18 research outputs found

    Metabolic Checkpoints in Cancer Cell Cycle

    Full text link
    Growth factors (GFs) as well as nutrient sufficiency regulate cell division in metazoans. The vast majority of mutations that contribute to cancer are in genes that regulate progression through the G1 phase of the cell cycle. A key regulatory site in G1 is the growth factor-dependent Restriction Point (R), where cells get permissive signals to divide. In the absence of GF instructions, cells enter the quiescent G0 state. Despite fundamental differences between GF signaling and nutrient sensing, they both have been confusingly referred to as R and therefore by definition considered to be a singular event in G1. Autonomy from GF signaling is one of the hallmarks in cancer; however, cancer cells also have metabolic rewiring enabling them to engage in anabolic biosynthetic pathways. In the absence of GF instructions and nutrients, cells commonly undergo apoptotic cell death. Thus, it is of importance to elucidate the differences between GF and nutrient deregulation in cancer to develop novel strategies in targeting tumor cell proliferation and survival. Here, we report that the GF-mediated mid-G1 restriction point (R) is distinct and distinguishable from a series of late-G1 metabolic checkpoints mediated by essential amino acids, conditionally essential amino acid - glutamine, and mTOR - the mammalian target of rapamycin. Our data indicate that the arrest sites mediated by various blocking conditions are in the order of GF -\u3e EAA -\u3e Q -\u3e mTOR. We temporally mapped the EAA and glutamine checkpoints at 12 hr from G0 and mTOR mediated arrest occurring at 16 hr from G0. Distinct profiles for cell cycle regulator expression and phosphorylation was observed when released from restriction point relative to the metabolic checkpoints. These data are consistent with a mid-G1 R where cells decide whether they should divide, followed by late-G1 metabolic checkpoints where cells determine whether they have sufficient nutrients to divide. Since mTOR inhibition using rapamycin or Torin1 arrested the cells latest in G1, mTOR may serve as the final arbiter for nutrient sufficiency prior to replicating the genome. Significantly we also observed that in addition to GF autonomy, several cancer cells also have dysregulated nutritional sensing, and arrest in S- and G2/M phase upon essential amino acid and glutamine deprivation. We identified K-Ras mutation as the underlying genetic cause for this phenomenon. We found that treating cancer cells harboring K-Ras mutation with aminooxyacetate (AOA) - drug that interferes with glutamine utilization - causes them to arrest in S- and G2/M-phase, where synthetic lethality could be created to phase-specific cytotoxic drugs. Thus, besides addressing the long standing assumption of GF and nutrients regulating G1 cell cycle progression, our work provides rationale and proof of principle for targeting metabolic deregulations in cancer cells

    Blocking anaplerotic entry of glutamine to TCA cycle sensitizes K-Ras mutant cancer cells to cytotoxic drugs

    Full text link
    Cancer cells undergo a metabolic transformation that allows for increased anabolic demands wherein glycolytic and TCA cycle intermediates are shunted away for the synthesis of biological molecules required for cell growth and division. One of the key shunts is the exit of citrate from the mitochondria and the TCA cycle for the generation of cytosolic acetyl-CoA that can be used for fatty acid and cholesterol biosynthesis. With the loss of mitochondrial citrate, cancer cells rely on the “conditionally essential” amino acid glutamine (Q) as an anaplerotic carbon source for TCA cycle intermediates. While Q deprivation causes G1 cell cycle arrest in non-transformed cells, its impact on the cancer cell cycle is not well characterized. We report here a correlation between bypass of the Q-dependent G1 checkpoint and cancer cells harboring K-Ras mutations. Instead of arresting in G1 in response to Q-deprivation, K-Ras driven cancer cells arrest in either S- or G2/M-phase. Inhibition of K-Ras effector pathways was able to revert cells to G1 arrest upon Q deprivation. Blocking anaplerotic utilization of Q mimicked Q deprivation – causing S- and G2/M-phase arrest in K-Ras mutant cancer cells. Significantly, Q deprivation or suppression of anaplerotic Q utilization created synthetic lethality to the cell cycle phase-specific cytotoxic drugs, capecitabine and paclitaxel. These data suggest that disabling of the G1 Q checkpoint could represent a novel vulnerability of cancer cells harboring K-Ras and possibly other mutations that disable the Q-dependent checkpoint

    Amino Acids and mTOR Mediate Distinct Metabolic Checkpoints in Mammalian G1 Cell Cycle

    Get PDF
    Objective In multicellular organisms, cell division is regulated by growth factors (GFs). In the absence of GFs, cells exit the cell cycle at a site in G1 referred to as the restriction point (R) and enter a state of quiescence known as G0. Additionally, nutrient availability impacts on G1 cell cycle progression. While there is a vast literature on G1 cell cycle progression, confusion remains – especially with regard to the temporal location of R relative to nutrient-mediated checkpoints. In this report, we have investigated the relationship between R and a series of metabolic cell cycle checkpoints that regulate passage into S-phase. Methods We used double-block experiments to order G1 checkpoints that monitor the presence of GFs, essential amino acids (EEAs), the conditionally essential amino acid glutamine, and inhibition of mTOR. Cell cycle progression was monitored by uptake of [3H]-thymidine and flow cytometry, and analysis of cell cycle regulatory proteins was by Western-blot. Results We report here that the GF-mediated R can be temporally distinguished from a series of late G1 metabolic checkpoints mediated by EAAs, glutamine, and mTOR – the mammalian/mechanistic target of rapamycin. R is clearly upstream from an EAA checkpoint, which is upstream from a glutamine checkpoint. mTOR is downstream from both the amino acid checkpoints, close to S-phase. Significantly, in addition to GF autonomy, we find human cancer cells also have dysregulated metabolic checkpoints. Conclusion The data provided here are consistent with a GF-dependent mid-G1 R where cells determine whether it is appropriate to divide, followed by a series of late-G1 metabolic checkpoints mediated by amino acids and mTOR where cells determine whether they have sufficient nutrients to accomplish the task. Since mTOR inhibition arrests cells the latest in G1, it is likely the final arbiter for nutrient sufficiency prior to committing to replicating the genome

    Drosophila insulin and target of rapamycin (TOR) pathways regulate GSK3 beta activity to control Myc stability and determine Myc expression in vivo

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Genetic studies in <it>Drosophila melanogaster </it>reveal an important role for Myc in controlling growth. Similar studies have also shown how components of the insulin and target of rapamycin (TOR) pathways are key regulators of growth. Despite a few suggestions that Myc transcriptional activity lies downstream of these pathways, a molecular mechanism linking these signaling pathways to Myc has not been clearly described. Using biochemical and genetic approaches we tried to identify novel mechanisms that control Myc activity upon activation of insulin and TOR signaling pathways.</p> <p>Results</p> <p>Our biochemical studies show that insulin induces Myc protein accumulation in <it>Drosophila </it>S2 cells, which correlates with a decrease in the activity of glycogen synthase kinase 3-beta (GSK3<it>β </it>) a kinase that is responsible for Myc protein degradation. Induction of Myc by insulin is inhibited by the presence of the TOR inhibitor rapamycin, suggesting that insulin-induced Myc protein accumulation depends on the activation of TOR complex 1. Treatment with amino acids that directly activate the TOR pathway results in Myc protein accumulation, which also depends on the ability of S6K kinase to inhibit GSK3<it>β </it>activity. Myc upregulation by insulin and TOR pathways is a mechanism conserved in cells from the wing imaginal disc, where expression of Dp110 and Rheb also induces Myc protein accumulation, while inhibition of insulin and TOR pathways result in the opposite effect. Our functional analysis, aimed at quantifying the relative contribution of Myc to ommatidial growth downstream of insulin and TOR pathways, revealed that Myc activity is necessary to sustain the proliferation of cells from the ommatidia upon Dp110 expression, while its contribution downstream of TOR is significant to control the size of the ommatidia.</p> <p>Conclusions</p> <p>Our study presents novel evidence that Myc activity acts downstream of insulin and TOR pathways to control growth in <it>Drosophila</it>. At the biochemical level we found that both these pathways converge at GSK3<it>β </it>to control Myc protein stability, while our genetic analysis shows that insulin and TOR pathways have different requirements for Myc activity during development of the eye, suggesting that Myc might be differentially induced by these pathways during growth or proliferation of cells that make up the ommatidia.</p

    Co-inhibition of SMAD and MAPK signaling enhances 124I uptake in BRAF-mutant thyroid cancers.

    Get PDF
    Constitutive MAPK activation silences genes required for iodide uptake and thyroid hormone biosynthesis in thyroid follicular cells. Accordingly, most BRAFV600E papillary thyroid cancers (PTC) are refractory to radioiodide (RAI) therapy. MAPK pathway inhibitors rescue thyroid-differentiated properties and RAI responsiveness in mice and patient subsets with BRAFV600E-mutant PTC. TGFB1 also impairs thyroid differentiation and has been proposed to mediate the effects of mutant BRAF. We generated a mouse model of BRAFV600E-PTC with thyroid-specific knockout of the Tgfbr1 gene to investigate the role of TGFB1 on thyroid-differentiated gene expression and RAI uptake in vivo. Despite appropriate loss of Tgfbr1, pSMAD levels remained high, indicating that ligands other than TGFB1 were engaging in this pathway. The activin ligand subunits Inhba and Inhbb were found to be overexpressed in BRAFV600E-mutant thyroid cancers. Treatment with follistatin, a potent inhibitor of activin, or vactosertib, which inhibits both TGFBR1 and the activin type I receptor ALK4, induced a profound inhibition of pSMAD in BRAFV600E-PTCs. Blocking SMAD signaling alone was insufficient to enhance iodide uptake in the setting of constitutive MAPK activation. However, combination treatment with either follistatin or vactosertib and the MEK inhibitor CKI increased 124I uptake compared to CKI alone. In summary, activin family ligands converge to induce pSMAD in Braf-mutant PTCs. Dedifferentiation of BRAFV600E-PTCs cannot be ascribed primarily to activation of SMAD. However, targeting TGFβ/activin-induced pSMAD augmented MAPK inhibitor effects on iodine incorporation into BRAF tumor cells, indicating that these two pathways exert interdependent effects on the differentiation state of thyroid cancer cells

    Restriction point and metabolic checkpoint arrest lead to differential patterns of cell cycle regulator expression and phosphorylation.

    No full text
    <p>(<b>A</b>) Cells were plated at 30% confluence in 10-cm plates in DMEM containing 10% FBS. After 24 hr, the cells were shifted to CM or blocking conditions for 4 hr, at which time the cells were harvested and the levels of the indicated protein or phosphoprotein was determined by Western blot analysis. The data shown are representative of experiments repeated at least two times. (<b>B</b>) Quantitative analysis of relative protein levels for Western blots shown in (<b>A</b>) using ImageJ software. (<b>C</b>–<b>F</b>) BJ cells were plated and shifted to various blocking conditions for 48 hr as in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0074157#pone-0074157-g001" target="_blank">Figure 1A</a>. The cells were subsequently released by shifting to CM, and the cells were harvested and lysates collected at indicated time points. The levels of the indicated protein or phosphoprotein were determined by Western blot analysis. The data shown are representative of experiments repeated at least two times. Also shown in the line graphs are the kinetic analyses of relative protein levels normalized to actin and quantitated using ImageJ.</p

    GF, EAA, Q, and rapamycin mediated G1 cell cycle arrests are distinct and distinguishable.

    No full text
    <p>(<b>A</b>–<b>D</b>) BJ hTERT cells were plated and shifted to various first blocking conditions for 48 hr as in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0074157#pone-0074157-g001" target="_blank">Figure 1A</a>. The cells were subsequently shifted to CM or different second block conditions containing [<sup>3</sup>H]-TdR for 24 hr, after which the cells were collected and the incorporated label was determined. Error bars represent the standard error for the experiment repeated at least four times. (<b>E</b>) Schematic model showing relative positions of different metabolic checkpoints relative to R (not drawn to represent precise time scales). G1-pm is post-mitotic phase in G1, G1-ps is pre-S phase of G1 [<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0074157#B4" target="_blank">4</a>].</p
    corecore