期刊论文详细信息
Cell & Bioscience
Influence of partial and complete glutamine-and glucose deprivation of breast-and cervical tumorigenic cell lines
Anna Margaretha Joubert2  George Edward Mathews1  Edward Henry Mathews1  Leon Liebenberg1  Thandi Vuyelwa Mqoco2  Michelle Helen Visagie2 
[1] Centre for Research and Continued Engineering Development, North-West University, Lynnwood Ridge, South Africa;Department of Physiology, University of Pretoria, Private Bag X323, Arcadia 0007, South Africa
关键词: Glucose;    Glutamine;    Glycolysis;    Reactive oxygen species;    Morphology;    Apoptosis;    Metabolism;   
Others  :  1223179
DOI  :  10.1186/s13578-015-0030-1
 received in 2015-05-07, accepted in 2015-06-26,  发布年份 2015
PDF
【 摘 要 】

Background

Due to their high proliferative requirements, tumorigenic cells possess altered metabolic systems whereby cells utilize higher quantities of glutamine and glucose. These altered metabolic requirements make it of interest to investigate the effects of physiological non-tumorigenic concentrations of glucose and glutamine on tumorigenic cells since deprivation of either results in a canonical amino acid response in mammalian cell.

Methods

The influence of short-term exposure of tumorigenic cells to correlating decreasing glutamine- and glucose quantities were demonstrated in a highly glycolytic metastatic breast cell line and a cervical carcinoma cell line. Thereafter, cells were propagated in medium containing typical physiological concentrations of 1 mM glutamine and 6 mM glucose for 7 days. The effects on morphology were investigated by means of polarization-optical transmitted light differential interference contrast. Flow cytometry was used to demonstrate the effects of glutamine-and glucose starvation on cell cycle progression and apoptosis induction. Fluorometrics were also conducted to investigate the effects on intrinsic apoptosis induction (mitocapture), reactive oxygen species production (2,7-dichlorofluorescein diacetate) and acidic vesicle formation (acridine orange).

Results

Morphological data suggests that glutamine-and glucose deprivation resulted in reduced cell density and rounded cells. Glutamine-and glucose starvation also resulted in an increase in the G 2 M phase and a sub-G 1peak. Complete starvation of glutamine and glucose resulted in the reduction of the mitochondrial membrane potential in both cell lines with MDA-MB-231 cells more prominently affected when compared to HeLa cells. Further, starved cells could not be rescued sufficiently by propagating since cells possessed an increase in reactive oxygen species, acidic compartments and vacuole formation.

Conclusion

Starvation from glutamine and glucose for short periods resulted in decreased cell density, rounded cells and apoptosis induction by means of reactive oxygen species generation and mitochondrial dysfunction. In addition, the metastatic cell line reacted more prominently to glutamine-and glucose starvation due to their highly glycolytic nature. Satisfactory cellular rescue was not possible as cells demonstrated oxidative stress and depolarized mitochondrial membrane potential. This study contributes to the knowledge regarding the in vitro effects and signal transduction of glucose and/or L-glutamine deprivation in tumorigenic cell lines.

【 授权许可】

   
2015 Visagie et al.

【 预 览 】
附件列表
Files Size Format View
20150901040102496.pdf 2917KB PDF download
Fig. 6. 68KB Image download
Fig. 5. 64KB Image download
Fig. 4. 69KB Image download
Fig. 3. 144KB Image download
Fig. 2. 117KB Image download
Fig. 1. 53KB Image download
【 图 表 】

Fig. 1.

Fig. 2.

Fig. 3.

Fig. 4.

Fig. 5.

Fig. 6.

【 参考文献 】
  • [1]Hensley CT, Wasti AT, BeBeradinis RJ. Glutamine and cancer: cell biology, physiology and clinical opportunities. J Clin Invest. 2015; 123:3678-3694.
  • [2]Cairns RA, Harris IS, Mak TW. Regulation of cancer cell metabolism. Nat Rev Cancer. 2011; 11:85-95.
  • [3]Shanware NP, Bray K, Abraham RT. The PI3K, metabolic, and autophagy networks: interactive partners in cellular health and disease. Annu Rev Pharmacol Toxicol. 2013; 53:89-106.
  • [4]Lozy F, Karantza VG. Autophagy and cancer cell metabolism. Semin Cell Dev Biol. 2012; 23:395-401.
  • [5]Qie S, Liang D, Yin C, Gu W, Meng M, Wang C et al.. Glutamine deprivation and glucose depletion trigger growth inhibition via distinctive gene expression reprogramming. Cell Cycle. 2012; 11:3679-3690.
  • [6]Kung H-N, Marks JR, Chi J-T. Glutamine synthetase is a genetic determinant of cell type-specific glutamine independence in breast epithelia. PLoS Genet. 2011; 7:e1002229.
  • [7]Sottnik JL, Lori JC, Rose BJ, Thamm DH. Glysolysis inhibition by 2-deoxy-D-glucose reverts the metastatic phenotype in vitro and in vivo. Clin Exp Metastasis. 2011; 28:865-875.
  • [8]Rahbari R, Sheahan T, Modes V, Collier P, Macfarlane C, Badge RM. A novel L1 retrotransposon marker for HeLa cell line identification. Biotechniques. 2009; 46:277-284.
  • [9]Jose C, Bellance N, Rossignoi R. Choosing between glycolysis and oxidative phosphorylation: a tumor’s dilemma? Biochim Biophys Acta. 1807; 2011:552-561.
  • [10]Rossignol R, Gilkerson R, Aggeler R, Yamagata K, Remington SJ, Capaldi RA. Energy substrate modulates mitochondrial structure and oxidative capacity in cancer cells. Cancer Res. 2014; 64:986-993.
  • [11]Corkery B, Crown J, Clynes M, Donovan N. Epidermal growth factor receptor as a potential therapeutic target in triple-negative breast cancer. Ann Oncol. 2009; 20:862-867.
  • [12]Hall DD, Wu Y, Domann FE, Spitz DR, Anderson ME. Mitochondrial calcium uniporter activity is dispensable for MDA-MB-231 breast carcinoma cell survival. PLoS One. 2014; 9:e96866.
  • [13]Liu B, Fan Z, Edgerton SM, Deng XS, Alimova IN, Lind SE et al.. Metformin induces unique biological and molecular responses in triple negative breast cancer cells. Cell Cycle. 2009; 8:2031-2040.
  • [14]Robey IF, Lien AD, Welsh SJ, Baggett BK, Gillies RJ. Hypoxia-inducible factor-1α and glycolytic phenotype in tumors. Neoplasia. 2005; 7:324-330.
  • [15]Palorini R, Simonetto T, Cirulli C, Chiaradonna F. Mitochondrial complex 1 inhibitors and forced oxidative phosphorylation synergize in inducing cancer cell death. Int J Cell Biol. 2013; doi:10.1155/2013/243876.
  • [16]Mqoco TV, Visagie MH, Albrecht C, Joubert AM. Differential cellular interaction of Sutherlandia frutescens extracts on tumorigenic and non-tumorigenic breast cells. S Afr J Bot. 2014; 90:59-67.
  • [17]Visagie MH, Birkholtz LM, Joubert AM. 17-Beta-estradiol analog inhibits cell proliferation by induction apoptosis in breast cell lines. Microsc Res Tech. 2014; 77:236-242.
  • [18]Ekim B, Magnuson B, Acosta-Jaquez HA, Keller JA, Feener EP, Fingar DC. mTOR Kinase domain phosphorylation promotes mTORC1 signaling, cell growth, and cell cycle progression. Mol Cell Biol. 2011; 31:2878-2801.
  • [19]Yoon JC, Ling AJY, Isik M, Lee DD, Steinbaugh MJ, Sack LM et al.. GLTSCR2/PICT1 links mitochondrial stress and Myc signaling. Proc Natl Acad Sci U S A. 2014; 11:3781-3786.
  • [20]Takai N, Ueda T, Nishida M, Nasu K, Nahara H. Bufalin induces growth inhibition, cell cycle arrest and apoptosis in human endometrial and ovarian cancer cells. Int J Mol Med. 2008; 21:637-643.
  • [21]Costa A, Scholer-Dahirel A, Mechta-Grigoriou F. The role of reactive oxygen species and metabolism on cancer cells and their microenvironment. Semin Cancer Biol. 2014; 26:23-32.
  • [22]Yao ZF, Cao J, Xu LM, Sun XC, Kang J, Yang G et al.. Perfluorooctane sulfonate blocked autophagy flux and induced lysosome membrane permeabilization in HepG2 cells. Food Chem Toxicol. 2014; 67:96-104.
  • [23]Kusuzaki K, Murata H, Takeshita H, Hashiguchi S, Nozaki T, Emoto K et al.. Intracellular binding sites of acridine orange in living osteosarcoma cells. Anticancer Res. 2000; 20:971-975.
  • [24]Koppenol WH, Bounds PL, Dang CV. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat Rev Cancer. 2011; 11:325-337.
  • [25]Lunt SY, Vander Heiden MG. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu Rev Cell Dev Biol. 2011; 27:441-464.
  • [26]Brand K. Aerobic glycolysis by proliferating cells: protection against oxidative stress at the expense of energy yield. J Bioenerg Biomembr. 1997; 29:355-364.
  • [27]Spitz DRS, Sim J, Didnour LA, Galforo SS, Lee YJ. Glucose deprivation-induced oxidative stress in human tumor cells: a fundamental defect in metabolism. Ann NY Acad Sci. 2006; 899:349-362.
  • [28]Lee YJ, Galoforo SS, Berns CM, Chen JC, Davis BH, Sim JE et al.. Glucose deprivation-induced cytotoxicity and alterations in mitogen-activated protein kinase activation are mediated by oxidative stress in multidrug-resistant human breast carcinoma cells. J Biol Chem. 1998; 273:5294-5299.
  • [29]Kansara M, Berridge MV. Oncogenes modulate cell sensitivity to apoptosis induced by glucose deprivation. Anticancer Res. 2004; 24:2503-2510.
  • [30]Schlappack OK, Zimmermann A, Hill RP. Glucose starvation and acidosis: effect on experimental metastatic potential, DNA content and MTX resistance of murine tumor cells. Br J Cancer. 1991; 64:663-670.
  • [31]Landes T, Martinou JC. Mitochondrial outer membrane permeabilization during apoptosis: the role of mitochondrial fission. Biochim Biophys Acta. 2011; 1813:540-5.
  • [32]Abcouwer SF, Schwarz C, Mequid RA. Glutamine deprivation induces the expression of GADD45 and GADD153 primarily by mRNA stabilization. J Biol Chem. 1999; 274:28645-28651.
  • [33]Saqcena, S Mukhopadhyay S, Hosny C, Alhamed A, Chatterjee A, Foster DA. Blocking anelerotic entry of glutamine into the TCA cycle sensitizes K-Ras mutant cancer cells to cytotoxic drugs. Oncogene. 2014; doi:10.1038/onc.2014.207.
  • [34]Yuneva M. Finding an “Achilles heel” of cancer. Cell Cycle. 2008; 7:2083-2089.
  • [35]Rambold AS, Kostelecky B, Elia N, Lippincott-Swartz J. Tubular network formation protects mitochondria from autophagosomal degradation during nutrient starvation. Proc Natl Acad Sci U S A. 2011; 108(5):101902-110195.
  • [36]Ahmad S, White CW, Chang L-Y, Schneider BK, Allen CB. Glutamine protects mitochondria structure and function in oxygen toxicity. Am J Physiol. 2001; 280(4):L779-L791.
  • [37]Maldonado AC, Muñoz-Pinedo C. Dying for something to eat: how cells respond to starvation. Open Cell Sign J. 2011; 3:13-23.
  • [38]Fuchs BC, Bode BP. Stressing out over survival: glutamine as an apoptotic modulator. J Surg Res. 2006; 131:26-40.
  • [39]Paquette JC, Guérin PJ, Gauthier ER. Rapid induction of the intrinsic apoptotic pathway by L-glutamine starvation. J Cell Physiol. 2004; 202:912-921.
  • [40]Fumarola C, Zerbini A, Guidotti GG. Glutamine deprivation-mediated cell shrinkage induces ligand-independent CD95 receptor signaling and apoptosis. Cell Death Differ. 2001; 8:1004-1013.
  • [41]Blackburn RV, Spitz D, Liu X, Galoforo SS, Sim JE, Ridnour LA et al.. Metabolic oxidative stress activates signal transduction and gene expression during glucose deprivation in human tumor cells. Free Radic Biol Med. 1999; 26:419-430.
  • [42]Lord-Fontaine S, Averill-Bates DA. Heat shock inactivates cellular antioxidant defences against hydrogen peroxide: protection by glucose. Free Radic Biol Med. 2002; 32:762-766.
  • [43]Owada S, Shimoda Y, Tsuchihara K, Esumi H. Critical role of H2O2 generated by NOX4 during cellular response under glucose deprivation. PLoS One. 2013; 8:e56628.
  • [44]Song JJ, Lee YL. Catalase but not MNSOD, inhibits glucose-activated ASK1-MEK-MAPK signal transduction pathway and prevents relocalization of DAXX: hydrogen peroxide as a major second messenger of metabolic oxidative stress. J Cell Biochem. 2003; 90:304-314.
  • [45]Aki T, Yamaguchi K, Fujimiya T, Mizukami Y. Phosphoinositide 3-kinase accelerates autophagic cell death during glucose deprivation in rat cardiomyocyte-derived cell line H9c2. Oncogene. 2003; 22:8529-8535.
  文献评价指标  
  下载次数:37次 浏览次数:16次