47 results on '"Avizonis D"'
Search Results
2. High Sensitivity of an Ha-RAS Transgenic Model of Superficial Bladder Cancer to Metformin Is Associated with  240-Fold Higher Drug Concentration in Urine than Serum
- Author
-
Liu, Z., Yokoyama, N. N, Blair, C. A, Li, X., Avizonis, D., Wu, X.-R., Uchio, E., Youssef, R., McClelland, M., Pollak, M., and Zi, X.
- Published
- 2016
3. Chromatin, TAFs, and a novel multiprotein coactivator are required for synergistic activation by Sp1 and SREBP-1a in vitro.
- Author
-
Näär, A M, Beaurang, P A, Robinson, K M, Oliner, J D, Avizonis, D, Scheek, S, Zwicker, J, Kadonaga, J T, and Tjian, R
- Abstract
The promoter selectivity factor Sp1 often cooperates with other enhancer-binding proteins to activate transcription. To study the molecular underpinnings of these regulatory events, we have reconstituted in vitro the synergy observed in vivo between Sp1 and the sterol-regulated factor SREBP-1a at the low density lipoprotein receptor (LDLR) promoter. Using a highly purified human transcription system, we found that chromatin, TAFs, and a novel SREBP-binding coactivator activity, which includes CBP, are all required to mediate full synergistic activation by Sp1 and SREBP-1a. The SREBP-binding domain of CBP inhibits activation by SREBP-1a and Sp1 in a dominant-negative fashion that is both chromatin- and activator-specific. Whereas recombinant CBP alone is not sufficient to mediate activation, a human cellular fraction containing CBP can support high levels of chromatin-dependent synergistic activation. Purification of this activity to near homogeneity resulted in the identification of a multiprotein coactivator, including CBP, that selectively binds to the SREBP-1a activation domain and is capable of mediating high levels of synergistic activation by SREBP/Sp1 on chromatin templates. The development of a reconstituted chromatin transcription system has allowed us to isolate a novel coactivator that is recruited by the SREBP-1a activation domain and that functions in concert with TFIID to coordinate the action of multiple activators at complex promoters in the context of chromatin.
- Published
- 1998
4. Solid phase synthesis of 5-hydroxymethyluracil containing DNA
- Author
-
Maria R. Conte, Victor L. Hsu, Daina Avizonis, Luciano Mayol, Aldo Galeone, David R. Kearns, Conte, M. R., Galeone, Aldo, Avizonis, D., Hsu, V. L., Mayol, Luciano, and Kearns, D. R.
- Subjects
chemistry.chemical_compound ,5-HYDROXYMETHYL-2'-DEOXYURIDINE, LEUKEMIA, SEQUENCE, BINDING PROTEIN ,Solid-phase synthesis ,Chemistry ,Organic Chemistry ,Clinical Biochemistry ,Drug Discovery ,Pharmaceutical Science ,Molecular Medicine ,Molecular Biology ,Biochemistry ,Combinatorial chemistry ,DNA - Abstract
The synthesis of 3′-O-(diisopropylamino-2-cyanoethoxyphosphinyl)-5′-O-(4,4′-diemthoxytriyl)-5-( tert -butyldimethylsiloxymethyl)-2′-deoxyuridine ( 5 ) and its utilization for the preparation of 5-hydroxymethyluracil (hmU) containing oligodeoxyribonucleotides by means of automated synthesis are described.
- Published
- 1992
5. Metastatic breast cancer cells are metabolically reprogrammed to maintain redox homeostasis during metastasis.
- Author
-
Biondini M, Lehuédé C, Tabariès S, Annis MG, Pacis A, Ma EH, Tam C, Hsu BE, Audet-Delage Y, Abu-Thuraia A, Girondel C, Sabourin V, Totten SP, de Sá Tavares Russo M, Bridon G, Avizonis D, Guiot MC, St-Pierre J, Ursini-Siegel J, Jones R, and Siegel PM
- Subjects
- Female, Humans, Animals, Mice, Cell Line, Tumor, Glycolysis, Neoplasm Metastasis, Gene Expression Regulation, Neoplastic, Tumor Microenvironment, Breast Neoplasms pathology, Breast Neoplasms metabolism, Breast Neoplasms genetics, Oxidation-Reduction, Glutathione metabolism, Reactive Oxygen Species metabolism, Liver Neoplasms metabolism, Liver Neoplasms pathology, Liver Neoplasms secondary, Liver Neoplasms genetics, Glutamate-Cysteine Ligase metabolism, Glutamate-Cysteine Ligase genetics, Homeostasis
- Abstract
Metabolic rewiring is essential for tumor growth and progression to metastatic disease, yet little is known regarding how cancer cells modify their acquired metabolic programs in response to different metastatic microenvironments. We have previously shown that liver-metastatic breast cancer cells adopt an intrinsic metabolic program characterized by increased HIF-1α activity and dependence on glycolysis. Here, we confirm by in vivo stable isotope tracing analysis (SITA) that liver-metastatic breast cancer cells retain a glycolytic profile when grown as mammary tumors or liver metastases. However, hepatic metastases exhibit unique metabolic adaptations including elevated expression of genes involved in glutathione (GSH) biosynthesis and reactive oxygen species (ROS) detoxification when compared to mammary tumors. Accordingly, breast-cancer-liver-metastases exhibited enhanced de novo GSH synthesis. Confirming their increased capacity to mitigate ROS-mediated damage, liver metastases display reduced levels of 8-Oxo-2'-deoxyguanosine. Depletion of the catalytic subunit of the rate-limiting enzyme in glutathione biosynthesis, glutamate-cysteine ligase (GCLC), strongly reduced the capacity of breast cancer cells to form liver metastases, supporting the importance of these distinct metabolic adaptations. Loss of GCLC also affected the early steps of the metastatic cascade, leading to decreased numbers of circulating tumor cells (CTCs) and impaired metastasis to the liver and the lungs. Altogether, our results indicate that GSH metabolism could be targeted to prevent the dissemination of breast cancer cells., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
6. Targeting fatty acid oxidation enhances response to HER2-targeted therapy.
- Author
-
Nandi I, Ji L, Smith HW, Avizonis D, Papavasiliou V, Lavoie C, Pacis A, Attalla S, Sanguin-Gendreau V, and Muller WJ
- Subjects
- Animals, Female, Humans, Mice, Cell Line, Tumor, Oxidative Stress, Tumor Microenvironment drug effects, Diet, Ketogenic, Cell Proliferation drug effects, Apoptosis drug effects, Glucose metabolism, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Lung Neoplasms secondary, Lung Neoplasms genetics, Lung Neoplasms pathology, Receptor, ErbB-2 metabolism, Receptor, ErbB-2 genetics, Receptor, ErbB-2 antagonists & inhibitors, Fatty Acids metabolism, Carnitine O-Palmitoyltransferase metabolism, Carnitine O-Palmitoyltransferase genetics, Oxidation-Reduction, Breast Neoplasms pathology, Breast Neoplasms metabolism, Breast Neoplasms drug therapy, Breast Neoplasms genetics, NF-E2-Related Factor 2 metabolism, NF-E2-Related Factor 2 genetics
- Abstract
Metabolic reprogramming, a hallmark of tumorigenesis, involves alterations in glucose and fatty acid metabolism. Here, we investigate the role of Carnitine palmitoyl transferase 1a (Cpt1a), a key enzyme in long-chain fatty acid (LCFA) oxidation, in ErbB2-driven breast cancers. In ErbB2+ breast cancer models, ablation of Cpt1a delays tumor onset, growth, and metastasis. However, Cpt1a-deficient cells exhibit increased glucose dependency that enables survival and eventual tumor progression. Consequently, these cells exhibit heightened oxidative stress and upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) activity. Inhibiting Nrf2 or silencing its expression reduces proliferation and glucose consumption in Cpt1a-deficient cells. Combining the ketogenic diet, composed of LCFAs, or an anti-ErbB2 monoclonal antibody (mAb) with Cpt1a deficiency significantly perturbs tumor growth, enhances apoptosis, and reduces lung metastasis. Using an immunocompetent model, we show that Cpt1a inhibition promotes an antitumor immune microenvironment, thereby enhancing the efficacy of anti-ErbB2 mAbs. Our findings underscore the importance of targeting fatty acid oxidation alongside HER2-targeted therapies to combat resistance in HER2+ breast cancer patients., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Tourniquet Use and Local Tissue Concentrations of Cefazolin During Total Knee Arthroplasty: A Randomized Clinical Trial.
- Author
-
Montreuil J, Tanzer M, Zhang YL, Rajda E, Avizonis D, and Hart A
- Subjects
- Humans, Female, Male, Aged, Middle Aged, Prospective Studies, Prosthesis-Related Infections prevention & control, Antibiotic Prophylaxis methods, Surgical Wound Infection prevention & control, Cefazolin pharmacokinetics, Cefazolin administration & dosage, Cefazolin blood, Arthroplasty, Replacement, Knee, Tourniquets, Anti-Bacterial Agents administration & dosage, Anti-Bacterial Agents pharmacokinetics, Anti-Bacterial Agents analysis, Anti-Bacterial Agents therapeutic use
- Abstract
Importance: Prophylactic administration of antibiotics before skin incision is an important component in the prevention of periprosthetic joint infection in arthroplasty surgery. For antibiotics to be effective, the local tissue concentration (LTC) must exceed the minimum inhibitory concentration of typical infecting organisms; however, the LTC of cefazolin during arthroplasty is poorly understood., Objective: To compare the systemic concentration of cefazolin in serum with the LTC in fat, synovium, and bone during primary total knee arthroplasty (TKA) while assessing the effect of tourniquet inflation., Design, Setting, and Participants: This prospective randomized clinical trial was conducted from March 1, 2022, to June 30, 2023, in patients undergoing TKA at a single academic center., Intervention: Total knee arthroplasty with or without a limb tourniquet., Main Outcomes and Measures: Systemic blood and local tissues from the surgical site (fat, synovium, and bone) were harvested at regular intervals during the surgery. The primary outcome was the LTC of cefazolin, quantified using the liquid chromatography-tandem mass spectrometry technique., Results: A total of 59 patients were included in the study, with 29 in the tourniquet group (mean [SD] age, 69.3 [9.6] years; 23 [79.3%] female) and 30 in the no tourniquet group (mean [SD] age, 69.9 [9.7] years; 21 [70.0%] female). In patients undergoing TKA without a tourniquet, the mean concentration of cefazolin in serum was 71.9 μg/mL (95% CI, 66.4-77.5 μg/mL), whereas the mean LTCs were 13.9 μg/g (95% CI, 12.1-15.7 μg/g) in fat, 27.7 μg/g (95% CI, 24.3-31.0 μg/g) in synovium, and 17.7 μg/g (95% CI, 14.8-20.5 μg/g) in bone. For patients undergoing TKA with a tourniquet, the mean concentration of cefazolin in serum was 72.0 μg/mL (95% CI, 66.3-77.7 μg/mL), and the mean LTCs were 9.9 μg/g (95% CI, 8.7-11.1 μg/g) in fat, 21.8 μg/g (95% CI, 18.7-25.0 μg/g) in synovium, and 13.0 μg/g (95% CI, 10.8-15.2 μg/g) in bone. The use of a tourniquet resulted in significantly lower mean LTCs by 60 minutes after cefazolin infusion (10.8 μg/g [95% CI, 9.1-12.4 μg/g] vs 16.9 μg/g [95% CI, 14.1-19.6 μg/g], P = .001 in fat; 18.9 μg/g [95% CI, 14.1-23.6 μg/g] vs 25.8 μg/g [95% CI, 21.4-30.3 μg/g], P = .03 in synovium; and 11.8 μg/g [95% CI, 9.3-14.2 μg/g] vs 19.4 μg/g [95% CI, 14.5-24.4 μg/g], P = .007 in bone)., Conclusions and Relevance: In this randomized clinical trial, the concentration of cefazolin was lower in local tissues (fat, synovium, and bone) than in systemic blood, and the use of a limb tourniquet further significantly reduced these concentrations. Although the current prophylactic dosing regimen for cefazolin provides sufficient serum concentrations, the levels in the periarticular tissue during TKA may be insufficient to prevent periprosthetic joint infection., Trial Registration: ClinicalTrials.gov Identifier: NCT05604157.
- Published
- 2024
- Full Text
- View/download PDF
8. p66ShcA promotes malignant breast cancer phenotypes by alleviating energetic and oxidative stress.
- Author
-
Lewis K, La Selva R, Maldonado E, Annis MG, Najyb O, Cepeda Cañedo E, Totten S, Hébert S, Sabourin V, Mirabelli C, Ciccolini E, Lehuédé C, Choinière L, Russo M, Avizonis D, Park M, St-Pierre J, Kleinman CL, Siegel PM, and Ursini-Siegel J
- Subjects
- Humans, Female, Shc Signaling Adaptor Proteins genetics, Shc Signaling Adaptor Proteins metabolism, Src Homology 2 Domain-Containing, Transforming Protein 1 metabolism, Oxidative Stress physiology, Phenotype, Cell Line, Tumor, Tumor Microenvironment, Breast Neoplasms metabolism
- Abstract
Significant efforts have focused on identifying targetable genetic drivers that support the growth of solid tumors and/or increase metastatic ability. During tumor development and progression to metastatic disease, physiological and pharmacological selective pressures influence parallel adaptive strategies within cancer cell sub-populations. Such adaptations allow cancer cells to withstand these stressful microenvironments. This Darwinian model of stress adaptation often prevents durable clinical responses and influences the emergence of aggressive cancers with increased metastatic fitness. However, the mechanisms contributing to such adaptive stress responses are poorly understood. We now demonstrate that the p66ShcA redox protein, itself a ROS inducer, is essential for survival in response to physiological stressors, including anchorage independence and nutrient deprivation, in the context of poor outcome breast cancers. Mechanistically, we show that p66ShcA promotes both glucose and glutamine metabolic reprogramming in breast cancer cells, to increase their capacity to engage catabolic metabolism and support glutathione synthesis. In doing so, chronic p66ShcA exposure contributes to adaptive stress responses, providing breast cancer cells with sufficient ATP and redox balance needed to withstand such transient stressed states. Our studies demonstrate that p66ShcA functionally contributes to the maintenance of aggressive phenotypes and the emergence of metastatic disease by forcing breast tumors to adapt to chronic and moderately elevated levels of oxidative stress., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. MTOR modulation induces selective perturbations in histone methylation which influence the anti-proliferative effects of mTOR inhibitors.
- Author
-
Kim H, Lebeau B, Papadopoli D, Jovanovic P, Russo M, Avizonis D, Morita M, Afzali F, Ursini-Siegel J, Postovit LM, Witcher M, and Topisirovic I
- Abstract
Emerging data suggest a significant cross-talk between metabolic and epigenetic programs. However, the relationship between the mechanistic target of rapamycin (mTOR), which is a pivotal metabolic regulator, and epigenetic modifications remains poorly understood. Our results show that mTORC1 activation caused by the abrogation of its negative regulator tuberous sclerosis complex 2 (TSC2) coincides with increased levels of the histone modification H3K27me3 but not H3K4me3 or H3K9me3. This selective H3K27me3 induction was mediated via 4E-BP-dependent increase in EZH2 protein levels. Surprisingly, mTOR inhibition also selectively induced H3K27me3. This was independent of TSC2, and was paralleled by reduced EZH2 and increased EZH1 protein levels. Notably, the ability of mTOR inhibitors to induce H3K27me3 levels was positively correlated with their anti-proliferative effects. Collectively, our findings demonstrate that both activation and inhibition of mTOR selectively increase H3K27me3 by distinct mechanisms, whereby the induction of H3K27me3 may potentiate the anti-proliferative effects of mTOR inhibitors., Competing Interests: The authors declare no conflict of interest., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Methionine oxidation activates pyruvate kinase M2 to promote pancreatic cancer metastasis.
- Author
-
He D, Feng H, Sundberg B, Yang J, Powers J, Christian AH, Wilkinson JE, Monnin C, Avizonis D, Thomas CJ, Friedman RA, Kluger MD, Hollingsworth MA, Grandgenett PM, Klute KA, Toste FD, Chang CJ, and Chio IIC
- Subjects
- Humans, Methionine, Methionine Sulfoxide Reductases chemistry, Methionine Sulfoxide Reductases metabolism, Oxidation-Reduction, Pyruvate Kinase metabolism, Thyroid Hormone-Binding Proteins, Pancreatic Neoplasms, Carcinoma, Pancreatic Ductal genetics, Carrier Proteins metabolism, Membrane Proteins metabolism, Pancreatic Neoplasms genetics, Thyroid Hormones metabolism
- Abstract
Cancer mortality is primarily a consequence of its metastatic spread. Here, we report that methionine sulfoxide reductase A (MSRA), which can reduce oxidized methionine residues, acts as a suppressor of pancreatic ductal adenocarcinoma (PDA) metastasis. MSRA expression is decreased in the metastatic tumors of PDA patients, whereas MSRA loss in primary PDA cells promotes migration and invasion. Chemoproteomic profiling of pancreatic organoids revealed that MSRA loss results in the selective oxidation of a methionine residue (M239) in pyruvate kinase M2 (PKM2). Moreover, M239 oxidation sustains PKM2 in an active tetrameric state to promote respiration, migration, and metastasis, whereas pharmacological activation of PKM2 increases cell migration and metastasis in vivo. These results demonstrate that methionine residues can act as reversible redox switches governing distinct signaling outcomes and that the MSRA-PKM2 axis serves as a regulatory nexus between redox biology and cancer metabolism to control tumor metastasis., Competing Interests: Declaration of interests C.J.C., F.D.T., and A.H.C. are inventors on patent applications related to the redox-active reagents for methionine conjugation. C.J.T. is listed as an inventor on patents related to PKM2 activators. The remaining authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
11. Mitochondrial hyperfusion via metabolic sensing of regulatory amino acids.
- Author
-
Abdullah MO, Zeng RX, Margerum CL, Papadopoli D, Monnin C, Punter KB, Chu C, Al-Rofaidi M, Al-Tannak NF, Berardi D, Rattray Z, Rattray NJW, Abraham SA, Eskelinen EL, Watson DG, Avizonis D, Topisirovic I, and Chan EYW
- Subjects
- Amines metabolism, Mitochondria metabolism, Mitochondrial Proteins metabolism, Purines metabolism, Amino Acids metabolism, Mitochondrial Dynamics
- Abstract
The relationship between nutrient starvation and mitochondrial dynamics is poorly understood. We find that cells facing amino acid starvation display clear mitochondrial fusion as a means to evade mitophagy. Surprisingly, further supplementation of glutamine (Q), leucine (L), and arginine (R) did not reverse, but produced stronger mitochondrial hyperfusion. Interestingly, the hyperfusion response to Q + L + R was dependent upon mitochondrial fusion proteins Mfn1 and Opa1 but was independent of MTORC1. Metabolite profiling indicates that Q + L + R addback replenishes amino acid and nucleotide pools. Inhibition of fumarate hydratase, glutaminolysis, or inosine monophosphate dehydrogenase all block Q + L + R-dependent mitochondrial hyperfusion, which suggests critical roles for the tricarboxylic acid (TCA) cycle and purine biosynthesis in this response. Metabolic tracer analyses further support the idea that supplemented Q promotes purine biosynthesis by serving as a donor of amine groups. We thus describe a metabolic mechanism for direct sensing of cellular amino acids to control mitochondrial fusion and cell fate., Competing Interests: Declarations of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
12. Autophagy-dependent glutaminolysis drives superior IL21 production in HIV-1-specific CD4 T cells.
- Author
-
Loucif H, Dagenais-Lussier X, Avizonis D, Choinière L, Beji C, Cassin L, Routy JP, Fritz JH, Olagnier D, and van Grevenynghe J
- Subjects
- Adenosine Monophosphate, Antiviral Agents pharmacology, CD4-Positive T-Lymphocytes, Humans, Interleukins, Leukocytes, Mononuclear, Protein Kinases, Autophagy physiology, HIV-1
- Abstract
The maintenance of a strong IL21 production in memory CD4 T cells, especially in HIV-1-specific cells, represents a major correlate of natural immune protection against the virus. However, the molecular mechanisms underlying IL21 production during HIV-1 infection, which is only elevated among the naturally protected elite controllers (EC), are still unknown. We recently found out that lipophagy is a critical immune mediator that control an antiviral metabolic state following CD8A T cell receptor engagement, playing an important role in the natural control of HIV-1 infection. This led us to investigate whether the beneficial role of a strong macroautophagy/autophagy, could also be used to ensure effective IL21 production as well. Herein, we confirm that after both polyclonal and HIV-1-specific activation, memory CD4 T cells (Mem) from EC display enhanced activity of the autophagy-mediated proteolysis compared to ART. Our results indicate that the enhanced autophagy activity in EC was controlled by the energy-sensing PRKAA1 (protein kinase AMP-activated catalytic subunit alpha 1). We further confirmed the critical role of the autophagy-mediated proteolysis in the strong IL21 production in EC by using BECN1 gene silencing as well as protease, PRKAA1, and lysosomal inhibitors. Finally, we established that high autophagy-mediated proteolysis in EC fuels their cellular rates of mitochondrial respiration due to glutaminolysis. Our data confirm the critical role of autophagy in dictating the metabolic input, which is required not only to ensure protective cytotoxic CD8A T cell responses, but also to provide strong IL21 production among antiviral CD4 T cells. Abbreviations: AKG: alpha-ketoglutarate; ART: patients under antiretroviral therapy; ATG7: autophagy related 7; BaF: bafilomycin A
1 ; BECN1: beclin 1; Chloro.: chloroquine; EC: elite controllers; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; FOXO3: forkhead box O3; GLS: glutaminase; GLUD1: glutamate dehydrogenase 1; HIVneg : HIV-1-uninfected control donors; IFNG/IFN-γ: interferon gamma; IL21: interleukin 21; MTOR: mechanistic target of rapamycin kinase; PBMC: peripheral blood mononuclear cells; PRKAA1: protein kinase AMP-activated catalytic subunit alpha 1; SQSTM1: sequestosome 1; TCA: tricarboxylic acid cycle; ULK1: unc-51 like autophagy activating kinase.- Published
- 2022
- Full Text
- View/download PDF
13. Mitochondrial complex IV defects induce metabolic and signaling perturbations that expose potential vulnerabilities in HCT116 cells.
- Author
-
Uchenunu O, Zhdanov AV, Hutton P, Jovanovic P, Wang Y, Andreev DE, Hulea L, Papadopoli DJ, Avizonis D, Baranov PV, Pollak MN, Papkovsky DB, and Topisirovic I
- Subjects
- HCT116 Cells, Humans, Mitochondria metabolism, Proto-Oncogene Proteins c-akt metabolism, Electron Transport Complex IV genetics, Electron Transport Complex IV metabolism, Molecular Chaperones metabolism
- Abstract
Mutations in genes encoding cytochrome c oxidase (mitochondrial complex IV) subunits and assembly factors [e.g., synthesis of cytochrome c oxidase 2 (SCO2)] are linked to severe metabolic syndromes. Notwithstanding that SCO2 is under transcriptional control of tumor suppressor p53, the role of mitochondrial complex IV dysfunction in cancer metabolism remains obscure. Herein, we demonstrate that the loss of SCO2 in HCT116 colorectal cancer cells leads to significant metabolic and signaling perturbations. Specifically, abrogation of SCO2 increased NAD
+ regenerating reactions and decreased glucose oxidation through citric acid cycle while enhancing pyruvate carboxylation. This was accompanied by a reduction in amino acid levels and the accumulation of lipid droplets. In addition, SCO2 loss resulted in hyperactivation of the insulin-like growth factor 1 receptor (IGF1R)/AKT axis with paradoxical downregulation of mTOR signaling, which was accompanied by increased AMP-activated kinase activity. Accordingly, abrogation of SCO2 expression appears to increase the sensitivity of cells to IGF1R and AKT, but not mTOR inhibitors. Finally, the loss of SCO2 was associated with reduced proliferation and enhanced migration of HCT116 cells. Collectively, herein we describe potential adaptive signaling and metabolic perturbations triggered by mitochondrial complex IV dysfunction., (© 2022 The Authors. FEBS Open Bio published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2022
- Full Text
- View/download PDF
14. Decrease of Intracellular Glutamine by STF-62247 Results in the Accumulation of Lipid Droplets in von Hippel-Lindau Deficient Cells.
- Author
-
Johnson M, Nowlan S, Sahin G, Barnett DA, Joy AP, Touaibia M, Cuperlovic-Culf M, Zofija Avizonis D, and Turcotte S
- Abstract
Kidney cancer is one of the top ten cancer diagnosed worldwide and its incidence has increased the last 20 years. Clear Cell Renal Cell Carcinoma (ccRCC) are characterized by mutations that inactivate the von Hippel-Lindau (VHL) tumor suppressor gene and evidence indicated alterations in metabolic pathways, particularly in glutamine metabolism. We previously identified a small molecule, STF-62247, which target VHL-deficient renal tumors by affecting late-stages of autophagy and lysosomal signaling. In this study, we investigated ccRCC metabolism in VHL-deficient and proficient cells exposed to the small molecule. Metabolomics profiling using 1H NMR demonstrated that STF-62247 increases levels of glucose, pyruvate, glycerol 3-phosphate while glutamate, asparagine, and glutathione significantly decreased. Diminution of glutamate and glutamine was further investigated using mass spectrometry, western blot analyses, enzymatic activities, and viability assays. We found that expression of SLC1A5 increases in VHL-deficient cells treated with STF-62247, possibly to stimulate glutamine uptake intracellularly to counteract the diminution of this amino acid. However, exogenous addition of glutamine was not able to rescue cell viability induced by the small molecule. Instead, our results showed that VHL-deficient cells utilize glutamine to produce fatty acid in response to STF-62247. Surprisingly, this occurs through oxidative phosphorylation in STF-treated cells while control cells use reductive carboxylation to sustain lipogenesis. We also demonstrated that STF-62247 stimulated expression of stearoyl-CoA desaturase (SCD1) and peripilin2 (PLIN2) to generate accumulation of lipid droplets in VHL-deficient cells. Moreover, the carnitine palmitoyltransferase 1A (CPT1A), which control the entry of fatty acid into mitochondria for β-oxidation, also increased in response to STF-62247. CPT1A overexpression in ccRCC is known to limit tumor growth. Together, our results demonstrated that STF-62247 modulates cellular metabolism of glutamine, an amino acid involved in the autophagy-lysosome process, to support lipogenesis, which could be implicated in the signaling driving to cell death., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Johnson, Nowlan, Sahin, Barnett, Joy, Touaibia, Cuperlovic-Culf, Zofija Avizonis and Turcotte.)
- Published
- 2022
- Full Text
- View/download PDF
15. Sarm1 activation produces cADPR to increase intra-axonal Ca++ and promote axon degeneration in PIPN.
- Author
-
Li Y, Pazyra-Murphy MF, Avizonis D, de Sá Tavares Russo M, Tang S, Chen CY, Hsueh YP, Bergholz JS, Jiang T, Zhao JJ, Zhu J, Ko KW, Milbrandt J, DiAntonio A, and Segal RA
- Subjects
- Animals, Calcium Channels metabolism, Cyclic ADP-Ribose antagonists & inhibitors, Female, HEK293 Cells, Humans, Mice, Inbred C57BL, Rats, Sprague-Dawley, Mice, Rats, Armadillo Domain Proteins metabolism, Axons metabolism, Calcium metabolism, Cyclic ADP-Ribose metabolism, Cytoskeletal Proteins metabolism, Nerve Degeneration pathology, Paclitaxel adverse effects, Peripheral Nervous System Diseases chemically induced, Peripheral Nervous System Diseases metabolism
- Abstract
Cancer patients frequently develop chemotherapy-induced peripheral neuropathy (CIPN), a painful and long-lasting disorder with profound somatosensory deficits. There are no effective therapies to prevent or treat this disorder. Pathologically, CIPN is characterized by a "dying-back" axonopathy that begins at intra-epidermal nerve terminals of sensory neurons and progresses in a retrograde fashion. Calcium dysregulation constitutes a critical event in CIPN, but it is not known how chemotherapies such as paclitaxel alter intra-axonal calcium and cause degeneration. Here, we demonstrate that paclitaxel triggers Sarm1-dependent cADPR production in distal axons, promoting intra-axonal calcium flux from both intracellular and extracellular calcium stores. Genetic or pharmacologic antagonists of cADPR signaling prevent paclitaxel-induced axon degeneration and allodynia symptoms, without mitigating the anti-neoplastic efficacy of paclitaxel. Our data demonstrate that cADPR is a calcium-modulating factor that promotes paclitaxel-induced axon degeneration and suggest that targeting cADPR signaling provides a potential therapeutic approach for treating paclitaxel-induced peripheral neuropathy (PIPN)., (© 2021 Li et al.)
- Published
- 2022
- Full Text
- View/download PDF
16. Glucose metabolism and pyruvate carboxylase enhance glutathione synthesis and restrict oxidative stress in pancreatic islets.
- Author
-
Fu A, van Rooyen L, Evans L, Armstrong N, Avizonis D, Kin T, Bird GH, Reddy A, Chouchani ET, Liesa-Roig M, Walensky LD, Shapiro AMJ, and Danial NN
- Subjects
- Adult, Animals, Antioxidants physiology, Female, Glutathione metabolism, Humans, Insulin metabolism, Islets of Langerhans metabolism, Male, Mice, Mice, Inbred C57BL, Middle Aged, Oxidation-Reduction, Oxidative Stress physiology, Primary Cell Culture, Glucose metabolism, Glutathione biosynthesis, Pyruvate Carboxylase metabolism
- Abstract
Glucose metabolism modulates the islet β cell responses to diabetogenic stress, including inflammation. Here, we probed the metabolic mechanisms that underlie the protective effect of glucose in inflammation by interrogating the metabolite profiles of primary islets from human donors and identified de novo glutathione synthesis as a prominent glucose-driven pro-survival pathway. We find that pyruvate carboxylase is required for glutathione synthesis in islets and promotes their antioxidant capacity to counter inflammation and nitrosative stress. Loss- and gain-of-function studies indicate that pyruvate carboxylase is necessary and sufficient to mediate the metabolic input from glucose into glutathione synthesis and the oxidative stress response. Altered redox metabolism and cellular capacity to replenish glutathione pools are relevant in multiple pathologies beyond obesity and diabetes. Our findings reveal a direct interplay between glucose metabolism and glutathione biosynthesis via pyruvate carboxylase. This metabolic axis may also have implications in other settings where sustaining glutathione is essential., Competing Interests: Declarations of interests L.D.W. is a scientific co-founder and shareholder in Aileron Therapeutics. M.L.-R. is a co-founder and consultant of Enspire Bio. E.T.C. is a founder, board member, and equity holder in EoCys Therapeutics., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
17. STAT1 potentiates oxidative stress revealing a targetable vulnerability that increases phenformin efficacy in breast cancer.
- Author
-
Totten SP, Im YK, Cepeda Cañedo E, Najyb O, Nguyen A, Hébert S, Ahn R, Lewis K, Lebeau B, La Selva R, Sabourin V, Martínez C, Savage P, Kuasne H, Avizonis D, Santos Martínez N, Chabot C, Aguilar-Mahecha A, Goulet ML, Dankner M, Witcher M, Petrecca K, Basik M, Pollak M, Topisirovic I, Lin R, Siegel PM, Kleinman CL, Park M, St-Pierre J, and Ursini-Siegel J
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Cell Line, Tumor, Drug Synergism, Electron Transport Complex I antagonists & inhibitors, Energy Metabolism drug effects, Female, Glutathione antagonists & inhibitors, Glutathione biosynthesis, Humans, Interferon-gamma administration & dosage, Interferon-gamma deficiency, Interferon-gamma metabolism, MCF-7 Cells, Mammary Neoplasms, Experimental drug therapy, Mammary Neoplasms, Experimental metabolism, Mice, Mice, Inbred BALB C, Mice, Knockout, Mice, SCID, NAD(P)H Dehydrogenase (Quinone) antagonists & inhibitors, NAD(P)H Dehydrogenase (Quinone) metabolism, Naphthoquinones administration & dosage, Oxidative Stress drug effects, Phenformin administration & dosage, Poly I-C administration & dosage, Reactive Oxygen Species metabolism, STAT1 Transcription Factor agonists, Xenograft Model Antitumor Assays, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Phenformin pharmacology, STAT1 Transcription Factor metabolism
- Abstract
Bioenergetic perturbations driving neoplastic growth increase the production of reactive oxygen species (ROS), requiring a compensatory increase in ROS scavengers to limit oxidative stress. Intervention strategies that simultaneously induce energetic and oxidative stress therefore have therapeutic potential. Phenformin is a mitochondrial complex I inhibitor that induces bioenergetic stress. We now demonstrate that inflammatory mediators, including IFNγ and polyIC, potentiate the cytotoxicity of phenformin by inducing a parallel increase in oxidative stress through STAT1-dependent mechanisms. Indeed, STAT1 signaling downregulates NQO1, a key ROS scavenger, in many breast cancer models. Moreover, genetic ablation or pharmacological inhibition of NQO1 using β-lapachone (an NQO1 bioactivatable drug) increases oxidative stress to selectively sensitize breast cancer models, including patient derived xenografts of HER2+ and triple negative disease, to the tumoricidal effects of phenformin. We provide evidence that therapies targeting ROS scavengers increase the anti-neoplastic efficacy of mitochondrial complex I inhibitors in breast cancer.
- Published
- 2021
- Full Text
- View/download PDF
18. Reprogramming of Nucleotide Metabolism Mediates Synergy between Epigenetic Therapy and MAP Kinase Inhibition.
- Author
-
Shorstova T, Su J, Zhao T, Dahabieh M, Leibovitch M, De Sa Tavares Russo M, Avizonis D, Rajkumar S, Watson IR, Del Rincón SV, Miller WH Jr, Foulkes WD, and Witcher M
- Subjects
- Animals, Azetidines pharmacology, Cell Cycle Checkpoints drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Down-Regulation drug effects, Drug Synergism, Female, Gene Knockdown Techniques, HEK293 Cells, Humans, Mice, Inbred NOD, Mice, SCID, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Mitogen-Activated Protein Kinases metabolism, Neoplasm Proteins metabolism, Ovarian Neoplasms drug therapy, Piperidines pharmacology, Protein Kinase Inhibitors therapeutic use, S Phase drug effects, Xenograft Model Antitumor Assays, Mice, Epigenesis, Genetic drug effects, Mitogen-Activated Protein Kinases antagonists & inhibitors, Nucleotides metabolism, Protein Kinase Inhibitors pharmacology
- Abstract
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare but often lethal cancer that is diagnosed at a median age of 24 years. Optimal management of patients is not well defined, and current treatment remains challenging, necessitating the discovery of novel therapeutic approaches. The identification of SMARCA4-inactivating mutations invariably characterizing this type of cancer provided insights facilitating diagnostic and therapeutic measures against this disease. We show here that the BET inhibitor OTX015 acts in synergy with the MEK inhibitor cobimetinib to repress the proliferation of SCCOHT in vivo Notably, this synergy is also observed in some SMARCA4-expressing ovarian adenocarcinoma models intrinsically resistant to BETi. Mass spectrometry, coupled with knockdown of newly found targets such as thymidylate synthase, revealed that the repression of a panel of proteins involved in nucleotide synthesis underlies this synergy both in vitro and in vivo , resulting in reduced pools of nucleotide metabolites and subsequent cell-cycle arrest. Overall, our data indicate that dual treatment with BETi and MEKi represents a rational combination therapy against SCCOHT and potentially additional ovarian cancer subtypes., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
19. Genome-Wide Screens Reveal that Resveratrol Induces Replicative Stress in Human Cells.
- Author
-
Benslimane Y, Bertomeu T, Coulombe-Huntington J, McQuaid M, Sánchez-Osuna M, Papadopoli D, Avizonis D, Russo MST, Huard C, Topisirovic I, Wurtele H, Tyers M, and Harrington L
- Subjects
- CRISPR-Cas Systems, Cell Line, Drug Resistance genetics, Humans, Hydroxyurea pharmacology, Jurkat Cells, Nucleotides metabolism, S Phase Cell Cycle Checkpoints drug effects, Sirtuin 1 metabolism, Stilbenes pharmacology, Cell Proliferation drug effects, DNA Replication drug effects, Resveratrol pharmacology
- Abstract
Resveratrol is a natural product associated with wide-ranging effects in animal and cellular models, including lifespan extension. To identify the genetic target of resveratrol in human cells, we conducted genome-wide CRISPR-Cas9 screens to pinpoint genes that confer sensitivity or resistance to resveratrol. An extensive network of DNA damage response and replicative stress genes exhibited genetic interactions with resveratrol and its analog pterostilbene. These genetic profiles showed similarity to the response to hydroxyurea, an inhibitor of ribonucleotide reductase that causes replicative stress. Resveratrol, pterostilbene, and hydroxyurea caused similar depletion of nucleotide pools, inhibition of replication fork progression, and induction of replicative stress. The ability of resveratrol to inhibit cell proliferation and S phase transit was independent of the histone deacetylase sirtuin 1, which has been implicated in lifespan extension by resveratrol. These results establish that a primary impact of resveratrol on human cell proliferation is the induction of low-level replicative stress., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
20. Repression of LKB1 by miR-17∼92 Sensitizes MYC -Dependent Lymphoma to Biguanide Treatment.
- Author
-
Izreig S, Gariepy A, Kaymak I, Bridges HR, Donayo AO, Bridon G, DeCamp LM, Kitchen-Goosen SM, Avizonis D, Sheldon RD, Laister RC, Minden MD, Johnson NA, Duchaine TF, Rudoltz MS, Yoo S, Pollak MN, Williams KS, and Jones RG
- Subjects
- AMP-Activated Protein Kinase Kinases drug effects, Animals, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Apoptosis genetics, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Drug Synergism, HEK293 Cells, Humans, Lymphoma genetics, Lymphoma pathology, Mice, Mice, Nude, Proto-Oncogene Proteins c-myc genetics, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, AMP-Activated Protein Kinase Kinases genetics, Biguanides therapeutic use, Lymphoma drug therapy, RNA, Long Noncoding physiology
- Abstract
Cancer cells display metabolic plasticity to survive stresses in the tumor microenvironment. Cellular adaptation to energetic stress is coordinated in part by signaling through the liver kinase B1 (LKB1)-AMP-activated protein kinase (AMPK) pathway. Here, we demonstrate that miRNA-mediated silencing of LKB1 confers sensitivity of lymphoma cells to mitochondrial inhibition by biguanides. Using both classic (phenformin) and newly developed (IM156) biguanides, we demonstrate that elevated miR-17∼92 expression in Myc
+ lymphoma cells promotes increased apoptosis to biguanide treatment in vitro and in vivo . This effect is driven by the miR-17 -dependent silencing of LKB1, which reduces AMPK activation in response to complex I inhibition. Mechanistically, biguanide treatment induces metabolic stress in Myc+ lymphoma cells by inhibiting TCA cycle metabolism and mitochondrial respiration, exposing metabolic vulnerability. Finally, we demonstrate a direct correlation between miR-17∼92 expression and biguanide sensitivity in human cancer cells. Our results identify miR-17∼92 expression as a potential biomarker for biguanide sensitivity in malignancies., Competing Interests: R.G.J. and M.N.P. serve on the Scientific Advisory Board of ImmunoMet Therapeutics. M.S.R. is a consultant to ImmunoMet Therapeutics. S.Y. is an employee of ImmunoMet Therapeutics. R.G.J. and ImmunoMet Therapeutics hold a provisional patent for the use of IM156 for treatment of malignancy based on miR-17∼92, Myc, or LKB1 expression., (© 2020 The Author(s).)- Published
- 2020
- Full Text
- View/download PDF
21. Methotrexate elicits pro-respiratory and anti-growth effects by promoting AMPK signaling.
- Author
-
Papadopoli DJ, Ma EH, Roy D, Russo M, Bridon G, Avizonis D, Jones RG, and St-Pierre J
- Subjects
- Biguanides pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, Cell Respiration drug effects, Drug Synergism, Humans, Mitochondria drug effects, Mitochondria metabolism, Mitochondria pathology, AMP-Activated Protein Kinases metabolism, Antineoplastic Agents pharmacology, Methotrexate pharmacology, Signal Transduction drug effects
- Abstract
One-carbon metabolism fuels the high demand of cancer cells for nucleotides and other building blocks needed for increased proliferation. Although inhibitors of this pathway are widely used to treat many cancers, their global impact on anabolic and catabolic processes remains unclear. Using a combination of real-time bioenergetics assays and metabolomics approaches, we investigated the global effects of methotrexate on cellular metabolism. We show that methotrexate treatment increases the intracellular concentration of the metabolite AICAR, resulting in AMPK activation. Methotrexate-induced AMPK activation leads to decreased one-carbon metabolism gene expression and cellular proliferation as well as increased global bioenergetic capacity. The anti-proliferative and pro-respiratory effects of methotrexate are AMPK-dependent, as cells with reduced AMPK activity are less affected by methotrexate treatment. Conversely, the combination of methotrexate with the AMPK activator, phenformin, potentiates its anti-proliferative activity in cancer cells. These data highlight a reciprocal effect of methotrexate on anabolic and catabolic processes and implicate AMPK activation as a metabolic determinant of methotrexate response.
- Published
- 2020
- Full Text
- View/download PDF
22. Glucose-dependent partitioning of arginine to the urea cycle protects β-cells from inflammation.
- Author
-
Fu A, Alvarez-Perez JC, Avizonis D, Kin T, Ficarro SB, Choi DW, Karakose E, Badur MG, Evans L, Rosselot C, Bridon G, Bird GH, Seo HS, Dhe-Paganon S, Kamphorst JJ, Stewart AF, James Shapiro AM, Marto JA, Walensky LD, Jones RG, Garcia-Ocana A, and Danial NN
- Subjects
- Adolescent, Adult, Aged, Aspartic Acid metabolism, Cell Survival, Citric Acid Cycle drug effects, Female, Humans, Inflammation pathology, Insulin-Secreting Cells pathology, Male, Metabolomics, Middle Aged, Nitric Oxide metabolism, Pyruvate Carboxylase metabolism, Urea Cycle Disorders, Inborn metabolism, Young Adult, Arginine metabolism, Glucose metabolism, Glucose pharmacology, Inflammation prevention & control, Insulin-Secreting Cells drug effects, Urea metabolism, Urea Cycle Disorders, Inborn pathology
- Abstract
Chronic inflammation is linked to diverse disease processes, but the intrinsic mechanisms that determine cellular sensitivity to inflammation are incompletely understood. Here, we show the contribution of glucose metabolism to inflammation-induced changes in the survival of pancreatic islet β-cells. Using metabolomic, biochemical and functional analyses, we investigate the protective versus non-protective effects of glucose in the presence of pro-inflammatory cytokines. When protective, glucose metabolism augments anaplerotic input into the TCA cycle via pyruvate carboxylase (PC) activity, leading to increased aspartate levels. This metabolic mechanism supports the argininosuccinate shunt, which fuels ureagenesis from arginine and conversely diminishes arginine utilization for production of nitric oxide (NO), a chief mediator of inflammatory cytotoxicity. Activation of the PC-urea cycle axis is sufficient to suppress NO synthesis and shield cells from death in the context of inflammation and other stress paradigms. Overall, these studies uncover a previously unappreciated link between glucose metabolism and arginine-utilizing pathways via PC-directed ureagenesis as a protective mechanism.
- Published
- 2020
- Full Text
- View/download PDF
23. eIF4A supports an oncogenic translation program in pancreatic ductal adenocarcinoma.
- Author
-
Chan K, Robert F, Oertlin C, Kapeller-Libermann D, Avizonis D, Gutierrez J, Handly-Santana A, Doubrovin M, Park J, Schoepfer C, Da Silva B, Yao M, Gorton F, Shi J, Thomas CJ, Brown LE, Porco JA Jr, Pollak M, Larsson O, Pelletier J, and Chio IIC
- Subjects
- Animals, Carcinogenesis, Carcinoma, Pancreatic Ductal metabolism, Cell Line, Tumor, Eukaryotic Initiation Factor-4A antagonists & inhibitors, Eukaryotic Initiation Factor-4A metabolism, Glutathione metabolism, Humans, Mice, Inbred C57BL, Molecular Targeted Therapy, Oxidation-Reduction, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms, Protein Biosynthesis
- Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal malignancy with limited treatment options. Although metabolic reprogramming is a hallmark of many cancers, including PDA, previous attempts to target metabolic changes therapeutically have been stymied by drug toxicity and tumour cell plasticity. Here, we show that PDA cells engage an eIF4F-dependent translation program that supports redox and central carbon metabolism. Inhibition of the eIF4F subunit, eIF4A, using the synthetic rocaglate CR-1-31-B (CR-31) reduced the viability of PDA organoids relative to their normal counterparts. In vivo, CR-31 suppresses tumour growth and extends survival of genetically-engineered murine models of PDA. Surprisingly, inhibition of eIF4A also induces glutamine reductive carboxylation. As a consequence, combined targeting of eIF4A and glutaminase activity more effectively inhibits PDA cell growth both in vitro and in vivo. Overall, our work demonstrates the importance of eIF4A in translational control of pancreatic tumour metabolism and as a therapeutic target against PDA.
- Published
- 2019
- Full Text
- View/download PDF
24. Translational and HIF-1α-Dependent Metabolic Reprogramming Underpin Metabolic Plasticity and Responses to Kinase Inhibitors and Biguanides.
- Author
-
Hulea L, Gravel SP, Morita M, Cargnello M, Uchenunu O, Im YK, Lehuédé C, Ma EH, Leibovitch M, McLaughlan S, Blouin MJ, Parisotto M, Papavasiliou V, Lavoie C, Larsson O, Ohh M, Ferreira T, Greenwood C, Bridon G, Avizonis D, Ferbeyre G, Siegel P, Jones RG, Muller W, Ursini-Siegel J, St-Pierre J, Pollak M, and Topisirovic I
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Amino Acids metabolism, Animals, Biguanides pharmacology, Cell Cycle Proteins, Eukaryotic Initiation Factors metabolism, Gene Expression Regulation, Neoplastic, HCT116 Cells, Humans, K562 Cells, Mechanistic Target of Rapamycin Complex 1 metabolism, Mice, Mice, Nude, Phosphoproteins metabolism, Protein Kinase Inhibitors pharmacology, RNA, Messenger metabolism, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, Drug Resistance, Neoplasm, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Neoplasms drug therapy, Neoplasms metabolism, Signal Transduction drug effects
- Abstract
There is increasing interest in therapeutically exploiting metabolic differences between normal and cancer cells. We show that kinase inhibitors (KIs) and biguanides synergistically and selectively target a variety of cancer cells. Synthesis of non-essential amino acids (NEAAs) aspartate, asparagine, and serine, as well as glutamine metabolism, are major determinants of the efficacy of KI/biguanide combinations. The mTORC1/4E-BP axis regulates aspartate, asparagine, and serine synthesis by modulating mRNA translation, while ablation of 4E-BP1/2 substantially decreases sensitivity of breast cancer and melanoma cells to KI/biguanide combinations. Efficacy of the KI/biguanide combinations is also determined by HIF-1α-dependent perturbations in glutamine metabolism, which were observed in VHL-deficient renal cancer cells. This suggests that cancer cells display metabolic plasticity by engaging non-redundant adaptive mechanisms, which allows them to survive therapeutic insults that target cancer metabolism., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
25. Leveraging increased cytoplasmic nucleoside kinase activity to target mtDNA and oxidative phosphorylation in AML.
- Author
-
Liyanage SU, Hurren R, Voisin V, Bridon G, Wang X, Xu C, MacLean N, Siriwardena TP, Gronda M, Yehudai D, Sriskanthadevan S, Avizonis D, Shamas-Din A, Minden MD, Bader GD, Laposa R, and Schimmer AD
- Subjects
- Animals, Cell Line, Tumor, Cells, Cultured, DNA Replication, Humans, Mice, SCID, NM23 Nucleoside Diphosphate Kinases metabolism, Nucleoside-Phosphate Kinase metabolism, Signal Transduction, Tumor Cells, Cultured, Zalcitabine metabolism, DNA, Mitochondrial genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Oxidative Phosphorylation, Phosphotransferases metabolism
- Abstract
Mitochondrial DNA (mtDNA) biosynthesis requires replication factors and adequate nucleotide pools from the mitochondria and cytoplasm. We performed gene expression profiling analysis of 542 human acute myeloid leukemia (AML) samples and identified 55% with upregulated mtDNA biosynthesis pathway expression compared with normal hematopoietic cells. Genes that support mitochondrial nucleotide pools, including mitochondrial nucleotide transporters and a subset of cytoplasmic nucleoside kinases, were also increased in AML compared with normal hematopoietic samples. Knockdown of cytoplasmic nucleoside kinases reduced mtDNA levels in AML cells, demonstrating their contribution in maintaining mtDNA. To assess cytoplasmic nucleoside kinase pathway activity, we used a nucleoside analog 2'3'-dideoxycytidine (ddC), which is phosphorylated to the activated antimetabolite, 2'3'-dideoxycytidine triphosphate by cytoplasmic nucleoside kinases. ddC is a selective inhibitor of the mitochondrial DNA polymerase γ. ddC was preferentially activated in AML cells compared with normal hematopoietic progenitor cells. ddC treatment inhibited mtDNA replication, oxidative phosphorylation, and induced cytotoxicity in a panel of AML cell lines. Furthermore, ddC preferentially inhibited mtDNA replication in a subset of primary human leukemia cells and selectively targeted leukemia cells while sparing normal progenitor cells. In animal models of human AML, treatment with ddC decreased mtDNA, electron transport chain proteins, and induced tumor regression without toxicity. ddC also targeted leukemic stem cells in secondary AML xenotransplantation assays. Thus, AML cells have increased cytidine nucleoside kinase activity that regulates mtDNA biogenesis and can be leveraged to selectively target oxidative phosphorylation in AML., (© 2017 by The American Society of Hematology.)
- Published
- 2017
- Full Text
- View/download PDF
26. Are Metformin Doses Used in Murine Cancer Models Clinically Relevant?
- Author
-
Chandel NS, Avizonis D, Reczek CR, Weinberg SE, Menz S, Neuhaus R, Christian S, Haegebarth A, Algire C, and Pollak M
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacology, Disease Models, Animal, Dose-Response Relationship, Drug, Humans, Hypoglycemic Agents administration & dosage, Hypoglycemic Agents pharmacology, Metformin administration & dosage, Metformin pharmacology, Mice, Mice, Inbred C57BL, Neoplasms pathology, Antineoplastic Agents therapeutic use, Hypoglycemic Agents therapeutic use, Metformin therapeutic use, Neoplasms drug therapy
- Published
- 2016
- Full Text
- View/download PDF
27. High Sensitivity of an Ha-RAS Transgenic Model of Superficial Bladder Cancer to Metformin Is Associated with ∼240-Fold Higher Drug Concentration in Urine than Serum.
- Author
-
Liu Z, Yokoyama NN, Blair CA, Li X, Avizonis D, Wu XR, Uchio E, Youssef R, McClelland M, Pollak M, and Zi X
- Subjects
- Animals, Biomarkers, Cell Line, Tumor, Cell Proliferation drug effects, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Disease Models, Animal, Disease Progression, Female, Homozygote, Humans, Male, Mice, Mice, Transgenic, Mutation, Neoplasm Grading, S-Phase Kinase-Associated Proteins metabolism, Signal Transduction drug effects, TOR Serine-Threonine Kinases, Tumor Burden drug effects, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms metabolism, Urinary Bladder Neoplasms pathology, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacokinetics, Drug Resistance, Neoplasm genetics, Genes, ras, Metformin pharmacokinetics, Urinary Bladder Neoplasms genetics
- Abstract
While pharmacoepidemiologic and laboratory studies have supported the hypothesis that the antidiabetic drug metformin may be useful in treating or preventing cancer, there is limited evidence to suggest which specific cancer sites may be particularly sensitive. Sensitivity likely is determined both by features of tumor pathophysiology and by pharmacokinetic factors. We used UPII-mutant Ha-ras transgenic mice that develop hyperplasia and low-grade, papillary urothelial cell carcinoma to determine whether metformin has activity in a model of superficial bladder cancer. Metformin significantly improved survival, reduced urinary tract obstruction, reduced bladder weight (a surrogate for tumor volume), and led to clear activation of AMP α kinase and inhibition of mTOR signaling in neoplastic tissue. We investigated the basis of the unusual sensitivity of this model to metformin, and observed that following oral dosing, urothelium is exposed to drug concentrations via the urine that are approximately 240-fold higher than those in the circulation. In addition, we observed that bladder cancer cell lines (RT4, UMUC-3, and J82) with homozygous deletion of either TSC1 or PTEN are more sensitive to metformin than those (TEU2, TCCSUP, and HT1376) with wild-type TSC1 and PTEN genes. Our findings provide a strong rationale for clinical trials of oral metformin in treatment of superficial bladder cancer., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
28. Metabolomics Analyses of Cancer Cells in Controlled Microenvironments.
- Author
-
Gravel SP, Avizonis D, and St-Pierre J
- Subjects
- Cell Line, Tumor, Cells, Cultured, Gas Chromatography-Mass Spectrometry, Humans, Hypoxia metabolism, Workflow, Metabolome, Metabolomics methods, Neoplasms metabolism, Neoplasms pathology, Tumor Microenvironment
- Abstract
The tumor microenvironment is a complex and heterogeneous milieu in which cancer cells undergo metabolic reprogramming to fuel their growth. Cancer cell lines grown in vitro using traditional culture methods represent key experimental models to gain a mechanistic understanding of tumor biology. This protocol describes the use of gas chromatography-mass spectrometry (GC-MS) to assess metabolic changes in cancer cells grown under varied levels of oxygen and nutrients that may better mimic the tumor microenvironment. Intracellular metabolite changes, metabolite uptake and release, as well as stable isotope ((13)C) tracer analyses are done in a single experimental setup to provide an integrated understanding of metabolic adaptation. Overall, this chapter describes some essential tools and methods to perform comprehensive metabolomics analyses.
- Published
- 2016
- Full Text
- View/download PDF
29. Mitochondrial Phosphoenolpyruvate Carboxykinase Regulates Metabolic Adaptation and Enables Glucose-Independent Tumor Growth.
- Author
-
Vincent EE, Sergushichev A, Griss T, Gingras MC, Samborska B, Ntimbane T, Coelho PP, Blagih J, Raissi TC, Choinière L, Bridon G, Loginicheva E, Flynn BR, Thomas EC, Tavaré JM, Avizonis D, Pause A, Elder DJ, Artyomov MN, and Jones RG
- Subjects
- Adaptation, Physiological genetics, Animals, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cell Proliferation, Citric Acid Cycle genetics, Glucose deficiency, Glutamine metabolism, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Metabolomics, Mice, Mice, Nude, Mitochondria metabolism, Neoplasms genetics, Neoplasms pathology, Phosphoenolpyruvate metabolism, Phosphoenolpyruvate Carboxykinase (ATP) genetics, Purines biosynthesis, Pyruvic Acid metabolism, Serine biosynthesis, Carcinoma, Non-Small-Cell Lung metabolism, Gene Expression Regulation, Neoplastic, Gluconeogenesis genetics, Lung Neoplasms metabolism, Neoplasms metabolism, Phosphoenolpyruvate Carboxykinase (ATP) metabolism
- Abstract
Cancer cells adapt metabolically to proliferate under nutrient limitation. Here we used combined transcriptional-metabolomic network analysis to identify metabolic pathways that support glucose-independent tumor cell proliferation. We found that glucose deprivation stimulated re-wiring of the tricarboxylic acid (TCA) cycle and early steps of gluconeogenesis to promote glucose-independent cell proliferation. Glucose limitation promoted the production of phosphoenolpyruvate (PEP) from glutamine via the activity of mitochondrial PEP-carboxykinase (PCK2). Under these conditions, glutamine-derived PEP was used to fuel biosynthetic pathways normally sustained by glucose, including serine and purine biosynthesis. PCK2 expression was required to maintain tumor cell proliferation under limited-glucose conditions in vitro and tumor growth in vivo. Elevated PCK2 expression is observed in several human tumor types and enriched in tumor tissue from non-small-cell lung cancer (NSCLC) patients. Our results define a role for PCK2 in cancer cell metabolic reprogramming that promotes glucose-independent cell growth and metabolic stress resistance in human tumors., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
30. Deletion of the gene encoding G0/G 1 switch protein 2 (G0s2) alleviates high-fat-diet-induced weight gain and insulin resistance, and promotes browning of white adipose tissue in mice.
- Author
-
El-Assaad W, El-Kouhen K, Mohammad AH, Yang J, Morita M, Gamache I, Mamer O, Avizonis D, Hermance N, Kersten S, Tremblay ML, Kelliher MA, and Teodoro JG
- Subjects
- Adiposity genetics, Animals, Energy Metabolism genetics, Female, Gene Deletion, Male, Mice, Mice, Knockout, Thermogenesis genetics, Adipocytes, Brown physiology, Adipose Tissue, White physiology, Cell Cycle Proteins genetics, Cell Transdifferentiation genetics, Diet, High-Fat adverse effects, Insulin Resistance genetics, Weight Gain genetics
- Abstract
Aims/hypothesis: Obesity is a global epidemic resulting from increased energy intake, which alters energy homeostasis and results in an imbalance in fat storage and breakdown. G0/G1 switch gene 2 (G0s2) has been recently characterised in vitro as an inhibitor of adipose triglyceride lipase (ATGL), the rate-limiting step in fat catabolism. In the current study we aim to functionally characterise G0s2 within the physiological context of a mouse model., Methods: We generated a mouse model in which G0s2 was deleted. The homozygous G0s2 knockout (G0s2 (-/-)) mice were studied over a period of 22 weeks. Metabolic variables were measured including body weight and body composition, food intake, glucose and insulin tolerance tests, energy metabolism and thermogenesis., Results: We report that G0s2 inhibits ATGL and regulates lipolysis and energy metabolism in vivo. G0s2 (-/-) mice are lean, resistant to weight gain induced by a high-fat diet and are glucose tolerant and insulin sensitive. The white adipose tissue of G0s2 (-/-) mice has enhanced lipase activity and adipocytes showed enhanced stimulated lipolysis. Energy metabolism in the G0s2 (-/-) mice is shifted towards enhanced lipid metabolism and increased thermogenesis. G0s2 (-/-) mice showed enhanced cold tolerance and increased expression of thermoregulatory and oxidation genes within white adipose tissue, suggesting enhanced 'browning' of the white adipose tissue., Conclusions/interpretation: Our data show that G0s2 is a physiological regulator of adiposity and energy metabolism and is a potential target in the treatment of obesity and insulin resistance.
- Published
- 2015
- Full Text
- View/download PDF
31. Synergy between the NAMPT inhibitor GMX1777(8) and pemetrexed in non-small cell lung cancer cells is mediated by PARP activation and enhanced NAD consumption.
- Author
-
Chan M, Gravel M, Bramoullé A, Bridon G, Avizonis D, Shore GC, and Roulston A
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cytokines antagonists & inhibitors, DNA Repair drug effects, Drug Synergism, Gene Expression Regulation, Neoplastic, Guanine administration & dosage, Humans, Mice, NAD metabolism, Nicotinamide Phosphoribosyltransferase antagonists & inhibitors, Pemetrexed, Poly(ADP-ribose) Polymerase Inhibitors, Poly(ADP-ribose) Polymerases genetics, Transcriptional Activation genetics, Xenograft Model Antitumor Assays, Carcinoma, Non-Small-Cell Lung drug therapy, Cytokines biosynthesis, Glutamates administration & dosage, Guanidines administration & dosage, Guanine analogs & derivatives, Nicotinamide Phosphoribosyltransferase biosynthesis, Poly(ADP-ribose) Polymerases biosynthesis
- Abstract
GMX1778 and its prodrug GMX1777 represent a new class of cancer drugs that targets nicotinamide phosphoribosyltransferase (NAMPT) as a new strategy to interfere with biosynthesis of the key enzymatic cofactor NAD, which is critical for a number of cell functions, including DNA repair. Using a genome-wide synthetic lethal siRNA screen, we identified the folate pathway-related genes, deoxyuridine triphosphatase and dihydrofolate reductase, the silencing of which sensitized non-small cell lung carcinoma (NSCLC) cells to the cytotoxic effects of GMX. Pemetrexed is an inhibitor of dihydrofolate reductase currently used to treat patients with nonsquamous NSCLC. We found that combining pemetrexed with GMX1777 produced a synergistic therapeutic benefit in A549 and H1299 NSCLC cells in vitro and in a mouse A549 xenograft model of lung cancer. Pemetrexed is known to activate PARPs, thereby accelerating NAD consumption. Genetic or pharmacologic blockade of PARP activity inhibited this effect, impairing cell death by pemetrexed either alone or in combination with GMX1777. Conversely, inhibiting the base excision repair pathway accentuated NAD decline in response to GMX and the cytotoxicity of both agents either alone or in combination. These findings provide a mechanistic rationale for combining GMX1777 with pemetrexed as an effective new therapeutic strategy to treat nonsquamous NSCLC., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
32. Stable isotope tracer analysis in isolated mitochondria from mammalian systems.
- Author
-
Gravel SP, Andrzejewski S, Avizonis D, and St-Pierre J
- Abstract
Mitochondria are a focal point in metabolism, given that they play fundamental roles in catabolic, as well as anabolic reactions. Alterations in mitochondrial functions are often studied in whole cells, and metabolomics experiments using 13C-labeled substrates, coupled with mass isotopomer distribution analyses, represent a powerful approach to study global changes in cellular metabolic activities. However, little is known regarding the assessment of metabolic activities in isolated mitochondria using this technology. Studies on isolated mitochondria permit the evaluation of whether changes in cellular metabolic activities are due to modifications in the intrinsic properties of the mitochondria. Here, we present a streamlined approach to accurately determine 13C, as well as 12C enrichments in isolated mitochondria from mammalian tissues or cultured cells by GC/MS. We demonstrate the relevance of this experimental approach by assessing the effects of drugs perturbing mitochondrial functions on the mass isotopomer enrichment of metabolic intermediates. Furthermore, we investigate 13C and 12C enrichments in mitochondria isolated from cancer cells given the emerging role of metabolic alterations in supporting tumor growth. This original method will provide a very sensitive tool to perform metabolomics studies on isolated mitochondria.
- Published
- 2014
- Full Text
- View/download PDF
33. Oxidative metabolism enables Salmonella evasion of the NLRP3 inflammasome.
- Author
-
Wynosky-Dolfi MA, Snyder AG, Philip NH, Doonan PJ, Poffenberger MC, Avizonis D, Zwack EE, Riblett AM, Hu B, Strowig T, Flavell RA, Jones RG, Freedman BD, and Brodsky IE
- Subjects
- Aconitate Hydratase metabolism, Animals, Apoptosis Regulatory Proteins metabolism, Bacterial Secretion Systems, Calcium-Binding Proteins metabolism, Citrate (si)-Synthase metabolism, Citric Acid Cycle, Genes, Bacterial genetics, Immunity, Isocitrate Dehydrogenase metabolism, Mice, Mice, Inbred C57BL, Mitochondria metabolism, Mutation genetics, NLR Family, Pyrin Domain-Containing 3 Protein, Oxidation-Reduction, Reactive Oxygen Species metabolism, Salmonella Infections, Animal immunology, Salmonella Infections, Animal pathology, Salmonella typhimurium enzymology, Salmonella typhimurium genetics, Salmonella typhimurium immunology, Carrier Proteins metabolism, Immune Evasion immunology, Inflammasomes metabolism, Salmonella typhimurium metabolism
- Abstract
Microbial infection triggers assembly of inflammasome complexes that promote caspase-1-dependent antimicrobial responses. Inflammasome assembly is mediated by members of the nucleotide binding domain leucine-rich repeat (NLR) protein family that respond to cytosolic bacterial products or disruption of cellular processes. Flagellin injected into host cells by invading Salmonella induces inflammasome activation through NLRC4, whereas NLRP3 is required for inflammasome activation in response to multiple stimuli, including microbial infection, tissue damage, and metabolic dysregulation, through mechanisms that remain poorly understood. During systemic infection, Salmonella avoids NLRC4 inflammasome activation by down-regulating flagellin expression. Macrophages exhibit delayed NLRP3 inflammasome activation after Salmonella infection, suggesting that Salmonella may evade or prevent the rapid activation of the NLRP3 inflammasome. We therefore screened a Salmonella Typhimurium transposon library to identify bacterial factors that limit NLRP3 inflammasome activation. Surprisingly, absence of the Salmonella TCA enzyme aconitase induced rapid NLRP3 inflammasome activation. This inflammasome activation correlated with elevated levels of bacterial citrate, and required mitochondrial reactive oxygen species and bacterial citrate synthase. Importantly, Salmonella lacking aconitase displayed NLRP3- and caspase-1/11-dependent attenuation of virulence, and induced elevated serum IL-18 in wild-type mice. Together, our data link Salmonella genes controlling oxidative metabolism to inflammasome activation and suggest that NLRP3 inflammasome evasion promotes systemic Salmonella virulence.
- Published
- 2014
- Full Text
- View/download PDF
34. Loss of the tumor suppressor LKB1 promotes metabolic reprogramming of cancer cells via HIF-1α.
- Author
-
Faubert B, Vincent EE, Griss T, Samborska B, Izreig S, Svensson RU, Mamer OA, Avizonis D, Shackelford DB, Shaw RJ, and Jones RG
- Subjects
- AMP-Activated Protein Kinase Kinases, Adenosine Triphosphate metabolism, Analysis of Variance, Animals, Apoptosis physiology, Blotting, Western, Cell Line, Tumor, Cell Proliferation, Fibroblasts, Gas Chromatography-Mass Spectrometry, Glucose metabolism, Glutamine metabolism, Humans, Mechanistic Target of Rapamycin Complex 1, Metabolic Networks and Pathways physiology, Mice, Multiprotein Complexes metabolism, Oxygen Consumption physiology, Protein Serine-Threonine Kinases metabolism, Reactive Oxygen Species metabolism, TOR Serine-Threonine Kinases metabolism, Energy Metabolism physiology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Metabolic Networks and Pathways genetics, Protein Serine-Threonine Kinases deficiency
- Abstract
One of the major metabolic changes associated with cellular transformation is enhanced nutrient utilization, which supports tumor progression by fueling both energy production and providing biosynthetic intermediates for growth. The liver kinase B1 (LKB1) is a serine/threonine kinase and tumor suppressor that couples bioenergetics to cell-growth control through regulation of mammalian target of rapamycin (mTOR) activity; however, the influence of LKB1 on tumor metabolism is not well defined. Here, we show that loss of LKB1 induces a progrowth metabolic program in proliferating cells. Cells lacking LKB1 display increased glucose and glutamine uptake and utilization, which support both cellular ATP levels and increased macromolecular biosynthesis. This LKB1-dependent reprogramming of cell metabolism is dependent on the hypoxia-inducible factor-1α (HIF-1α), which accumulates under normoxia in LKB1-deficient cells and is antagonized by inhibition of mTOR complex I signaling. Silencing HIF-1α reverses the metabolic advantages conferred by reduced LKB1 signaling and impairs the growth and survival of LKB1-deficient tumor cells under low-nutrient conditions. Together, our data implicate the tumor suppressor LKB1 as a central regulator of tumor metabolism and growth control through the regulation of HIF-1α-dependent metabolic reprogramming.
- Published
- 2014
- Full Text
- View/download PDF
35. PGC-1α supports glutamine metabolism in breast cancer.
- Author
-
McGuirk S, Gravel SP, Deblois G, Papadopoli DJ, Faubert B, Wegner A, Hiller K, Avizonis D, Akavia UD, Jones RG, Giguère V, and St-Pierre J
- Abstract
Background: Glutamine metabolism is a central metabolic pathway in cancer. Recently, reductive carboxylation of glutamine for lipogenesis has been shown to constitute a key anabolic route in cancer cells. However, little is known regarding central regulators of the various glutamine metabolic pathways in cancer cells., Methods: The impact of PGC-1α and ERRα on glutamine enzyme expression was assessed in ERBB2+ breast cancer cell lines with quantitative RT-PCR, chromatin immunoprecipitation, and immunoblotting experiments. Glutamine flux was quantified using 13C-labeled glutamine and GC/MS analyses. Functional assays for lipogenesis were performed using 14C-labeled glutamine. The expression of glutamine metabolism genes in breast cancer patients was determined by bioinformatics analyses using The Cancer Genome Atlas., Results: We show that the transcriptional coactivator PGC-1α, along with the transcription factor ERRα, is a positive regulator of the expression of glutamine metabolism genes in ERBB2+ breast cancer. Indeed, ERBB2+ breast cancer cells with increased expression of PGC-1α display elevated expression of glutamine metabolism genes. Furthermore, ERBB2+ breast cancer cells with reduced expression of PGC-1α or when treated with C29, a pharmacological inhibitor of ERRα, exhibit diminished expression of glutamine metabolism genes. The biological relevance of the control of glutamine metabolism genes by the PGC-1α/ERRα axis is demonstrated by consequent regulation of glutamine flux through the citric acid cycle. PGC-1α and ERRα regulate both the canonical citric acid cycle (forward) and the reductive carboxylation (reverse) fluxes; the latter can be used to support de novo lipogenesis reactions, most notably in hypoxic conditions. Importantly, murine and human ERBB2+ cells lines display a significant dependence on glutamine availability for their growth. Finally, we show that PGC-1α expression is positively correlated with that of the glutamine pathway in ERBB2+ breast cancer patients, and high expression of this pathway is associated with reduced patient survival., Conclusions: These data reveal that the PGC-1α/ERRα axis is a central regulator of glutamine metabolism in ERBB2+ breast cancer. This novel regulatory link, as well as the marked reduction in patient survival time associated with increased glutamine pathway gene expression, suggests that targeting glutamine metabolism may have therapeutic potential in the treatment of ERBB2+ breast cancer.
- Published
- 2013
- Full Text
- View/download PDF
36. mTORC1 controls mitochondrial activity and biogenesis through 4E-BP-dependent translational regulation.
- Author
-
Morita M, Gravel SP, Chénard V, Sikström K, Zheng L, Alain T, Gandin V, Avizonis D, Arguello M, Zakaria C, McLaughlan S, Nouet Y, Pause A, Pollak M, Gottlieb E, Larsson O, St-Pierre J, Topisirovic I, and Sonenberg N
- Subjects
- Adenosine Triphosphate biosynthesis, Animals, Autophagy genetics, Cell Cycle Proteins, Cell Nucleus metabolism, Cell Respiration, DNA, Mitochondrial biosynthesis, DNA-Binding Proteins metabolism, Genome, Human genetics, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Mechanistic Target of Rapamycin Complex 1, Mechanistic Target of Rapamycin Complex 2, Mice, Mitochondrial Proteins metabolism, Models, Biological, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, RNA, Messenger genetics, RNA, Messenger metabolism, Ribosomal Protein S6 Kinases metabolism, Transcription Factors metabolism, Adaptor Proteins, Signal Transducing metabolism, Eukaryotic Initiation Factors metabolism, Gene Expression Regulation, Mitochondria metabolism, Mitochondrial Turnover, Multiprotein Complexes metabolism, Phosphoproteins metabolism, Protein Biosynthesis, TOR Serine-Threonine Kinases metabolism
- Abstract
mRNA translation is thought to be the most energy-consuming process in the cell. Translation and energy metabolism are dysregulated in a variety of diseases including cancer, diabetes, and heart disease. However, the mechanisms that coordinate translation and energy metabolism in mammals remain largely unknown. The mechanistic/mammalian target of rapamycin complex 1 (mTORC1) stimulates mRNA translation and other anabolic processes. We demonstrate that mTORC1 controls mitochondrial activity and biogenesis by selectively promoting translation of nucleus-encoded mitochondria-related mRNAs via inhibition of the eukaryotic translation initiation factor 4E (eIF4E)-binding proteins (4E-BPs). Stimulating the translation of nucleus-encoded mitochondria-related mRNAs engenders an increase in ATP production capacity, a required energy source for translation. These findings establish a feed-forward loop that links mRNA translation to oxidative phosphorylation, thereby providing a key mechanism linking aberrant mTOR signaling to conditions of abnormal cellular energy metabolism such as neoplasia and insulin resistance., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
37. LKB1 is a central regulator of tumor initiation and pro-growth metabolism in ErbB2-mediated breast cancer.
- Author
-
Dupuy F, Griss T, Blagih J, Bridon G, Avizonis D, Ling C, Dong Z, Siwak DR, Annis MG, Mills GB, Muller WJ, Siegel PM, and Jones RG
- Abstract
Background: Germline and somatic mutations in STK11, the gene encoding the serine/threonine kinase LKB1, are strongly associated with tumorigenesis. While loss of LKB1 expression has been linked to breast cancer, the mechanistic role of LKB1 in regulating breast cancer development, metastasis, and tumor metabolism has remained unclear., Methods: We have generated and analyzed transgenic mice expressing ErbB2 in the mammary epithelium of LKB1 wild-type or LKB1-deficient mice. We have also utilized ErbB2-expressing breast cancer cells in which LKB1 levels have been reduced using shRNA approaches. These transgenic and xenograft models were characterized for the effects of LKB1 loss on tumor initiation, growth, metastasis and tumor cell metabolism., Results: We demonstrate that loss of LKB1 promotes tumor initiation and induces a characteristic shift to aerobic glycolysis ('Warburg effect') in a model of ErbB2-mediated breast cancer. LKB1-deficient breast cancer cells display enhanced early tumor growth coupled with increased cell migratory and invasive properties in vitro. We show that ErbB2-positive tumors deficient for LKB1 display a pro-growth molecular and phenotypic signature characterized by elevated Akt/mTOR signaling, increased glycolytic metabolism, as well as increased bioenergetic markers both in vitro and in vivo. We also demonstrate that mTOR contributes to the metabolic reprogramming of LKB1-deficient breast cancer, and is required to drive glycolytic metabolism in these tumors; however, LKB1-deficient breast cancer cells display reduced metabolic flexibility and increased apoptosis in response to metabolic perturbations., Conclusions: Together, our data suggest that LKB1 functions as a tumor suppressor in breast cancer. Loss of LKB1 collaborates with activated ErbB2 signaling to drive breast tumorigenesis and pro-growth metabolism in the resulting tumors.
- Published
- 2013
- Full Text
- View/download PDF
38. AMPK is a negative regulator of the Warburg effect and suppresses tumor growth in vivo.
- Author
-
Faubert B, Boily G, Izreig S, Griss T, Samborska B, Dong Z, Dupuy F, Chambers C, Fuerth BJ, Viollet B, Mamer OA, Avizonis D, DeBerardinis RJ, Siegel PM, and Jones RG
- Subjects
- AMP-Activated Protein Kinases antagonists & inhibitors, AMP-Activated Protein Kinases genetics, Animals, B-Lymphocytes metabolism, Cell Line, Glycolysis, HCT116 Cells, Humans, Hypoxia-Inducible Factor 1, alpha Subunit antagonists & inhibitors, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Kaplan-Meier Estimate, Mice, Mice, Transgenic, Neoplasms metabolism, Neoplasms mortality, Neoplasms pathology, Proto-Oncogene Proteins c-myc metabolism, RNA Interference, RNA, Small Interfering metabolism, Signal Transduction, AMP-Activated Protein Kinases metabolism
- Abstract
AMPK is a metabolic sensor that helps maintain cellular energy homeostasis. Despite evidence linking AMPK with tumor suppressor functions, the role of AMPK in tumorigenesis and tumor metabolism is unknown. Here we show that AMPK negatively regulates aerobic glycolysis (the Warburg effect) in cancer cells and suppresses tumor growth in vivo. Genetic ablation of the α1 catalytic subunit of AMPK accelerates Myc-induced lymphomagenesis. Inactivation of AMPKα in both transformed and nontransformed cells promotes a metabolic shift to aerobic glycolysis, increased allocation of glucose carbon into lipids, and biomass accumulation. These metabolic effects require normoxic stabilization of the hypoxia-inducible factor-1α (HIF-1α), as silencing HIF-1α reverses the shift to aerobic glycolysis and the biosynthetic and proliferative advantages conferred by reduced AMPKα signaling. Together our findings suggest that AMPK activity opposes tumor development and that its loss fosters tumor progression in part by regulating cellular metabolic pathways that support cell growth and proliferation., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
39. The complete targeted profile of the organic acid intermediates of the citric acid cycle using a single stable isotope dilution analysis, sodium borodeuteride reduction and selected ion monitoring GC/MS.
- Author
-
Mamer O, Gravel SP, Choinière L, Chénard V, St-Pierre J, and Avizonis D
- Abstract
The quantitative profiling of the organic acid intermediates of the citric acid cycle (CAC) presents a challenge due to the lack of commercially available internal standards for all of the organic acid intermediates. We developed an analytical method that enables the quantitation of all the organic acids in the CAC in a single stable isotope dilution GC/MS analysis with deuterium-labeled analogs used as internal standards. The unstable α-keto acids are rapidly reduced with sodium borodeuteride to the corresponding stable α-deutero-α-hydroxy acids and these, along with their unlabeled analogs and other CAC organic acid intermediates, are converted to their tert -butyldimethylsilyl derivatives. Selected ion monitoring is employed with electron ionization. We validated this method by treating an untransformed mouse mammary epithelial cell line with well-known mitochondrial toxins affecting the electron transport chain and ATP synthase, which resulted in profound perturbations of the concentration of CAC intermediates.
- Published
- 2013
- Full Text
- View/download PDF
40. PGC-1α promotes the growth of ErbB2/Neu-induced mammary tumors by regulating nutrient supply.
- Author
-
Klimcakova E, Chénard V, McGuirk S, Germain D, Avizonis D, Muller WJ, and St-Pierre J
- Subjects
- Animals, Cell Line, Tumor, Down-Regulation, Female, Glucose analysis, Glucose metabolism, Humans, Mice, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, Unfolded Protein Response, Breast Neoplasms metabolism, Heat-Shock Proteins metabolism, Mammary Neoplasms, Experimental metabolism, Receptor, ErbB-2 metabolism, Trans-Activators metabolism, Transcription Factors metabolism
- Abstract
Cancer cells display an increased reliance on glycolysis despite the presence of sufficient oxygen levels to support mitochondrial functions. In this study, we asked whether ameliorating mitochondrial functions in cancer cells might limit their proliferative capacity. Specifically, we increased mitochondrial metabolism in a murine cellular model of ErbB2/Neu-induced breast cancer by ectopically expressing the transcriptional coactivator peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a master regulator of mitochondrial metabolism. As predicted, ErbB2/Neu cells ectopically expressing PGC-1α displayed an increased level of mitochondrial metabolism and reduced proliferative capacity in vitro, compared with controls. In contrast, ErbB2/Neu cells ectopically expressing PGC-1α formed larger tumors in vivo. These tumors exhibited increased concentrations of glucose and the angiogenic factor VEGF as well as higher expression of ErbB2/Neu compared with controls. We discovered that ErbB2/Neu levels were sensitive to nutrient availability, such that reduced glucose concentrations resulted in diminished ErbB2/Neu protein levels. Therefore, our data indicate that PGC-1α prevents the nutrient-mediated downregulation of ErbB2/Neu in tumors by increasing glucose supply. Mechanistic investigations revealed that the regulation of ErbB2/Neu levels by glucose was mediated by the unfolded protein response (UPR). Incubation of ErbB2/Neu-induced breast cancer cells in limited glucose concentrations or with drugs that activate the UPR led to significant reductions in ErbB2/Neu protein levels. Also, ErbB2/Neu-induced tumors ectopically expressing PGC-1α displayed lowered UPR activation compared with controls. Together, our findings uncover an unexpected link between PGC-1α-mediated nutrient availability, UPR, and ErbB2/Neu levels.
- Published
- 2012
- Full Text
- View/download PDF
41. Alterations in cellular energy metabolism associated with the antiproliferative effects of the ATM inhibitor KU-55933 and with metformin.
- Author
-
Zakikhani M, Bazile M, Hashemi S, Javeshghani S, Avizonis D, St Pierre J, and Pollak MN
- Subjects
- Ataxia Telangiectasia Mutated Proteins, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation, Citric Acid chemistry, Citric Acid Cycle, DNA-Binding Proteins metabolism, Fumarates chemistry, HeLa Cells, Hep G2 Cells, Humans, Hypoglycemic Agents pharmacology, Ketoglutaric Acids chemistry, Malates chemistry, Membrane Potential, Mitochondrial, Metformin pharmacology, Oxygen Consumption, Phosphorylation, Protein Serine-Threonine Kinases metabolism, Succinic Acid chemistry, Tumor Suppressor Proteins metabolism, Antineoplastic Agents pharmacology, Metformin metabolism, Morpholines pharmacology, Pyrones pharmacology
- Abstract
KU-55933 is a specific inhibitor of the kinase activity of the protein encoded by Ataxia telangiectasia mutated (ATM), an important tumor suppressor gene with key roles in DNA repair. Unexpectedly for an inhibitor of a tumor suppressor gene, KU-55933 reduces proliferation. In view of prior preliminary evidence suggesting defective mitochondrial function in cells of patients with Ataxia Telangiectasia (AT), we examined energy metabolism of cells treated with KU-55933. The compound increased AMPK activation, glucose uptake and lactate production while reducing mitochondrial membrane potential and coupled respiration. The stimulation of glycolysis by KU-55933 did not fully compensate for the reduction in mitochondrial functions, leading to decreased cellular ATP levels and energy stress. These actions are similar to those previously described for the biguanide metformin, a partial inhibitor of respiratory complex I. Both compounds decreased mitochondrial coupled respiration and reduced cellular concentrations of fumarate, malate, citrate, and alpha-ketogluterate. Succinate levels were increased by KU-55933 levels and decreased by metformin, indicating that the effects of ATM inhibition and metformin are not identical. These observations suggest a role for ATM in mitochondrial function and show that both KU-55933 and metformin perturb the TCA cycle as well as oxidative phosphorylation.
- Published
- 2012
- Full Text
- View/download PDF
42. Electronic referencing techniques for quantitative NMR: pitfalls and how to avoid them using amplitude-corrected referencing through signal injection.
- Author
-
Mehr K, John B, Russell D, and Avizonis D
- Abstract
NMR spectroscopy can be a superior analytical technique for quantification of compounds dissolved in solution. Traditionally a chemical reference standard of known concentration is added to the sample. The concentration of the solute can then be determined by comparing the signal integrals. However, it can be inconvenient or impossible to use internal references. Electronic referencing was developed to circumvent problems with internal standards and has been used successfully in well-controlled situations. However, it is not always possible or convenient to have samples where the dielectric sample properties do not change from one to the next. We propose a modification of the old electronic referencing technique that takes into account the electronic changes between dissimilar samples. We have called this new technique Amplitude-corrected Referencing Through Signal Injection or ARTSI.
- Published
- 2008
- Full Text
- View/download PDF
43. Improving NMR sensitivity by use of salt-tolerant cryogenically cooled probes.
- Author
-
Robosky LC, Reily MD, and Avizonis D
- Subjects
- Cold Temperature, Equipment Design, Magnetic Resonance Spectroscopy methods, Sensitivity and Specificity, Magnetic Resonance Spectroscopy instrumentation, Magnetic Resonance Spectroscopy standards, Salts
- Published
- 2007
- Full Text
- View/download PDF
44. The Internet for nuclear magnetic resonance spectroscopists.
- Author
-
Avizonis D and Farr-Jones S
- Subjects
- Computer Graphics, Computer Simulation, Congresses as Topic, Directories as Topic, Gene Expression, Isotopes, Macromolecular Substances, Models, Molecular, Models, Theoretical, Nucleic Acids chemistry, Proteins chemistry, Proteins genetics, Software, User-Computer Interface, Internet, Nuclear Magnetic Resonance, Biomolecular
- Published
- 2001
- Full Text
- View/download PDF
45. Structural characterization of d(CAACCCGTTG) and d(CAACGGGTTG) mini-hairpin loops by heteronuclear NMR: the effects of purines versus pyrimidines in DNA hairpins.
- Author
-
Avizonis DZ and Kearns DR
- Subjects
- Base Sequence, Carbohydrates chemistry, Magnetic Resonance Spectroscopy methods, Models, Molecular, Molecular Sequence Data, Molecular Structure, Nucleic Acid Conformation, Protons, Purines chemistry, Pyrimidines chemistry, Thermodynamics, DNA chemistry, Deoxyribonucleotides chemistry
- Abstract
The DNA decamers, d(CAACCCGTTG) and d(CAACGGGTTG) were studied in solution by proton and heteronuclear NMR. Under appropriate conditions of pH, temperature, salt concentration and DNA concentration, both decamers form hairpin conformations with similar stabilities [Avizonis and Kearns (1995) Biopolymers, 35, 187-200]. Both decamers adopt mini-hairpin loops, where the first and last four nucleotides are involved in Watson-Crick hydrogen bonding and the central two nucleotides, CC or GG respectively, form the loop. Through the use of proton-proton, proton-phosphorus and natural abundance proton-carbon NMR experiments, backbone torsion angles (beta, gamma and epsilon), sugar puckers and interproton distances were measured. The nucleotides forming the loops of these decamers were found to stack upon one another in an L1 type of loop conformation. Both show gamma tr and unusual beta torsion angles in the loop-closing nucleotide G7, as expected for mini-hairpin loop formation. Our results indicate that the beta and epsilon torsion angles of the fifth and sixth nucleotides that form the loop and the loop-closing nucleotide G7 are not in the standard trans conformation as found in B-DNA. Although the loop structures calculated from NMR-derived constraints are not well defined, the stacking of the bases in the two different hairpins is different. This difference in the base stacking of the loop may provide an explanation as to why the cytosine-containing hairpin is thermodynamically more stable than the guanine-containing hairpin.
- Published
- 1995
- Full Text
- View/download PDF
46. Kinetic and thermodynamic characterization of DNA duplex-hairpin interconversion for two DNA decamers: d(CAACGGGTTG) and d(CAACCCGTTG).
- Author
-
Avizonis DZ and Kearns DR
- Subjects
- Base Sequence, Kinetics, Magnetic Resonance Spectroscopy, Molecular Sequence Data, Thermodynamics, DNA, Nucleic Acid Conformation, Nucleic Acid Heteroduplexes
- Abstract
The duplex-hairpin interconversion of two DNA decamers, d(CAACGGGTTG) and d(CAACCCGTTG), has been characterized thermodynamically and kinetically by using uv-melting and nmr relaxation methods. Separately, each decamer shows slow exchange between hairpin and duplex conformations. The hairpin conformations have melting points of 47 and 50 degrees C, respectively, and exhibit similar thermodynamic stabilities. The enthalpies of duplex formation, measured by nmr, were found to be very similar (delta HDH = 26 +/- 3 kcal/mole) for both decamers at low salt concentrations (< 50 mM NaCl). However, as the salt concentration was increased the behavior of delta HDH and kinetics is significantly different for each decamer. The d(CAACGGGTTG) decamer forms a duplex containing two central G.G mismatches at high salt and DNA concentration. Based upon the measurement of high interconversion activation energies and a decrease in hairpin formation rate with increasing salt, the interconversion between hairpin and duplex was concluded to proceed by complete strand dissociation. In contrast, the d(CAACCCGTTG) decamer was determined to form a duplex with two centrally located C.C mismatches at pH values less than 6.2, consistent with the formation of a hemiprotonated C+.C mismatch. At pH values greater than 6.4, the hairpin-duplex equilibrium is almost completely shifted toward the hairpin conformation at DNA concentrations of 0.5-7.0 mM and salt concentrations of 10-100 mM. The interconversion of duplex and hairpin conformations was ascertained by means of both kinetic and thermodynamic measurements to proceed by a slightly different mechanism than its complementary decamer. Although the interconversion proceeds by complete strand separation as suggested by high duplex-hairpin interconversion activation enthalpies, the increasing hairpin formation rate with increasing ionic strength as well as the delta HDH dependence on salt indicate that an intermediate internally bulged duplex (no C+.C formation) is stabilized by increasing ionic strength. These data support an interconversion mechanism where an intermediate internally bulged duplex may be the rate limiting step before strand separation.
- Published
- 1995
- Full Text
- View/download PDF
47. Assignment of hyperfine-shifted resonances in yeast ferricytochrome c isozyme 2 using the proton pre-steady-state nuclear Overhauser effect.
- Author
-
Satterlee JD, Avizonis DZ, and Moench SJ
- Subjects
- Animals, Cytochromes c2, Horses, Species Specificity, Tuna, Cytochrome c Group isolation & purification, Isoenzymes isolation & purification, Magnetic Resonance Spectroscopy methods, Saccharomyces cerevisiae enzymology
- Abstract
Yeast cytochrome c isozyme 2 is one of two cytochrome c isozymes which are found in yeast mitochondria. Unlike isozyme 1, which can dimerize in vitro due to a free sulfhydryl group at primary sequence position 102, isozyme 2 (Ala-102) is a monomer. The hyperfine proton NMR resonance pattern of ferric isozyme 2 is somewhat different from the horse and tuna ferricytochromes c. Thus, resonance assignments would help determine how similar the yeast, horse and tuna proteins actually are. In this work, many of the unassigned proton hyperfine resonances of the ferric protein have been assigned using the proton pre-steady-state nuclear Overhauser effect. Assigned resonances include those attributable to the heme 7-propionic acid alpha-CH2, His-18 alpha-CH and beta-CH2, Met-80 beta-CH2, and heme 4-beta-CH3 protons. The overall pattern of NOE connectivities is similar to the horse and tuna proteins. Combining shift and NOE patterns leads to the conclusion that the heme environment of yeast ferric isozyme 2 in solution is similar, but not identical, to the heme environment of horse and tuna cytochromes c.
- Published
- 1988
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.