to a cellular model of early-stage SCLC. Among the top hits was MAX, the obligate heterodimerization partner for MYC family proteins that is mutated in human SCLC. Max deletion increases growth and transformation in cells and dramatically accelerates SCLC progression in an Rb1/Trp53-deleted mouse model. In contrast, deletion of Max abrogates tumorigenesis in MYCL-overexpressing SCLC. Max deletion in SCLC resulted in derepression of metabolic genes involved in serine and one-carbon metabolism. By increasing serine biosynthesis, Max-deleted cells exhibit resistance to serine depletion. Thus, Max loss results in metabolic rewiring and context-specific tumor suppression.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Disease Models, Animal , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , Tumor Suppressor Proteins/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cells, Cultured , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , HEK293 Cells , Hep G2 Cells , Humans , K562 Cells , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Mice, Knockout , Mice, Transgenic , Small Cell Lung Carcinoma/metabolism , Tumor Suppressor Proteins/metabolism
8.
Mol Cell Biol ; 25(16): 6990-7004, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16055712

ABSTRACT

The corepressor mSin3A is the core component of a chromatin-modifying complex that is recruited by multiple gene-specific transcriptional repressors. In order to understand the role of mSin3A during development, we generated constitutive germ line as well as conditional msin3A deletions. msin3A deletion in the developing mouse embryo results in lethality at the postimplantation stage, demonstrating that it is an essential gene. Blastocysts derived from preimplantation msin3A null embryos and mouse embryo fibroblasts (MEFs) lacking msin3A display a significant reduction in cell division. msin3A null MEFs also show mislocalization of the heterochromatin protein, HP1alpha, without alterations in global histone acetylation. Heterozygous msin3A(+/-) mice with a systemic twofold decrease in mSin3A protein develop splenomegaly as well as kidney disease indicative of a disruption of lymphocyte homeostasis. Conditional deletion of msin3A from developing T cells results in reduced thymic cellularity and a fivefold decrease in the number of cytotoxic (CD8) T cells, while helper (CD4) T cells are unaffected. We show that CD8 development is dependent on mSin3A at a step downstream of T-cell receptor signaling and that loss of mSin3A specifically decreases survival of double-positive and CD8 T cells. Thus, msin3A is a pleiotropic gene which, in addition to its role in cell cycle progression, is required for the development and homeostasis of cells in the lymphoid lineage.


Subject(s)
Chromatin/metabolism , Repressor Proteins/physiology , T-Lymphocytes/cytology , Animals , Apoptosis , Blastocyst , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Chromatin/chemistry , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Exons , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Gene Deletion , Gene Expression Regulation, Developmental , Genotype , Glomerulonephritis, Membranous , Heterochromatin/metabolism , Heterozygote , Mice , Mice, Transgenic , Models, Biological , Models, Genetic , Recombination, Genetic , Sin3 Histone Deacetylase and Corepressor Complex , Splenomegaly , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/cytology , Thymus Gland/cytology , Time Factors
9.
Mol Cell Biol ; 25(16): 7078-91, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16055719

ABSTRACT

The Myc-Max-Mad/Mnt network of transcription factors has been implicated in oncogenesis and the regulation of proliferation in vertebrate cells. The identification of Myc and Max homologs in Drosophila melanogaster has demonstrated a critical role for dMyc in cell growth control. In this report, we identify and characterize the third member of this network, dMnt, the sole fly homolog of the mammalian Mnt and Mad family of transcriptional repressors. dMnt possesses two regions characteristic of Mad and Mnt proteins: a basic helix-loop-helix-zipper domain, through which it dimerizes with dMax to form a sequence-specific DNA binding complex, and a Sin-interacting domain, which mediates interaction with the dSin3 corepressor. Using the upstream activation sequence/GAL4 system, we show that expression of dMnt results in an inhibition of cellular growth and proliferation. Furthermore, we have generated a dMnt null allele, which results in flies with larger cells, increased weight, and decreased life span compared to wild-type flies. Our results demonstrate that dMnt is a transcriptional repressor that regulates D. melanogaster body size.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Repressor Proteins/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Alleles , Alternative Splicing , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Body Size , Cell Proliferation , Cell Separation , DNA/metabolism , Dimerization , Drosophila melanogaster/physiology , Flow Cytometry , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Insulin/metabolism , Longevity , Models, Genetic , Mutation , Phenotype , Phylogeny , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic , Two-Hybrid System Techniques
10.
Cancer Cell ; 27(2): 271-85, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25640402

ABSTRACT

Deregulated Myc transcriptionally reprograms cell metabolism to promote neoplasia. Here we show that oncogenic Myc requires the Myc superfamily member MondoA, a nutrient-sensing transcription factor, for tumorigenesis. Knockdown of MondoA, or its dimerization partner Mlx, blocks Myc-induced reprogramming of multiple metabolic pathways, resulting in apoptosis. Identification and knockdown of genes coregulated by Myc and MondoA have allowed us to define metabolic functions required by deregulated Myc and demonstrate a critical role for lipid biosynthesis in survival of Myc-driven cancer. Furthermore, overexpression of a subset of Myc and MondoA coregulated genes correlates with poor outcome of patients with diverse cancers. Coregulation of cancer metabolism by Myc and MondoA provides the potential for therapeutics aimed at inhibiting MondoA and its target genes.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Neuroblastoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Carcinogenesis/genetics , Cellular Reprogramming/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Neuroblastoma/metabolism , Neuroblastoma/pathology , Protein Multimerization , Proto-Oncogene Proteins c-myc/biosynthesis , Xenograft Model Antitumor Assays
11.
Cell Stem Cell ; 3(6): 611-24, 2008 Dec 04.
Article in English | MEDLINE | ID: mdl-19041778

ABSTRACT

Myc activity is emerging as a key element in acquisition and maintenance of stem cell properties. We have previously shown that c-Myc deficiency results in accumulation of defective hematopoietic stem cells (HSCs) due to niche-dependent differentiation defects. Here we report that immature HSCs coexpress c-myc and N-myc mRNA at similar levels. Although conditional deletion of N-myc in the bone marrow does not affect hematopoiesis, combined deficiency of c-Myc and N-Myc (dKO) results in pancytopenia and rapid lethality. Interestingly, proliferation of HSCs depends on both myc genes during homeostasis, but is c-Myc/N-Myc independent during bone marrow repair after injury. Strikingly, while most dKO hematopoietic cells undergo apoptosis, only self-renewing HSCs accumulate the cytotoxic molecule Granzyme B, normally employed by the innate immune system, thereby revealing an unexpected mechanism of stem cell apoptosis. Collectively, Myc activity (c-Myc and N-Myc) controls crucial aspects of HSC function including proliferation, differentiation, and survival.


Subject(s)
Cell Differentiation/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , Animals , Cell Lineage/genetics , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Graft Survival/genetics , Granzymes/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , Pancytopenia/genetics , Pancytopenia/physiopathology , Signal Transduction/genetics , Stress, Physiological/genetics
12.
EMBO J ; 25(12): 2723-34, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16724113

ABSTRACT

The family of myc proto-oncogenes encodes transcription factors (c-, N-, and L-Myc) that regulate cell growth and proliferation and are involved in the etiology of diverse cancers. Myc proteins are thought to function by binding and regulating specific target genes. Here we report that Myc proteins are required for the widespread maintenance of active chromatin. Disruption of N-myc in neuronal progenitors and other cell types leads to nuclear condensation accompanied by large-scale changes in histone modifications associated with chromatin inactivation, including hypoacetylation and altered methylation. These effects are largely reversed by exogenous Myc as well as by differentiation and are mimicked by the Myc antagonist Mad1. The first chromatin changes are evident within 6 h of Myc loss and lead to changes in chromatin structure. Myc widely influences chromatin in part through upregulation of the histone acetyltransferase GCN5. This study provides the first evidence for regulation of global chromatin structure by an oncoprotein and may explain the broad effects of Myc on cell behavior and tumorigenesis.


Subject(s)
Chromatin/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Acetylation , Animals , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Differentiation , DNA/metabolism , Embryo, Mammalian/cytology , Fibroblasts/cytology , Heterochromatin/metabolism , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Methylation , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/deficiency , Rats , Stem Cells/ultrastructure , Transcription Factors/metabolism , p300-CBP Transcription Factors
13.
Genes Dev ; 16(20): 2699-712, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12381668

ABSTRACT

To address the role of N-myc in neurogenesis and in nervous system tumors, it was conditionally disrupted in neuronal progenitor cells (NPCs) with a nestin-Cre transgene. Null mice display ataxia, behavioral abnormalities, and tremors that correlate with a twofold decrease in brain mass that disproportionately affects the cerebellum (sixfold reduced in mass) and the cerebral cortex, both of which show signs of disorganization. In control mice at E12.5, we observe a domain of high N-Myc protein expression in the rapidly proliferating cerebellar primordium. Targeted deletion of N-myc results in severely compromised proliferation as shown by a striking decrease in S phase and mitotic cells as well as in cells expressing the Myc target gene cyclin D2, whereas apoptosis is unaffected. Null progenitor cells also have comparatively high levels of the cdk inhibitors p27(Kip1) and p18(Ink4c), whereas p15(Ink4b), p21(Cip1), and p19(Ink4d) levels are unaffected. Many null progenitors also exhibit altered nuclear morphology and size. In addition, loss of N-myc disrupts neuronal differentiation as evidenced by ectopic staining of the neuron specific marker betaTUBIII in the cerebrum. Furthermore, in progenitor cell cultures derived from null embryonic brain, we observe a dramatic increase in neuronal differentiation compared with controls. Thus, N-myc is essential for normal neurogenesis, regulating NPC proliferation, differentiation, and nuclear size. Its effects on proliferation and differentiation appear due, at least in part, to down-regulation of a specific subset of cyclin-dependent kinase inhibitors.


Subject(s)
Cerebellum/embryology , Genes, myc/physiology , Neurons/physiology , Stem Cells/physiology , Animals , Apoptosis , Ataxia , Behavior, Animal , Cell Differentiation/physiology , Cell Division , Cerebellum/physiology , Cyclin D2 , Cyclin-Dependent Kinases , Cyclins/metabolism , DNA/metabolism , DNA Primers/chemistry , Gene Expression Regulation , Immunoenzyme Techniques , Integrases/metabolism , Mice , Mice, Knockout , Morphogenesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tremor , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL