全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
Cancers  2013 

Metabolomic Dynamic Analysis of Hypoxia in MDA-MB-231 and the Comparison with Inferred Metabolites from Transcriptomics Data

DOI: 10.3390/cancers5020491

Keywords: 1H-NMR spectroscopy, metabolic network, metabolomics, multivariate analysis, tumor hypoxia

Full-Text   Cite this paper   Add to My Lib

Abstract:

Hypoxia affects the tumor microenvironment and is considered important to metastasis progression and therapy resistance. Thus far, the majority of global analyses of tumor hypoxia responses have been limited to just a single omics level. Combining multiple omics data can broaden our understanding of tumor hypoxia. Here, we investigate the temporal change of the metabolite composition with gene expression data from literature to provide a more comprehensive insight into the system level in response to hypoxia. Nuclear magnetic resonance spectroscopy was used to perform metabolomic profiling on the MDA-MB-231 breast cancer cell line under hypoxic conditions. Multivariate statistical analysis revealed that the metabolic difference between hypoxia and normoxia was similar over 24 h, but became distinct over 48 h. Time dependent microarray data from the same cell line in the literature displayed different gene expressions under hypoxic and normoxic conditions mostly at 12 h or earlier. The direct metabolomic profiles show a large overlap with theoretical metabolic profiles deduced from previous transcriptomic studies. Consistent pathways are glycolysis/gluconeogenesis, pyruvate, purine and arginine and proline metabolism. Ten metabolic pathways revealed by metabolomics were not covered by the downstream of the known transcriptomic profiles, suggesting new metabolic phenotypes. These results confirm previous transcriptomics understanding and expand the knowledge from existing models on correlation and co-regulation between transcriptomic and metabolomics profiles, which demonstrates the power of integrated omics analysis.

References

[1]  Hiraga, T.; Kizaka-Kondoh, S.; Hirota, K.; Yoneda, T. Hypoxia and hypoxia-inducible factor-1 expression enhance osteolytic bone metastases of breast cancer. Cancer Res. 2007, 67, 4157–4163, doi:10.1158/0008-5472.CAN-06-2355.
[2]  Semenza, G.L. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 2009, 29, 625–634, doi:10.1038/onc.2009.441.
[3]  Zhong, H.; de Marzo, A.M.; Laughner, E.; Lim, M.; Hilton, D.A.; Zagzag, D.; Buechler, P.; Isaacs, W.B.; Semenza, G.L.; Simons, J.W. Overexpression of hypoxia-inducible factor 1 alpha in common human cancers and their metastases. Cancer Res. 1999, 59, 5830–5835.
[4]  Erb, G.; Elbayed, K.; Piotto, M.; Raya, J.; Neuville, A.; Mohr, M.; Maitrot, D.; Kehrli, P.; Namer, I.J. Toward improved grading of malignancy in oligodendrogliomas using metabolomics. Magn. Reson. Med. 2008, 59, 959–965, doi:10.1002/mrm.21486.
[5]  Chan, E.C.Y.; Koh, P.K.; Mal, M.; Cheah, P.Y.; Eu, K.W.; Backshall, A.; Cavill, R.; Nicholson, J.K.; Keun, H.C. Metabolic profiling of human colorectal cancer using high-resolution magic angle spinning nuclear magnetic resonance (HR-MAS NMR) spectroscopy and gas chromatography mass spectrometry (GC/MS). J. Proteome Res. 2009, 8, 352–361, doi:10.1021/pr8006232.
[6]  Lundgren, K.; Holm, C.; Landberg, G. Hypoxia and breast cancer: Prognostic and therapeutic implications. Cell. Mol. Life Sci. 2007, 64, 3233–3247, doi:10.1007/s00018-007-7390-6.
[7]  Tennant, D.A.; Duran, R.V.; Boulahbel, H.; Gottlieb, E. Metabolic transformation in cancer. Carcinogenesis 2009, 30, 1269–1280, doi:10.1093/carcin/bgp070.
[8]  Brizel, D.M.; Scully, S.P.; Harrelson, J.M.; Layfield, L.J.; Bean, J.M.; Prosnitz, L.R.; Dewhirst, M.W. Tumor oxygenation predicts for the likelihood of distant metastases in human soft tissue sarcoma. Cancer Res. 1996, 56, 941–943.
[9]  Raghunand, N.; Gatenby, R.A.; Gillies, R.J. Microenvironmental and cellular consequences of altered blood flow in tumours. Br. J. Radiol. 2003, 76, S11–S22, doi:10.1259/bjr/12913493.
[10]  Fulda, S.; Debatin, K.M. HIF-1-regulated glucose metabolism a key to apoptosis resistance? Cell Cycle 2007, 6, 790–792, doi:10.4161/cc.6.7.4084.
[11]  Weidner, N.; Semple, N.; Welch, W.R.; Folkman, J. Tumor angiogenesis and metastasis-correlation in invasive breast-carcinoma. N. Engl. J. Med. 1991, 324, 1–8, doi:10.1056/NEJM199101033240101.
[12]  Galanis, A.; Pappa, A.; Giannakakis, A.; Lanitis, E.; Dangaj, D.; Sandaltzopoulos, R. Reactive oxygen species and HIF-1 signalling in cancer. Cancer Lett. 2008, 266, 12–20, doi:10.1016/j.canlet.2008.02.028.
[13]  Brahimi-Horn, C.; Berra, E.; Pouyssegur, J. Hypoxia: The tumor’s gateway to progression along the angiogenic pathway. Trends Cell Biol. 2001, 11, S32–S36.
[14]  Keith, B.; Johnson, R.S.; Simon, M.C. HIF1 alpha and HIF2 alpha: Sibling rivalry in hypoxic tumour growth and progression. Nat. Rev. Cancer 2012, 12, 9–22.
[15]  Nicholson, J.K. Global systems biology, personalized medicine and molecular epidemiology. Mol. Syst. Biol. 2006, 2, doi:10.1038/msb4100095.
[16]  Lenz, E.M.; Bright, J.; Knight, R.; Westwood, F.R.; Davies, D.; Major, H.; Wilson, I.D. Metabonomics with H-1-NMR spectroscopy and liquid chromatography-mass spectrometry applied to the investigation of metabolic changes caused by gentamicin-induced nephrotoxicity in the rat. Biomarkers 2005, 10, 173–187, doi:10.1080/13547500500094034.
[17]  Hara, T.A.; Bansal, T.R.; de Grado, T.R. Effect of hypoxia on the uptake of [methyl-H-3]choline, [1-C-14] acetate and [F-18]FDG in cultured prostate cancer cells. Nucl. Med. Biol. 2006, 33, 977–984, doi:10.1016/j.nucmedbio.2006.08.002.
[18]  Glunde, K.; Shah, T.; Winnard, P.T.; Raman, V.; Takagi, T.; Vesuna, F.; Artemov, D.; Bhujwalla, Z.M. Hypoxia regulates choline kinase expression through hypoxia-inducible factor-lot signaling in a human prostate cancer model. Cancer Res. 2008, 68, 172–180, doi:10.1158/0008-5472.CAN-07-2678.
[19]  Yoshii, Y.; Furukawa, T.; Yoshii, H.; Mori, T.; Kiyono, Y.; Waki, A.; Kobayashi, M.; Tsujikawa, T.; Kudo, T.; Okazawa, H.; et al. Cytosolic acetyl-CoA synthetase affected tumor cell survival under hypoxia: The possible function in tumor acetyl-CoA/acetate metabolism. Cancer Sci. 2009, 100, 821–827, doi:10.1111/j.1349-7006.2009.01099.x.
[20]  Richardson, A.D.; Yang, C.; Osterman, A.; Smith, J.W. Central carbon metabolism in the progression of mammary carcinoma. Breast Cancer Res. Treat. 2008, 110, 297–307, doi:10.1007/s10549-007-9732-3.
[21]  Morse, D.L.; Carroll, D.; Day, S.; Gray, H.; Sadarangani, P.; Murthi, S.; Job, C.; Baggett, B.; Raghunand, N.; Gillies, R.J. Characterization of breast cancers and therapy response by MRS and quantitative gene expression profiling in the choline pathway. NMR Biomed. 2009, 22, 114–127, doi:10.1002/nbm.1318.
[22]  Weljie, A.M.; Bondareva, A.; Zang, P.; Jirik, F.R. 1H-NMR metabolomics identification of markers of hypoxia-induced metabolic shifts in a breast cancer model system. J. Biomol. NMR 2011, 49, 185–193, doi:10.1007/s10858-011-9486-4.
[23]  Troy, H.; Chung, Y.L.; Mayr, M.; Ly, L.; Williams, K.; Stratford, I.; Harris, A.; Griffiths, J.; Stubbs, M. Metabolic profiling of hypoxia-inducible factor-1 beta-deficient and wild type Hepa-1 cells: Effects of hypoxia measured by H-1 magnetic resonance spectroscopy. Metabolomics 2005, 1, 293–303.
[24]  Zhu, J.; Sova, P.; Xu, Q.W.; Dombek, K.M.; Xu, E.Y.; Vu, H.; Tu, Z.D.; Brem, R.B.; Bumgarner, R.E.; Schadt, E.E. Stitching together multiple data dimensions reveals interacting metabolomic and transcriptomic networks that modulate cell regulation. PLoS Biol. 2012, 10, e1001301, doi:10.1371/journal.pbio.1001301.
[25]  Jain, M.; Nilsson, R.; Sharma, S.; Madhusudhan, N.; Kitami, T.; Souza, A.L.; Kafri, R.; Kirschner, M.W.; Clish, C.B.; Mootha, V.K. Metabolite profiling identifies a key role for glycine in rapid cancer cell proliferation. Science 2012, 336, 1040–1044, doi:10.1126/science.1218595.
[26]  Mahadevan, S.; Shah, S.L.; Marrie, T.J.; Slupsky, C.M. Analysis of metabolomic data using support vector machines. Anal. Chem. 2008, 80, 7562–7570, doi:10.1021/ac800954c.
[27]  Greco, O.; Marples, B.; Joiner, M.C.; Scott, S.D. How to overcome (and exploit) tumor hypoxia for targeted gene therapy. J. Cell. Physiol. 2003, 197, 312–325, doi:10.1002/jcp.10374.
[28]  Graeber, T.G.; Peterson, J.F.; Tsai, M.; Monica, K.; Fornace, A.J.; Giaccia, A.J. Hypoxia induces accumulation of P53 protein, but activation of a G(1)-phase checkpoint by low-oxygen conditions is independent of P53 status. Mol. Cell. Biol. 1994, 14, 6264–6277, doi:10.1128/MCB.14.9.6264.
[29]  Koong, A.C.; Chen, E.Y.; Giaccia, A.J. Hypoxia causes the activation of nuclear factor kappa-B through the phosphorylation of I-kappa-B-alpha on tyrosine residues. Cancer Res. 1994, 54, 1425–1430.
[30]  Semenza, G.L.; Wang, G.L. A nuclear factor induced by hypoxia via denovo protein-synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol. Cell. Biol. 1992, 12, 5447–5454.
[31]  Freitas, I.; Bertone, V.; Griffini, P.; Accossato, P.; Baronzio, G.F.; Pontiggia, P.; Stoward, P.J. In situ lactate-dehydrogenase patterns as markers of tumor oxygenation. Anticancer Res. 1991, 11, 1293–1299.
[32]  Semenza, G.L.; Jiang, B.H.; Leung, S.W.; Passantino, R.; Concordet, J.P.; Maire, P.; Giallongo, A. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J. Biol. Chem. 1996, 271, 32529–32537.
[33]  Semenza, G.L.; Roth, P.H.; Fang, H.M.; Wang, G.L. Transcriptional regulation of genes encoding glycolytic-enzymes by hypoxia-inducible factor-1. J. Biol. Chem. 1994, 269, 23757–23763.
[34]  Firth, J.D.; Ebert, B.L.; Pugh, C.W.; Ratcliffe, P.J. Oxygen-regulated control elements in the phosphoglycerate kinase-1 and lactate-dehydrogenase-A genes-similarities with the erythropoietin 3' enhancer. Proc. Natl. Acad. Sci. USA 1994, 91, 6496–6500, doi:10.1073/pnas.91.14.6496.
[35]  Sivitz, W.I.; Lund, D.D.; Yorek, B.; Grovermckay, M.; Schmid, P.G. Pretranslational regulation of 2 cardiac glucose transporters in rats exposed to hypobaric hypoxia. Am. J. Physiol. 1992, 263, E562–E569.
[36]  Mathupala, S.P.; Rempel, A.; Pedersen, P.L. Aberrant glycolytic metabolism of cancer cells: A remarkable coordination of genetic, transcriptional, post-translational, and mutational events that lead to a critical role for Type II hexokinase. J. Bioenerg. Biomembr. 1997, 29, 339–343, doi:10.1023/A:1022494613613.
[37]  Minchenko, A.; Leshchinsky, I.; Opentanova, I.; Sang, N.L.; Srinivas, V.; Armstead, V.; Caro, J. Hypoxia-inducible factor-1-mediated expression of the 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) gene—Its possible role in the Warburg effect. J. Biol. Chem. 2002, 277, 6183–6187.
[38]  Airley, R.; Loncaster, J.; Davidson, S.; Bromley, M.; Roberts, S.; Patterson, A.; Hunter, R.; Stratford, I.; West, C. Glucose transporter Glut-1 expression correlates with tumor hypoxia and predicts metastasis-free survival in advanced carcinoma of the cervix. Clin. Cancer Res. 2001, 7, 928–934.
[39]  Chen, C.H.; Pore, N.; Behrooz, A.; Ismail-Beigi, F.; Maity, A. Regulation of glut1 mRNA by hypoxia-inducible factor-1—Interaction between H-ras and hypoxia. J. Biol. Chem. 2001, 276, 9519–9525.
[40]  Kolev, Y.; Uetake, H.; Takagi, Y.; Sugihara, K. Lactate dehydrogenase-5 (LDH-5) expression in human gastric cancer: Association with hypoxia-inducible factor (HIF-1 alpha) pathway, angiogenic factors production and poor prognosis. Ann. Surg. Oncol. 2008, 15, 2336–2344, doi:10.1245/s10434-008-9955-5.
[41]  Robey, I.F.; Lien, A.D.; Welsh, S.J.; Baggett, B.K.; Gillies, R.J. Hypoxia-inducible factor-1 alpha and the glycolytic phenotype in tumors. Neoplasia 2005, 7, 324–330, doi:10.1593/neo.04430.
[42]  Shiu, R.P.; Watson, P.H.; Dubik, D. C-myc oncognen expression in estrogen-dependent and estrogen-independent breast-cancer. Clin. Chem. 1993, 39, 353–355.
[43]  Wise, D.R.; DeBerardinis, R.J.; Mancuso, A.; Sayed, N.; Zhang, X.Y.; Pfeiffer, H.K.; Nissim, I.; Daikhin, E.; Yudkoff, M.; McMahon, S.B.; et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc. Natl. Acad. Sci. USA 2008, 105, 18782–18787, doi:10.1073/pnas.0810199105.
[44]  Soh, H.; Wasa, M.; Fukuzawa, M. Hypoxia upregulates amino acid transport in a human neuroblastoma cell line. J. Pediatr. Surg. 2007, 42, 608–612, doi:10.1016/j.jpedsurg.2006.12.010.
[45]  Yoshii, Y.; Waki, A.; Furukawa, T.; Kiyono, Y.; Mori, T.; Yoshii, H.; Kudo, T.; Okazawa, H.; Welch, M.J.; Fujibayashi, Y. Tumor uptake of radiolabeled acetate reflects the expression of cytosolic acetyl-CoA synthetase: Implications for the mechanism of acetate PET. Nucl. Med. Biol. 2009, 36, 771–777, doi:10.1016/j.nucmedbio.2009.05.006.
[46]  Loffler, M.; Schneider, F. Lipogenesis in ehrlich ascites tumor-cells under anaerobic culture conditions. J. Cancer Res. Clin. Oncol. 1979, 95, 115–122, doi:10.1007/BF00401005.
[47]  Vavere, A.L.; Kridel, S.J.; Wheeler, F.B.; Lewis, J.S. 1-C-11-acetate as a PET radiopharmaceutical for imaging fatty acid synthase expression in prostate cancer. J. Nucl. Med. 2008, 49, 327–334, doi:10.2967/jnumed.107.046672.
[48]  Furuta, E.; Pai, S.K.; Zhan, R.; Bandyopadhyay, S.; Watabe, M.; Mo, Y.Y.; Hirota, S.; Hosobe, S.; Tsukada, T.; Miura, K.; et al. Fatty acid synthase gene is up-regulated by hypoxia via activation of Akt and sterol regulatory element binding protein-1. Cancer Res. 2008, 68, 1003–1011, doi:10.1158/0008-5472.CAN-07-2489.
[49]  Southam, A.D.; Easton, J.M.; Stentiford, G.D.; Ludwig, C.; Arvanitis, T.N.; Viant, M.R. Metabolic changes in flatfish hepatic tumours revealed by NMR-based metabolomics and metabolic correlation networks. J. Proteome Res. 2008, 7, 5277–5285.
[50]  Israel, M.; Schwartz, L. The metabolic advantage of tumor cells. Mol. Cancer 2011, 10, 70:1–70:12.
[51]  Rofstad, E.K.; Demuth, P.; Fenton, B.M.; Sutherland, R.M. P-31 nuclear magnetic-resonance spectroscopy studies of tumor energy-metabolism and its relationship to intracapillary oxyhemoglobin saturation status and tumor hypoxia. Cancer Res. 1988, 48, 5440–5446.
[52]  Griffin, J.L.; Shockcor, J.P. Metabolic profiles of cancer cells. Nat. Rev. Cancer 2004, 4, 551–561, doi:10.1038/nrc1390.
[53]  El-Sayed, S.; Bezabeh, T.; Odlum, O.; Patel, R.; Ahing, S.; MacDonald, K.; Somorjai, R.L.; Smith, I.C.P. An ex vivo study exploring the diagnostic potential of H-1 magnetic resonance spectroscopy in squamous cell carcinoma of the head and neck region. Head Neck 2002, 24, 766–772, doi:10.1002/hed.10125.
[54]  Moreno, A.; Lopez, L.A.; Fabra, A.; Arus, C. H-1 MRS markers of tumour growth in intrasplenic tumour and liver metastasis induced by injection of HT-29 cells in nude mice spleen. NMR Biomed. 1998, 11, 93–106, doi:10.1002/(SICI)1099-1492(199805)11:3<93::AID-NBM520>3.0.CO;2-H.
[55]  Schaffer, S.W.; Pastukh, V.; Solodushko, V.; Kramer, J.; Azuma, J. Effect of ischemia, calcium depletion and repletion, acidosis and hypoxia on cellular taurine content. Amino Acids 2002, 23, 395–400, doi:10.1007/s00726-002-0201-3.
[56]  Gatenby, R.A.; Smallbone, K.; Maini, P.K.; Rose, F.; Averill, J.; Nagle, R.B.; Worrall, L.; Gillies, R.J. Cellular adaptations to hypoxia and acidosis during somatic evolution of breast cancer. Br. J. Cancer 2007, 97, 646–653, doi:10.1038/sj.bjc.6603922.
[57]  Beckonert, O.; Monnerjahn, K.; Bonk, U.; Leibfritz, D. Visualizing metabolic changes in breast-cancer tissue using 1H-NMR spectroscopy and self-organizing maps. NMR Biomed. 2003, 16, 1–11, doi:10.1002/nbm.797.
[58]  Bando, H.; Toi, M.; Kitada, K.; Koike, M. Genes commonly upregulated by hypoxia in human breast cancer cells MCF-7 and MDA-MB-231. Biomed. Pharmacother. 2003, 57, 333–340, doi:10.1016/S0753-3322(03)00098-2.
[59]  Kanehisa, M.; Goto, S.; Kawashima, S.; Okuno, Y.; Hattori, M. The KEGG resource for deciphering the genome. Nucleic Acids Res. 2004, 32, D277–D280, doi:10.1093/nar/gkh063.
[60]  Dennis, G., Jr.; Sherman, B.T.; Hosack, D.A.; Yang, J.; Gao, W.; Lane, H.C.; Lempicki, R.A. DAVID: Database for annotation, visualization, and integrated discovey. Genome Biol. 2003, 4, P3, doi:10.1186/gb-2003-4-5-p3.
[61]  Christofk, H.R.; Vander Heiden, M.G.; Harris, M.H.; Ramanathan, A.; Gerszten, R.E.; Wei, R.; Fleming, M.D.; Schreiber, S.L.; Cantley, L.C. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 2008, 452, 230–233, doi:10.1038/nature06734.
[62]  Nicholson, J.K.; Wilson, I.D. Understanding “global” systems biology: Metabonomics and the continuum of metabolism. Nat. Rev. Drug Discov. 2003, 2, 668–676, doi:10.1038/nrd1157.
[63]  R Development Core Team. R: A Language and Environment for Statistical Computing; R Foundation for Statistical Computing: Vienna, Austria, 2008.
[64]  Chang, C.; Lin, C. LIBSVM: A Library for Support Vector Machines. ACM Trans. Intell. Syst. Technol. 2001, 2, 27.
[65]  Kanehisa, M.; Goto, S.; Furumichi, M.; Tanabe, M.; Hirakawa, M. KEGG for representation and analysis of molecular networks involving diseases and drugs. Nucleic Acids Res. 2010, 38, D355–D360, doi:10.1093/nar/gkp896.
[66]  Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13, 2498–2504, doi:10.1101/gr.1239303.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413