18 research outputs found

    Interaction between SNAI2 and MYOD enhances oncogenesis and suppresses differentiation in Fusion Negative Rhabdomyosarcoma

    Get PDF
    Rhabdomyosarcoma (RMS) is an aggressive pediatric malignancy of the muscle, that includes Fusion Positive (FP)-RMS harboring PAX3/7-FOXO1 and Fusion Negative (FN)-RMS commonly with RAS pathway mutations. RMS express myogenic master transcription factors MYOD and MYOG yet are unable to terminally differentiate. Here, we report that SNAI2 is highly expressed in FN-RMS, is oncogenic, blocks myogenic differentiation, and promotes growth. MYOD activates SNAI2 transcription via super enhancers with striped 3D contact architecture. Genome wide chromatin binding analysis demonstrates that SNAI2 preferentially binds enhancer elements and competes with MYOD at a subset of myogenic enhancers required for terminal differentiation. SNAI2 also suppresses expression of a muscle differentiation program modulated by MYOG, MEF2, and CDKN1A. Further, RAS/MEK-signaling modulates SNAI2 levels and binding to chromatin, suggesting that the differentiation blockade by oncogenic RAS is mediated in part by SNAI2. Thus, an interplay between SNAI2, MYOD, and RAS prevents myogenic differentiation and promotes tumorigenesis. Rhabdomyosarcomas are tumours blocked in myogenic differentiation, which despite the expression of master muscle regulatory factors, including MYOD, are unable to differentiate. Here, the authors show that SNAI2 is upregulated by MYOD through super enhancers, binds to MYOD target enhancers, and arrests differentiation

    MYOD-SKP2 axis boosts tumorigenesis in fusion negative rhabdomyosarcoma by preventing differentiation through p57Kip2^{Kip2} targeting

    Get PDF
    Rhabdomyosarcomas (RMS) are pediatric mesenchymal-derived malignancies encompassing PAX3/7-FOXO1 Fusion Positive (FP)-RMS, and Fusion Negative (FN)-RMS with frequent RAS pathway mutations. RMS express the master myogenic transcription factor MYOD that, whilst essential for survival, cannot support differentiation. Here we discover SKP2, an oncogenic E3-ubiquitin ligase, as a critical pro-tumorigenic driver in FN-RMS. We show that SKP2 is overexpressed in RMS through the binding of MYOD to an intronic enhancer. SKP2 in FN-RMS promotes cell cycle progression and prevents differentiation by directly targeting p27Kip1^{Kip1} and p57Kip2^{Kip2}, respectively. SKP2 depletion unlocks a partly MYOD-dependent myogenic transcriptional program and strongly affects stemness and tumorigenic features and prevents in vivo tumor growth. These effects are mirrored by the investigational NEDDylation inhibitor MLN4924. Results demonstrate a crucial crosstalk between transcriptional and post-translational mechanisms through the MYOD-SKP2 axis that contributes to tumorigenesis in FN-RMS. Finally, NEDDylation inhibition is identified as a potential therapeutic vulnerability in FN-RMS

    Quercetin attenuates Monocyte Chemoattractant Protein-1 gene expression in glucose primed aortic endothelial cells through NF-κB and AP-1

    No full text
    Monocyte Chemoattractant Protein-1 (MCP-1) is involved in the diapedesis of blood monocytes into the arterial intima, an early critical event in atherogenesis. Modulating MCP-1 expression can be a key strategy to decrease the risk for atherosclerosis in diabetes. We hypothesized that quercetin, an anti-inflammatory molecule could modulate high glucose concentration (HG) induced MCP-1 expression in aortic endothelial cells in vitro because of its regulatory effects on Activator Protein-1 (AP-1) and Nuclear Factor-κB (NF-κB). Rat aortic endothelial cells (RAECs) were exposed to HG in the presence or absence of quercetin. Quercetin attenuated HG induced MCP-1 mRNA (42%) and protein synthesis (45%) when estimated using real-time reverse transcriptase-polymerase chain reaction and enzyme-linked immunosorbent assay respectively. Western blot analysis found quercetin to maintain cytosolic p65 protein levels to that seen in control. Quercetin was found to attenuate HG induced increased NF-κB and AP-1 DNA binding activity in electrophoretic mobility shift assay. Immunofluorescence studies revealed quercetin to prevent HG induced nuclear localization of p65 and c-jun. Quercetin was also found to decrease HG induced activation of NF-κB (71% ± 14%), AP-1 (69% ± 24%) and MCP-1 promoter (79% ± 25%) in EA.hy926 cells when analyzed using luciferase reporter assay. We conclude that quercetin attenuates MCP-1 expression in HG treated RAECs, probably by regulating both NF-κB and AP-1 pathways. The findings provide new insights into HG induced MCP-1 gene regulation in aortic endothelial cells and the potential of quercetin in abating the risk for atherosclerosis in diabetes

    Distinct Transcriptional Networks in Quiescent Myoblasts: A Role for Wnt Signaling in Reversible vs. Irreversible Arrest

    Get PDF
    <div><p>Most cells in adult mammals are non-dividing: differentiated cells exit the cell cycle permanently, but stem cells exist in a state of reversible arrest called quiescence. In damaged skeletal muscle, quiescent satellite stem cells re-enter the cell cycle, proliferate and subsequently execute divergent programs to regenerate both post-mitotic myofibers and quiescent stem cells. The molecular basis for these alternative programs of arrest is poorly understood. In this study, we used an established myogenic culture model (C2C12 myoblasts) to generate cells in alternative states of arrest and investigate their global transcriptional profiles. Using cDNA microarrays, we compared G<sub>0</sub> myoblasts with post-mitotic myotubes. Our findings define the transcriptional program of quiescent myoblasts in culture and establish that distinct gene expression profiles, especially of tumour suppressor genes and inhibitors of differentiation characterize reversible arrest, distinguishing this state from irreversibly arrested myotubes. We also reveal the existence of a tissue-specific quiescence program by comparing G<sub>0</sub> C2C12 myoblasts to isogenic G<sub>0</sub> fibroblasts (10T1/2). Intriguingly, in myoblasts but not fibroblasts, quiescence is associated with a signature of Wnt pathway genes. We provide evidence that different levels of signaling via the canonical Wnt pathway characterize distinct cellular states (proliferation vs. quiescence vs. differentiation). Moderate induction of Wnt signaling in quiescence is associated with critical properties such as clonogenic self-renewal. Exogenous Wnt treatment subverts the quiescence program and negatively affects clonogenicity. Finally, we identify two new quiescence-induced regulators of canonical Wnt signaling, Rgs2 and Dkk3, whose induction in G<sub>0</sub> is required for clonogenic self-renewal. These results support the concept that active signal-mediated regulation of quiescence contributes to stem cell properties, and have implications for pathological states such as cancer and degenerative disease.</p></div

    Chromatin-IP analysis of Myf5 and MyoG promoters in different cellular states.

    No full text
    <p>Antibodies against β-cat or HBP were used to assess the association of these Wnt-regulated transcription factors with chromatin in different cellular states as described in Materials and Methods. Control pulldowns used IgG and all values shown represent fold enrichment of the specific transcription factor after normalization against control IgG values. (<b>A</b>)<b>.</b> The Wnt target transcription factor β-cat associates with the known Wnt-responsive site in Myf5 enhancer preferentially in G<sub>0</sub> (G<sub>0</sub>) but shows low enrichment in either proliferating (MB) or differentiating muscle cells (MT) (Blue bars-MB; pink bars-G<sub>0</sub>; green bars-MT). This observation is consistent with the hypothesis that Wnt signaling is active in quiescent myoblasts. Comparison of β-cat association on another myogenic promoter (Myogenin promoter) shows greater enrichment in MT. Taken together, these observations suggest that Wnt/β-cat regulates different genes in different cellular states. (<b>B</b>)<b>.</b> ChIP analysis shows that HBP1 (a Wnt-induced repressor) co-associates with the Myf5 enhancer only in G<sub>0</sub> (Blue bars-Mb; pink bars-G<sub>0</sub>; green bars-MT) and does not associate with this element in either proliferating or differentiated muscle cells. This observation suggests that fine-tuning of Myf5 expression by both activating and repressive mechanisms may occur in quiescent cells by association of two types of Wnt-responsive transcription factors. Taken together, this observation would account for the absence of induction of Myf5 mRNA in quiescent myoblasts despite the association of the transcriptional activator β-cat.</p

    Moderate induction of Wnt signaling in two models of quiescence.

    No full text
    <p>(<b>A</b>) Wnt signaling revealed by TCF-dependent luciferase activity in stabily transfected TOPflash myoblast clone TFC1. Asynchronously proliferating myoblasts (As), suspension-arrested quiescent myoblasts (G<sub>0</sub>), suspension-synchronized myoblasts after re-activatation for 2–24 hrs (R2–R24), or myoblasts induced to differentiate for 72 hrs (MT). Transcriptional activity of the Wnt responsive TCF reporter is induced as MB enter quiescence and rapidly suppressed upon re-activation into the cell cycle. TCF-dependent luciferase activity (rlu/µg protein) is moderately induced in G<sub>0</sub> arrest but strongly induced in differentiation-associated arrest (MT, inset). Data represents mean +SE from atleast 3 independent experiments. (<b>B</b>) Conditioned medium (CM) from G<sub>0</sub> MB cultures contains more Wnt/TCF reporter-inducing activity than CM from proliferating or differentiated cultures. TOPflash reporter cells were exposed to CM derived from G<sub>0</sub> cultures (G<sub>0</sub>-CM), or proliferating cultures (Mb-CM), or differentiated cultures (Mt-CM). Fresh growth medium (GM) and differentiation medium (DM) were used as controls. Data represents mean +SE from 3 independent experiments. (<b>C</b>) Secreted Wnt agonist R-spondin expression is strongly induced in G<sub>0</sub>. RNA isolated from growing (Mb), arrested (G<sub>0</sub>) and differentiated (Mt) muscle cells was analysed by Q-RT-PCR (n = 3). (<b>D–F</b>) Wnt signaling is induced in an independent culture model of G<sub>0</sub> MB (reserve cells). (D) Phase contrast photographs of quiescent mononucleated undifferentiated reserve cells (RC) after differential trypsinization specifically removed myotubes (MT) in 5-day differentiated TFC1cultures. (E) RC isolated from away from MT do not induce MyoG (QRT-PCR analysis), confirming their undifferentiated state. (F) TOPflash TCF reporter activity is induced in purified quiescent reserve cells (G<sub>0</sub>rc) and its decline in reserve cells that have been reactivated by the addition of GM for 2–24 hrs (R2–R24). Inset shows TOPflash activity in G<sub>0</sub>rc compared to purified MT cultures depleted of reserve cells [MT(-rc)]. Data represents mean +SE from 3 independent experiments.</p

    C2C12 myoblasts enter alternate states of arrest.

    No full text
    <p>(<b>A</b>) Asynchronous proliferating myoblasts (MB) were held in suspension in mitogen-rich media to induce quiescence (arrest in an undifferentiated mono-nucleated state; G<sub>0</sub> MB), or shifted to mitogen-poor medium for 96 hrs to induce differentiation (arrest coupled with fusion into multinucleated myotubes; MT). Actin staining (Oregon Green-Phalloidin, top panel) reveals the distinct morphology of these 3 states. Nuclei are detected with Hoechst 33352. DNA synthesis was analyzed by detection of BrdU incorporated in a 15-minute pulse (middle panel)- ∼40% of cycling MB are labeled, while less than 1% of G<sub>0</sub> MB synthesize DNA. In differentiated cultures (MT), only residual mono-nucleated cycling cells (5–10%) incorporate label whereas myotube nuclei do not. Expression of determination factor MyoD can be detected in ∼50% of proliferating MB, is lost in G<sub>0</sub> but sustained in MT (lower panel). (<b>B</b>) FACS analysis of DNA content of cycling (Asynchronous MB) and 48-hr suspension-cultured populations confirms that >97% of cells in suspension (G<sub>0</sub> MB) possess a 2C DNA content while >40% of cells in an asynchronous culture have replicated their genomes (representative graphs from 4 independent experiments). (<b>C</b>) Muscle transcription factor expression distinguishes alternate states of arrest. Q-RT-PCR analysis of specification/survival factor Pax7, determination factor MyoD, and early differentiation marker Myogenin (MyoG) in asynchronous proliferating myoblasts (As), suspension-arrested MB (G<sub>0</sub>) and differentiated MT (MT). Values represent normalized fold differences between GAPDH (control) mRNA and myogenic mRNAs in each sample (n = 3). (<b>D</b>) Cell cycle inhibitor expression distinguishes alternate states of arrest-G<sub>0</sub> MB and MT express different combinations of cyclin-dependent kinase inhibitors p21/p27 and Rb-related p130. Immuno-detection of p21, p130 or p27 in MT and G<sub>0</sub>; p21 (green) co-localizes with MyoG (red) in all nuclei of differentiated MT but neither factor is expressed in G<sub>0</sub>; p130 is specific to G<sub>0</sub> and p27 is expressed in both G<sub>0</sub> and MT. Data depicted is representative of three independent experiments. (<b>E</b>) G<sub>0</sub> arrest is reversible. DNA synthesis in asynchronous MB (Mb), 48-hour suspension cultures (G<sub>0</sub>) and G<sub>0</sub> MB replated for 6 or 24 hours (R6, R24). BrdU detected as in Fig. 1A. Values represent mean<u>+</u>SEM (n = 3). (<b>F</b>) MyoD expression is suppressed in G<sub>0</sub> and restored during early reactivation. Asynchronous cultures (A) were arrested in G<sub>0</sub> (48 hrs) and reactivated by replating for 2 to 24 hrs (R2–R24). MyoD expression detected as in Fig. 1A. Values represent mean<u>+</u>SEM (n = 3).</p

    Exogenous Wnt treatment compromises a G0-induced program that promotes clonogenic potential.

    No full text
    <p>(A) Wnt3A treatment of MB reduces clonogenic potential. Colony formation was measured after 48 hrs in control culture conditions (either in proliferating conditions-Mb, or in suspension culture-G<sub>0</sub>), or in the presence of 50 ng/ml of rWnt3A. Cloning efficiency (a measure of self-renewal) was strongly reduced by Wnt3A supplementation and restored by simultaneous addition of 50ng/ml sFRP2. Values represent the mean±SEM from three independent experiments, <i>p</i><0.05 (denoted by asterisk *). (B) Knockdown of Rgs2 and Dkk3 transcripts using siRNAs. siRNAs were designed against the putative Wnt regulators Rgs2, Dkk3 or an irrelevant gene (GAPDH) or a control scrambled siRNA sequence and transfected into C2C12 myoblasts along with a GFP plasmid. GFP<b><sup>+</sup></b> transfected cells were enriched by FACS, RNA isolated and analysed by Q-RT-PCR and the relative mRNA levels calculated. In each pair, the mRNA level is depicted of cells transfected with scrambled siRNA (blue bars) and cells transfected with the targeting siRNA (pink bars). Values represent the mean and SEM of 3 independent experiments. In each case, modest but reproducible reduction of the target transcript level is observed. (C) Reduction of Rgs2 and Dkk3 protein expression by siRNA-mediated knockdown. Western blot analysis of total protein isolated from control and knockdown C2C12 muscle cells probed with antibodies against Rgs2 (top) and Dkk3 (bottom). GAPDH protein levels indicate equal loading. Data depicted is representative of 3 independent experiments. (D) Rgs2 and Dkk3 expression is necessary for Wnt signaling. Knockdown of either Rgs2 or Dkk3 in growing or quiescent MB leads to suppression of TOPflash activity. Cells were treated and enriched as described in (B) and luciferase activity measured. Despite modest reduction of protein levels, strong reduction in TOPflash activity are seen, indicating a critical role for Rgs2 and Dkk3 in Wnt-βcat signaling. Values represent the mean and SEM of 3 independent experiments. (E,F) Knockdown cells (‘Rgs sh’ and ‘Dkk sh’) were enriched as described in (B) cultured in quiescence-inducing conditions, recovered from suspension culture and plated at clonogenic density for assessment of self-renewal (colony formation). Controls include untransfected cells (‘UT’) and control shRNA transfected cells (‘Con sh’). Typical plates with colony assays are shown in (E) and data are quantified as CFU (colony forming units) in (F). Values represent the mean and SEM of 3 independent experiments.</p

    Microarray analysis reveals distinct genetic programs in reversible and irreversible arrest.

    No full text
    <p>(<b>A</b>) Schematic of microarray loop experiment. RNA isolated from G<sub>0</sub> MB was analyzed by 2-color competitive hybridization to NIA15K mouse cDNA arrays. Pair-wise analysis: G<sub>0</sub> MB vs. either proliferating MB, differentiated MT or G<sub>0</sub> FB. In each pair, genes consistently up- or down-regulated (>1.6 fold, normalized log ratio (NLR) 0.67, false discovery rate 4%) were selected for further analysis. The numbers of genes regulated between each pair of samples are shown: induced (red), suppressed (green). Expression of key regulators characteristic of the different samples is indicated, confirming the identity of each cellular state. (B) Hierarchical clustering (MEV software, TIGR) shows the relatedness of the myoblast and fibroblast sample pairs drawn from independent biological experiments including a dye reversal test. Based on the total transcriptional response, genes up-regulated in G<sub>0</sub> MB fall into 3 broad categories-genes specifically induced in G<sub>0</sub> MB (the largest number), genes induced in both G<sub>0</sub> MB and MT but down-regulated in G<sub>0</sub> FB, and genes induced in G<sub>0</sub> MB and G<sub>0</sub> FB but not in MT. (<b>C</b>) In muscle cells, common features of two types of cell cycle exit (reversible vs irreversible) are overshadowed by differences. Venn diagram reveals minimal overlap between the three myogenic sample pairs (MB vs G<sub>0</sub> MB, MB vs MT, G<sub>0</sub> MB vs MT), and show that distinct transcriptional programs characterize reversible arrest vs. differentiation-coupled irreversible arrest. (D) Validation of microarray analysis using Northern blots. RNA samples used for microarray analysis were also interrogated with <sup>32</sup>P-labelled probes representing genes selected to test the range of quantitative changes in expression (i) genes that showed no change (log ratio of 0), (ii) genes that were down-regulated in G<sub>0</sub> (negative log ratio) or (iii) genes that were up regulated in G<sub>0</sub> (positive log ratio). Numbers on the left of blots represent log ratios derived from G<sub>0</sub> vs. asynchronous myoblast array. Gene symbols & transcript sizes are shown on the right of each blot. 28S rRNA was used as a loading control. Rfg, a gene expressed equally in all conditions. Cofilin, a regulator of microfilament dynamics - down-regulated in both G<sub>0</sub> Mb and in Mt. Of the 12 transcripts selected from the array as enriched in G<sub>0</sub>, all 12 showed G0 induced expression. Of these, 8 (FGFR1, BMP1, GSTa4, TM7SF1, IGFBP5, TIMP3, Cathepsin L, Col3A1) were specifically induced in G<sub>0</sub> MB, and not in differentiated MT. The other 4 genes, IGF2, Rho GAP, LAMP2 and Map1LC3b were induced in both G<sub>0</sub> MB and MT. Histone 2B and MyoD transcripts (not derived from array analysis, marked by *) were detected to confirm the cellular state. E. Functional diversity of genes specifically enriched in G<sub>0</sub> MB indicates a signature of <b><i>recycling</i></b> (autophagy, ubiquitin pathway), <b><i>resistance</i></b> (anti-apoptosis) and <b><i>repair</i></b> (DNA repair). Gene ontology classification of 663 annotated gene from the list of genes upregulated in quiescent (G0) MB suggests that enhanced survival mechanisms may compensate for depressed metabolic functions in quiescence (see Supplementary Information for altered cell cycle and metabolic pathways). In addition, there is evidence of altered expression of genome reprogramming factors (chromatin and transcriptional regulators), signaling (growth factors, GF receptors, signaling adaptors) and developmental regulators [Wnt (indicated by arrow), Hh, BMP, Notch pathways)]. The surprising increase in induction of Wnt genes in quiescence was further investigated.</p
    corecore