Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Nature ; 589(7842): 437-441, 2021 01.
Article in English | MEDLINE | ID: mdl-33299176

ABSTRACT

The formation of arteries is thought to occur by the induction of a highly conserved arterial genetic programme in a subset of vessels that will later experience an increase in oxygenated blood flow1,2. The initial steps of arterial specification require both the VEGF and Notch signalling pathways3-5. Here, we combine inducible genetic mosaics and transcriptomics to modulate and define the function of these signalling pathways in cell proliferation, arteriovenous differentiation and mobilization. We show that endothelial cells with high levels of VEGF or Notch signalling are intrinsically biased to mobilize and form arteries; however, they are not genetically pre-determined, and can also form veins. Mechanistically, we found that increased levels of VEGF and Notch signalling in pre-arterial capillaries suppresses MYC-dependent metabolic and cell-cycle activities, and promotes the incorporation of endothelial cells into arteries. Mosaic lineage-tracing studies showed that endothelial cells that lack the Notch-RBPJ transcriptional activator complex rarely form arteries; however, these cells regained the ability to form arteries when the function of MYC was suppressed. Thus, the development of arteries does not require the direct induction of a Notch-dependent arterial differentiation programme, but instead depends on the timely suppression of endothelial cell-cycle progression and metabolism, a process that precedes arterial mobilization and complete differentiation.


Subject(s)
Arteries/cytology , Arteries/growth & development , Cell Proliferation , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Differentiation/genetics , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Male , Mice , Mosaicism , Mutation , Phenotype , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Notch/deficiency , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Time Factors , Transcription, Genetic , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Veins/cytology
2.
Nat Immunol ; 13(11): 1092-100, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23001146

ABSTRACT

Germinal centers (GCs) are sites of intense B cell proliferation and are central for T cell-dependent antibody responses. However, the role of c-Myc, a key cell-cycle regulator, in this process has been questioned. Here we identified c-Myc(+) B cell subpopulations in immature and mature GCs and found, by genetic ablation of Myc, that they had indispensable roles in the formation and maintenance of GCs. The identification of these functionally critical cellular subsets has implications for human B cell lymphomagenesis, which originates mostly from GC B cells and frequently involves MYC chromosomal translocations. As these translocations are generally dependent on transcription of the recombining partner loci, the c-Myc(+) GC subpopulations may be at a particularly high risk for malignant transformation.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/metabolism , Cell Cycle/genetics , Germinal Center/metabolism , Proto-Oncogene Proteins c-myc/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Cycle/immunology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Gene Deletion , Gene Expression Regulation/immunology , Genes, Reporter , Genetic Loci , Germinal Center/immunology , Germinal Center/pathology , Green Fluorescent Proteins , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/immunology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Translocation, Genetic
3.
FASEB J ; 36(1): e22077, 2022 01.
Article in English | MEDLINE | ID: mdl-34878671

ABSTRACT

Endothelial cells play an essential role in inflammation through synthesis and secretion of chemoattractant cytokines and expression of adhesion molecules required for inflammatory cell attachment and infiltration. The mechanisms by which endothelial cells control the pro-inflammatory response depend on the type of inflammatory stimuli, endothelial cell origin, and tissue involved. In the present study, we investigated the role of the transcription factor c-Myc in inflammation using a conditional knockout mouse model in which Myc is specifically deleted in the endothelium. At a systemic level, circulating monocytes, the chemokine CCL7, and the extracellular-matrix protein osteopontin were significantly increased in endothelial c-Myc knockout (EC-Myc KO) mice, whereas the cytokine TNFSF11 was downregulated. Using an experimental model of steatohepatitis, we investigated the involvement of endothelial c-Myc in diet-induced inflammation. EC-Myc KO animals displayed enhanced pro-inflammatory response, characterized by increased expression of pro-inflammatory cytokines and leukocyte infiltration, and worsened liver fibrosis. Transcriptome analysis identified enhanced expression of genes associated with inflammation, fibrosis, and hepatocellular carcinoma in EC-Myc KO mice relative to control (CT) animals after short-exposure to high-fat diet. Analysis of a single-cell RNA-sequencing dataset of human cirrhotic livers indicated downregulation of MYC in endothelial cells relative to healthy controls. In summary, our results suggest a protective role of endothelial c-Myc in diet-induced liver inflammation and fibrosis. Targeting c-Myc and its downstream pathways in the endothelium may constitute a potential strategy for the treatment of inflammatory disease.


Subject(s)
Diet, High-Fat/adverse effects , Endothelium/metabolism , Fatty Liver , Liver Cirrhosis , Proto-Oncogene Proteins c-myc/deficiency , Animals , Endothelium/pathology , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Gene Knockout Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myc/metabolism
4.
Nature ; 529(7585): 216-20, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26735015

ABSTRACT

Endothelial cells (ECs) are plastic cells that can switch between growth states with different bioenergetic and biosynthetic requirements. Although quiescent in most healthy tissues, ECs divide and migrate rapidly upon proangiogenic stimulation. Adjusting endothelial metabolism to the growth state is central to normal vessel growth and function, yet it is poorly understood at the molecular level. Here we report that the forkhead box O (FOXO) transcription factor FOXO1 is an essential regulator of vascular growth that couples metabolic and proliferative activities in ECs. Endothelial-restricted deletion of FOXO1 in mice induces a profound increase in EC proliferation that interferes with coordinated sprouting, thereby causing hyperplasia and vessel enlargement. Conversely, forced expression of FOXO1 restricts vascular expansion and leads to vessel thinning and hypobranching. We find that FOXO1 acts as a gatekeeper of endothelial quiescence, which decelerates metabolic activity by reducing glycolysis and mitochondrial respiration. Mechanistically, FOXO1 suppresses signalling by MYC (also known as c-MYC), a powerful driver of anabolic metabolism and growth. MYC ablation impairs glycolysis, mitochondrial function and proliferation of ECs while its EC-specific overexpression fuels these processes. Moreover, restoration of MYC signalling in FOXO1-overexpressing endothelium normalizes metabolic activity and branching behaviour. Our findings identify FOXO1 as a critical rheostat of vascular expansion and define the FOXO1-MYC transcriptional network as a novel metabolic checkpoint during endothelial growth and proliferation.


Subject(s)
Endothelium, Vascular/growth & development , Endothelium, Vascular/metabolism , Forkhead Transcription Factors/metabolism , Animals , Cell Proliferation , Cell Respiration , Endothelium, Vascular/cytology , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Glycolysis , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction
5.
Nature ; 534(7607): 341-6, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281222

ABSTRACT

Chronic myeloid leukaemia (CML) arises after transformation of a haemopoietic stem cell (HSC) by the protein-tyrosine kinase BCR-ABL. Direct inhibition of BCR-ABL kinase has revolutionized disease management, but fails to eradicate leukaemic stem cells (LSCs), which maintain CML. LSCs are independent of BCR-ABL for survival, providing a rationale for identifying and targeting kinase-independent pathways. Here we show--using proteomics, transcriptomics and network analyses--that in human LSCs, aberrantly expressed proteins, in both imatinib-responder and non-responder patients, are modulated in concert with p53 (also known as TP53) and c-MYC regulation. Perturbation of both p53 and c-MYC, and not BCR-ABL itself, leads to synergistic cell kill, differentiation, and near elimination of transplantable human LSCs in mice, while sparing normal HSCs. This unbiased systems approach targeting connected nodes exemplifies a novel precision medicine strategy providing evidence that LSCs can be eradicated.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Acetamides/pharmacology , Acetamides/therapeutic use , Animals , Antigens, CD34/metabolism , Azepines/pharmacology , Azepines/therapeutic use , Cell Death/drug effects , Cell Differentiation/drug effects , DNA-Binding Proteins/metabolism , Female , Fusion Proteins, bcr-abl/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Imidazolines/pharmacology , Imidazolines/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Male , Mice , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/transplantation , Proteomics , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Transcriptome , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Genes Dev ; 27(10): 1101-14, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23699408

ABSTRACT

Tumorigenesis results from dysregulation of oncogenes and tumor suppressors that influence cellular proliferation, differentiation, apoptosis, and/or senescence. Many gene products involved in these processes are substrates of the E3 ubiquitin ligase Mule/Huwe1/Arf-BP1 (Mule), but whether Mule acts as an oncogene or tumor suppressor in vivo remains controversial. We generated K14Cre;Mule(flox/flox(y)) (Mule kKO) mice and subjected them to DMBA/PMA-induced skin carcinogenesis, which depends on oncogenic Ras signaling. Mule deficiency resulted in increased penetrance, number, and severity of skin tumors, which could be reversed by concomitant genetic knockout of c-Myc but not by knockout of p53 or p19Arf. Notably, in the absence of Mule, c-Myc/Miz1 transcriptional complexes accumulated, and levels of p21CDKN1A (p21) and p15INK4B (p15) were down-regulated. In vitro, Mule-deficient primary keratinocytes exhibited increased proliferation that could be reversed by Miz1 knockdown. Transfer of Mule-deficient transformed cells to nude mice resulted in enhanced tumor growth that again could be abrogated by Miz1 knockdown. Our data demonstrate in vivo that Mule suppresses Ras-mediated tumorigenesis by preventing an accumulation of c-Myc/Miz1 complexes that mediates p21 and p15 down-regulation.


Subject(s)
Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Nuclear Proteins/antagonists & inhibitors , Oncogene Protein p21(ras)/metabolism , Protein Inhibitors of Activated STAT/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Genes, ras , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Protein p21(ras)/antagonists & inhibitors , Oncogene Protein p21(ras)/genetics , Protein Inhibitors of Activated STAT/deficiency , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Suppressor Protein p53 , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
7.
Nature ; 507(7491): 243-7, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24509714

ABSTRACT

The transcription factors c-Myc and N-Myc--encoded by Myc and Mycn, respectively--regulate cellular growth and are required for embryonic development. A third paralogue, Mycl1, is dispensable for normal embryonic development but its biological function has remained unclear. To examine the in vivo function of Mycl1 in mice, we generated an inactivating Mycl1(gfp) allele that also reports Mycl1 expression. We find that Mycl1 is selectively expressed in dendritic cells (DCs) of the immune system and controlled by IRF8, and that during DC development, Mycl1 expression is initiated in the common DC progenitor concurrent with reduction in c-Myc expression. Mature DCs lack expression of c-Myc and N-Myc but maintain L-Myc expression even in the presence of inflammatory signals such as granulocyte-macrophage colony-stimulating factor. All DC subsets develop in Mycl1-deficient mice, but some subsets such as migratory CD103(+) conventional DCs in the lung and liver are greatly reduced at steady state. Importantly, loss of L-Myc by DCs causes a significant decrease in in vivo T-cell priming during infection by Listeria monocytogenes and vesicular stomatitis virus. The replacement of c-Myc by L-Myc in immature DCs may provide for Myc transcriptional activity in the setting of inflammation that is required for optimal T-cell priming.


Subject(s)
Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Proto-Oncogene Proteins c-myc/metabolism , T-Lymphocytes/immunology , Animals , Antigens, CD/metabolism , Cell Division , Dendritic Cells/cytology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation/immunology , Inflammation/metabolism , Integrin alpha Chains/metabolism , Interferon Regulatory Factors/metabolism , Listeria monocytogenes/immunology , Liver/cytology , Liver/immunology , Lung/cytology , Lung/immunology , Male , Mice , Proto-Oncogene Proteins c-myc/deficiency , Transcription, Genetic , Vesiculovirus/immunology
8.
Cell Biochem Funct ; 37(4): 266-272, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31025404

ABSTRACT

MYCL1 protein expression encoded by a proto-oncogene MYCL1, a member of the MYC family, is correlated with poor prognosis in gastric cancer patients. Nevertheless, the role of MYCL1 in gastric cancer cells remains unknown. In this study, the expression levels of MYCL1 mRNA and protein were downregulated by lentiviral-mediated RNA interference (RNAi) in the MGC-803 gastric cancer cell line. Then, the influence of MYCL1 on the biological behaviour of gastric cancer cells was investigated. Finally, a stable animal model of the MGC-803 human gastric cancer tumour model in nude mice was made successfully. Functionally, silencing of MYCL1 inhibited migration and invasion of the MGC-803 line in vitro and was accompanied with some ultrastructural changes. These results provide some evidences that lentiviral-mediated MYCL1 silencing may be a novel therapeutic strategy for the treatment of gastric cancer. SIGNIFICANCE OF THE STUDY: Gastric cancer is one of the most common malignant tumours worldwide and the second leading cause of cancer-related death in China. Our previous study revealed that expression of MYCL1 in gastric cancer tissue was associated with poor prognosis of patients. However, the potential underlying mechanism is still unclear. In the current study, we displayed the influence of MYCL1 gene on invasion and migration phenotype of gastric cancer cells and provided a possible explanation from the aspect of structural alteration. Our results suggested that downregulation of MYCL1 may be a potential therapeutic strategy for gastric cancer.


Subject(s)
Cell Movement/genetics , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Down-Regulation , Humans , Proto-Oncogene Mas , Stomach Neoplasms/metabolism , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 113(8): 2258-63, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26873106

ABSTRACT

High-throughput, genome-wide translocation sequencing (HTGTS) studies of activated B cells have revealed that DNA double-strand breaks (DSBs) capable of translocating to defined bait DSBs are enriched around the transcription start sites (TSSs) of active genes. We used the HTGTS approach to investigate whether a similar phenomenon occurs in primary neural stem/progenitor cells (NSPCs). We report that breakpoint junctions indeed are enriched around TSSs that were determined to be active by global run-on sequencing analyses of NSPCs. Comparative analyses of transcription profiles in NSPCs and B cells revealed that the great majority of TSS-proximal junctions occurred in genes commonly expressed in both cell types, possibly because this common set has higher transcription levels on average than genes transcribed in only one or the other cell type. In the latter context, among all actively transcribed genes containing translocation junctions in NSPCs, those with junctions located within 2 kb of the TSS show a significantly higher transcription rate on average than genes with junctions in the gene body located at distances greater than 2 kb from the TSS. Finally, analysis of repair junction signatures of TSS-associated translocations in wild-type versus classical nonhomologous end-joining (C-NHEJ)-deficient NSPCs reveals that both C-NHEJ and alternative end-joining pathways can generate translocations by joining TSS-proximal DSBs to DSBs on other chromosomes. Our studies show that the generation of transcription-associated DSBs is conserved across divergent cell types.


Subject(s)
DNA Breaks, Double-Stranded , Neural Stem Cells/metabolism , Transcription, Genetic , Translocation, Genetic , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , B-Lymphocytes/metabolism , Cells, Cultured , DNA End-Joining Repair , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, myc , Genes, p53 , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcription Initiation Site , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
Arterioscler Thromb Vasc Biol ; 34(8): 1739-46, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24925976

ABSTRACT

OBJECTIVE: Nitric oxide synthase 3 (NOS3) prevents neointima hyperplasia by still unknown mechanisms. To demonstrate the significance of endothelial nitric oxide in the polarization of infiltrated macrophages through the expression of matrix metalloproteinase (MMP)-13 in neointima formation. APPROACH AND RESULTS: After aortic endothelial denudation, NOS3 null mice show elevated neointima formation, detecting increased mobilization of LSK (lineage-negative [Lin]-stem-cell antigen 1 [SCA1]+KIT+) progenitor cells, and high ratios of M1 (proinflammatory) to M2 (resolving) macrophages, accompanied by high expression of interleukin-5, interleukin-6, MCP-1 (monocyte chemoattractant protein), VEGF (vascular endothelial growth factor), GM-CSF (granulocyte-macrophage colony stimulating factor), interleukin-1ß, and interferon-γ. In conditional c-Myc knockout mice, in which M2 polarization is defective, denuded aortas showed extensive wall thickening as well. Conditioned medium from NOS3-deficient endothelium induced extensive repolarization of M2 macrophages to an M1 phenotype, and vascular smooth muscle cells proliferated and migrated faster in conditioned medium from M1 macrophages. Among the different proteins participating in cell migration, MMP-13 was preferentially expressed by M1 macrophages. M1-mediated vascular smooth muscle cell migration was inhibited when macrophages were isolated from MMP-13-deficient mice, whereas exogenous administration of MMP-13 to vascular smooth muscle cell fully restored migration. Excess vessel wall thickening in mice lacking NOS3 was partially reversed by simultaneous deletion of MMP-13, indicating that NOS3 prevents neointimal hyperplasia by preventing MMP-13 activity. An excess of M1-polarized macrophages that coexpress MMP-13 was also detected in human carotid samples from endarterectomized patients. CONCLUSIONS: These findings indicate that at least M1 macrophage-mediated expression of MMP-13 in NOS3 null mice induces neointima formation after vascular injury, suggesting that MMP-13 may represent a new promising target in vascular disease.


Subject(s)
Aortic Diseases/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Nitric Oxide/metabolism , Aged , Aged, 80 and over , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Biomarkers/metabolism , Case-Control Studies , Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Humans , Hyperplasia , Inflammation Mediators/metabolism , Macrophages/enzymology , Macrophages/pathology , Male , Matrix Metalloproteinase 13/deficiency , Matrix Metalloproteinase 13/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Phenotype , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Time Factors
11.
Exp Cell Res ; 319(20): 3190-200, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24095950

ABSTRACT

Induced pluripotent stem cells (iPSCs) hold tremendous potential for the development of new regenerative medicine therapies and the study of molecular mechanisms of pluripotency and development. However, reactivation of c-Myc, which results in tumor formation in chimeric mice, is a major roadblock in the translation of iPSCs into therapies. Although ectopic expression of c-Myc is not absolutely required for somatic reprogramming, in the absence of c-Myc, the overall efficiency of reprogramming is drastically reduced and the reprogramming time is increased. Subtle, abnormal epigenetic modifications in iPSCs derived in the absence of c-Myc have also been documented. Therefore, we developed a reprogramming method without c-Myc to generate high-quality iPSCs, a prerequisite to harnessing the full potential of iPSCs. In this study, we determined that serum replacement (SR)-based culture conditions dramatically increased the transcription factor-mediated reprogramming of mouse embryonic fibroblast cells (MEFs). The process was shortened to approximately 8 days when Oct4/Sox2/Klf4 (3F)-transduced MEFs were first cultured for 3 days under low serum conditions (LS protocol). The 3F-derived iPSCs that were generated by this method resembled mouse ES cells (mESCs) in morphology, gene expression, and in vitro differentiation. Finally, we observed that 3F-derived iPSC colonies were able to reach definite pluripotency in terms of molecular signatures when the catalytic function of c-Myc was tolerated. The 3F induction of pluripotency described here should facilitate the use of iPSCs and may also facilitate the mechanistic dissection of somatic reprogramming.


Subject(s)
Cell Separation/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/deficiency , Animals , Cells, Cultured , Kruppel-Like Factor 4 , Mice
12.
J Biol Chem ; 287(8): 5627-38, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22184109

ABSTRACT

Although p63 and MYC are important in the control of epidermal homeostasis, the underlying molecular mechanisms governing keratinocyte proliferation or differentiation downstream of these two genes are not completely understood. By analyzing the transcriptional changes and phenotypic consequences of the loss of either p63 or MYC in human developmentally mature keratinocytes, we have characterized the networks acting downstream of these two genes to control epidermal homeostasis. We show that p63 is required to maintain growth and to commit to differentiation by two distinct mechanisms. Knockdown of p63 led to down-regulation of MYC via the Wnt/ß-catenin and Notch signaling pathways and in turn reduced keratinocyte proliferation. We demonstrate that a p63-controlled keratinocyte cell fate network is essential to induce the onset of keratinocyte differentiation. This network contains several secreted proteins involved in cell migration/adhesion, including fibronectin 1 (FN1), interleukin-1ß (IL1B), cysteine-rich protein 61 (CYR61), and jagged-1 (JAG1), that act downstream of p63 as key effectors to trigger differentiation. Our results characterized for the first time a connection between p63 and MYC and a cell adhesion-related network that controls differentiation. Furthermore, we show that the balance between the MYC-controlled cell cycle progression network and the p63-controlled cell adhesion-related network could dictate skin cell fate.


Subject(s)
Cell Differentiation/genetics , Gene Regulatory Networks , Keratinocytes/cytology , Keratinocytes/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Cell Adhesion/genetics , Cell Cycle Checkpoints/genetics , Cell Movement/genetics , Cell Proliferation , Down-Regulation/genetics , Gene Knockdown Techniques , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , RNA, Small Interfering/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics , Transcriptome/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism
13.
Crit Rev Immunol ; 32(4): 321-34, 2012.
Article in English | MEDLINE | ID: mdl-23237508

ABSTRACT

Since the discovery of the myc gene, few genes are likely to have such influence on biomedical research. The diversity of the biological functions regulated by this transcription factor and its impact in human health have attracted investigators from many different fields. The development of conditional knockout mouse models has allowed for the characterization of Myc-driven molecular mechanisms in primary cells in physiological and pathological conditions. In this review, we discuss some of the main functions and recent findings regarding c-Myc in in vivo B lymphocyte differentiation from early progenitors to terminally differentiated cells.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation , Proto-Oncogene Proteins c-myc/metabolism , Animals , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics
14.
Nature ; 446(7136): 676-9, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17377531

ABSTRACT

The APC gene encodes the adenomatous polyposis coli tumour suppressor protein, germline mutation of which characterizes familial adenomatous polyposis (FAP), an autosomal intestinal cancer syndrome. Inactivation of APC is also recognized as the key early event in the development of sporadic colorectal cancers, and its loss results in constitutive activity of the beta-catenin-Tcf4 transcription complex. The proto-oncogene c-MYC has been identified as a target of the Wnt pathway in colorectal cancer cells in vitro, in normal crypts in vivo and in intestinal epithelial cells acutely transformed on in vivo deletion of the APC gene; however, the significance of this is unclear. Therefore, to elucidate the role Myc has in the intestine after Apc loss, we have simultaneously deleted both Apc and Myc in the adult murine small intestine. Here we show that loss of Myc rescued the phenotypes of perturbed differentiation, migration, proliferation and apoptosis, which occur on deletion of Apc. Remarkably, this rescue occurred in the presence of high levels of nuclear beta-catenin. Array analysis revealed that Myc is required for the majority of Wnt target gene activation following Apc loss. These data establish Myc as the critical mediator of the early stages of neoplasia following Apc loss.


Subject(s)
Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Gene Deletion , Genes, APC , Genes, myc/genetics , Intestine, Small/metabolism , Proto-Oncogene Proteins c-myc/deficiency , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Animals , Enterocytes/metabolism , Intestine, Small/cytology , Male , Mice , Phenotype , Proto-Oncogene Proteins c-myc/genetics
15.
Nature ; 448(7152): 445-51, 2007 Jul 26.
Article in English | MEDLINE | ID: mdl-17597761

ABSTRACT

The c-Myc proto-oncogene encodes a transcription factor that is essential for cell growth and proliferation and is broadly implicated in tumorigenesis. However, the biological functions required by c-Myc to induce oncogenesis remain elusive. Here we show that c-Myc has a direct role in the control of DNA replication. c-Myc interacts with the pre-replicative complex and localizes to early sites of DNA synthesis. Depletion of c-Myc from mammalian (human and mouse) cells as well as from Xenopus cell-free extracts, which are devoid of RNA transcription, demonstrates a non-transcriptional role for c-Myc in the initiation of DNA replication. Overexpression of c-Myc causes increased replication origin activity with subsequent DNA damage and checkpoint activation. These findings identify a critical function of c-Myc in DNA replication and suggest a novel mechanism for its normal and oncogenic functions.


Subject(s)
DNA Replication/genetics , Proto-Oncogene Proteins c-myc/metabolism , Replication Origin/genetics , Animals , Cell Extracts , Cell Transformation, Neoplastic , Cells, Cultured , DNA Damage/genetics , Fibroblasts , HeLa Cells , Humans , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Transcription, Genetic , Xenopus
16.
Proc Natl Acad Sci U S A ; 106(21): 8641-6, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19423665

ABSTRACT

The molecular requirements for invariant Valpha14-bearing natural killer T cells (iNKT) in the thymus are poorly understood. A minute population of approximately 500 newly selected CD69(+)CD24(+) stage 0 (ST0) iNKT cells gives rise to approximately 100 times more CD44(neg/lo)CD24(-) stage 1 (ST1) cells, which then generate similar frequencies of CD44(hi)CD24(-) stage 2 (ST2) and mature iNKT cells. Although the increased number of ST1 compared with ST0 cells indicates the initiation of a proliferation wave in the very early stages of iNKT cell development, details about the controlling mechanism are currently lacking. Here, we show that the transcription factor c-Myc is required for iNKT cell development. Conditional ablation of c-Myc in double-positive thymocytes specifically impacted iNKT but not conventional T cell development. Within the iNKT population, a progressive reduction of iNKT cells was observed starting at ST1 (approximately 50-fold) and ST2 (approximately 350-fold), with a complete lack of mature cells in thymus, spleen, and liver. ST0/ST1 c-Myc-deficient iNKT cells showed reduced proliferation. In contrast, annexin V staining did not reveal increased apoptosis, and transgenic overexpression of BCL-2 did not rescue iNKT cell development in c-Myc-deficient mice. Moreover, expression of known iNKT differentiation factors such as Plzf and Gata3 was not dramatically altered. These, findings provide compelling evidence that c-Myc mediates an intrathymic proliferation wave immediately after agonist selection of iNKT cells and illustrate the importance of this expansion for the generation of mature iNKT cells in vivo.


Subject(s)
Cell Differentiation/immunology , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Cell Lineage/immunology , Cell Proliferation , DNA, Complementary/genetics , Gene Expression Regulation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics
17.
Int J Mol Sci ; 13(3): 3598-3617, 2012.
Article in English | MEDLINE | ID: mdl-22489170

ABSTRACT

The only curative treatment for hepatic failure is liver transplantation. Unfortunately, this treatment has several major limitations, as for example donor organ shortage. A previous report demonstrated that transplantation of induced pluripotent stem cells without reprogramming factor c-Myc (3-genes iPSCs) attenuates thioacetamide-induced hepatic failure with minimal incidence of tumorigenicity. In this study, we investigated whether 3-genes iPSC transplantation is capable of rescuing carbon tetrachloride (CCl(4))-induced fulminant hepatic failure and hepatic encephalopathy in mice. Firstly, we demonstrated that 3-genes iPSCs possess the capacity to differentiate into hepatocyte-like cells (iPSC-Heps) that exhibit biological functions and express various hepatic specific markers. 3-genes iPSCs also exhibited several antioxidant enzymes that prevented CCl(4)-induced reactive oxygen species production and cell death. Intraperitoneal transplantation of either 3-genes iPSCs or 3-genes iPSC-Heps significantly reduced hepatic necrotic areas, improved hepatic functions, and survival rate in CCl(4)-treated mice. CCl(4)-induced hepatic encephalopathy was also improved by 3-genes iPSC transplantation. Hoechst staining confirmed the successful engraftment of both 3-genes iPSCs and 3-genes iPSC-Heps, indicating the homing properties of these cells. The most pronounced hepatoprotective effect of iPSCs appeared to originate from the highest antioxidant activity of 3-gene iPSCs among all transplanted cells. In summary, our findings demonstrated that 3-genes iPSCs serve as an available cell source for the treatment of an experimental model of acute liver diseases.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Chemical and Drug Induced Liver Injury/therapy , Hepatic Encephalopathy/therapy , Induced Pluripotent Stem Cells/transplantation , Liver Failure, Acute/therapy , Animals , Antioxidants/metabolism , Carbon Tetrachloride/adverse effects , Cell Differentiation , Cell Survival , Cells, Cultured , Cellular Reprogramming , Disease Models, Animal , Hepatocytes/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Liver/pathology , Liver Failure, Acute/chemically induced , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Octamer Transcription Factor-3/biosynthesis , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Reactive Oxygen Species/metabolism , SOXB1 Transcription Factors/biosynthesis
18.
Cell Rep ; 35(11): 109264, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34133930

ABSTRACT

MYC activates different metabolic programs in a cell-type- and cell-status-dependent manner. However, the role of MYC in inflammatory macrophages has not yet been determined. Metabolic and molecular analyses reveal that MYC, but not hypoxia inducible factor 1 (HIF1), is involved in enhancing early glycolytic flux during inflammatory macrophage polarization. Ablation of MYC decreases lactate production by regulating lactate dehydrogenase (LDH) activity and causes increased inflammatory cytokines by regulating interferon regulatory factor 4 (IRF4) in response to lipopolysaccharide. Moreover, myeloid-specific deletion of MYC and pharmacological inhibition of the MYC/LDH axis enhance inflammation and the bacterial clearance in vivo. These results elucidate the potential role of the MYC/LDH/IRF4 axis in inflammatory macrophages by connecting early glycolysis with inflammatory responses and suggest that modulating early glycolytic flux mediated by the MYC/LDH axis can be used to open avenues for the therapeutic modulation of macrophage polarization to fight against bacterial infection.


Subject(s)
Glycolysis , Inflammation/metabolism , Inflammation/pathology , Interferon Regulatory Factors/metabolism , Macrophages/metabolism , Macrophages/pathology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Bacteria/metabolism , Cytokines/biosynthesis , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity, Innate , Inflammation Mediators/metabolism , Lactic Acid/metabolism , Lipopolysaccharides , Male , Mice, Knockout , Proto-Oncogene Proteins c-myc/deficiency
19.
Cerebellum ; 9(4): 537-47, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20658325

ABSTRACT

Separate murine knockout (KO) of either c- or N-myc genes in neural stem and precursor cells (NSC) driven by nestin-cre causes microcephaly. The cerebellum is particularly affected in the N-myc KO, leading to a strong reduction in cerebellar granule neural progenitors (CGNP) and mature granule neurons. In humans, mutation of N-myc also causes microcephaly in Feingold Syndrome. We created a double KO (DKO) of c- and N-myc using nestin-cre, which strongly impairs brain growth, particularly that of the cerebellum. Granule neurons were almost absent from the Myc DKO cerebellum, and other cell types were relatively overrepresented, including astroglia, oligodendrocytes, and Purkinje neurons. These findings are indicative of a profound disruption of cell fate of cerebellar stem and precursors. DKO Purkinje neurons were strikingly lacking in normal arborization. Inhibitory neurons were ectopic and exhibited very abnormal GAD67 staining patterns. Also consistent with altered cell fate, the adult DKO cerebellum still retained a residual external germinal layer (EGL). CGNP in the DKO EGL were almost uniformly NeuN and p27KIP1 positive as well as negative for Math1 and BrdU at the peak of normal cerebellar proliferation at P6. The presence of some mitotic CGNP in the absence of S phase cells suggests a possible arrest in M phase. CGNP and NSC metabolism also was affected by loss of Myc as DKO cells exhibited weak nucleolin staining. Together these findings indicate that c- and N-Myc direct cerebellar development by maintaining CGNP and NSC populations through inhibiting differentiation as well as directing rapid cell cycling and active cellular metabolism.


Subject(s)
Cell Cycle/physiology , Cerebellum , Gene Expression Regulation, Developmental/genetics , Neural Stem Cells/physiology , Proto-Oncogene Proteins c-myc/metabolism , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Age Factors , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Brain/growth & development , Brain/metabolism , Bromodeoxyuridine/metabolism , Cerebellum/cytology , Cerebellum/growth & development , Cerebellum/metabolism , Embryo, Mammalian , Glutamate Decarboxylase/metabolism , Intermediate Filament Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Proto-Oncogene Proteins c-myc/deficiency , Tubulin/metabolism
20.
Cell Death Differ ; 26(7): 1283-1298, 2019 07.
Article in English | MEDLINE | ID: mdl-30258097

ABSTRACT

ZNF322A, a C2H2 zinc finger transcription factor, is an oncoprotein in lung cancer. However, the transcription mechanisms of ZNF322A in lung cancer stem cell-like reprogramming remain elusive. By integrating our chromatin immunoprecipitation-sequencing and RNA-sequencing datasets, we identified and validated the transcriptional targets of ZNF322A, which were significantly enriched in tumorigenic functions and developmental processes. Indeed, overexpression of ZNF322A promoted self-renewal ability and increased stemness-related gene expressions in vitro and in vivo. Importantly, ZNF322A bound directly to c-Myc promoter and recruited histone deacetylase 3 to transcriptionally suppress c-Myc expression, which in turn increased mitochondrial oxidative phosphorylation and promoted cell motility, thus maintaining stem cell-like properties of lung cancer. Clinically, ZNF322AHigh/c-MycLow expression profile was revealed as an independent indicator of poor prognosis in lung cancer patients. Our study provides the first evidence that ZNF322A-centered transcriptome promotes lung tumorigenesis and ZNF322A acts as a transcription suppressor of c-Myc to maintain lung cancer stem cell-like properties by shifting metabolism towards oxidative phosphorylation.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/metabolism , Transcription, Genetic , A549 Cells , Humans , Lung Neoplasms/metabolism , Neoplastic Stem Cells/pathology , Oncogene Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-myc/deficiency , Transcription Factors/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL