Skip to main content

Advertisement

Log in

Impact of hexavalent chromium on mammalian cell bioenergetics: phenotypic changes, molecular basis and potential relevance to chromate-induced lung cancer

  • Published:
BioMetals Aims and scope Submit manuscript

Abstract

Occupational exposure to hexavalent chromium [Cr(VI)] has been firmly associated with the development of several pathologies, notably lung cancer. According to the current paradigm, the evolution of normal cells to a neoplastic state is accompanied by extensive metabolic reprogramming, namely at the level of energy-transducing processes. Thus, a complete understanding of the molecular basis of Cr(VI)-induced lung cancer must encompass the elucidation of the impact of Cr(VI) on metabolism. Research in this area is still in its infancy. Nonetheless, Cr(VI)-induced metabolic phenotypes are beginning to emerge. Specifically, it is now well documented that Cr(VI) exposure inhibits respiration and negatively affects the cellular energy status. Furthermore, preliminary results suggest that it also upregulates glucose uptake and lactic acid fermentation. From a mechanistic point of view, there is evidence that Cr(VI) exposure can interfere with energy transducing pathways at different levels, namely gene expression, intracellular protein levels and/or protein function. Loss of thiol redox control likely plays a key role in these processes. The transcriptional networks that control energy transduction can likewise be affected. Data also suggest that Cr(VI) exposure might compromise energy transducing processes through changes in the intracellular pools of their substrates. This article reviews, for the first time, the information available on Cr(VI) impact on mammalian cell bioenergetics. It aims to provide a framework for the understanding of the role played by bioenergetics in Cr(VI)-induced carcinogenesis and is also intended as a guide for future research efforts in this area.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

Notes

  1. EC = ([ATP] + ½[ADP])/[ATP] + [ADP] + [AMP]).

Abbreviations

BDH:

β-Hydroxybutyrate dehydrogenase

Cr(III):

Trivalent chromium

Cr(IV):

Tetravalent chromium

Cr(V):

Pentavalent chromium

Cr(VI):

Hexavalent chromium

DCFH:

Dichlorodihydrofluorescein

DHR:

Dihydrorhodamine

EC:

Energy charge

ETC:

Electron transport chain

FDG-PET:

18-Fluorodeoxyglucose positron emission tomography

G6PDH:

Glucose-6-phosphate dehydrogenase

GAPDH:

Glyceraldehyde-3-phosphate dehydrogenase

GPx:

Glutathione peroxidase

GSH:

Reduced glutathione

GSR:

Glutathione reductase

GST:

Glutathione S-transferase

KGDH:

α-Ketoglutarate dehydrogenase

LDH:

Lactate dehydrogenase NHBE Normal human bronchial epithelial

MCAD:

Medium-chain acyl-CoA dehydrogenase

MDH:

Malate dehydrogenase

NADPH:

Nicotinamide adenine dinucleotide phosphate

OCR:

Oxygen consumption rate

OXPHOS:

Oxidative phosphorylation

PDH:

Pyruvate dehydrogenase

PDK2:

Pyruvate dehydrogenase kinase isoform 2

3-PGK:

3-Phosphoglycerate kinase

PGM:

Phosphoglucomutase

PK:

Pyruvate kinase

PPP:

Pentose phosphate pathway

Prx:

Peroxiredoxin

ROS:

Reactive oxygen species

SCO2:

Synthesis of cytochrome c oxidase 2

SOD:

Superoxide dismutase

TCA:

Tricarboxylic acid

TPI:

Triosephosphate isomerase

Trx:

Thioredoxin

Trx1:

Cytosolic thioredoxin

Trx2:

Mitochondrial thioredoxin

TrxR:

Thioredoxin reductase

TrxR1:

Cytosolic thioredoxin reductase

TrxR2:

Mitochondrial thioredoxin reductase

References

  • Afolaranmi GA, Henderson C, Grant MH (2011) Effect of chromium and cobalt ions on phase I and phase II enzymatic activities in vitro in freshly isolated rat hepatocytes. Toxicol In Vitro 25(1):125–130. doi:10.1016/j.tiv.2010.10.003

    CAS  PubMed  Google Scholar 

  • Ahmad MK, Syma S, Mahmood R (2011) Cr(VI) induces lipid peroxidation, protein oxidation and alters the activities of antioxidant enzymes in human erythrocytes. Biol Trace Elem Res 144(1–3):426–435. doi:10.1007/s12011-011-9119-5

    CAS  PubMed  Google Scholar 

  • Ali AH, Kondo K, Namura T, Senba Y, Takizawa H, Nakagawa Y, Toba H, Kenzaki K, Sakiyama S, Tangoku A (2011) Aberrant DNA methylation of some tumor suppressor genes in lung cancers from workers with chromate exposure. Mol Carcinog 50(2):89–99. doi:10.1002/mc.20697

    CAS  PubMed  Google Scholar 

  • Amedei A, Niccolai E, Benagiano M, Della Bella C, Cianchi F, Bechi P, Taddei A, Bencini L, Farsi M, Cappello P, Prisco D, Novelli F, D’Elios MM (2013) Ex vivo analysis of pancreatic cancer-infiltrating T lymphocytes reveals that ENO-specific Tregs accumulate in tumor tissue and inhibit Th1/Th17 effector cell functions. Cancer Immunol Immunother 62(7):1249–1260. doi:10.1007/s00262-013-1429-3

    CAS  PubMed  Google Scholar 

  • Andreoli T, Daldegan B, Deangelis I, Fortunati E, Reggiani D, Bianchi V, Tiozzo R, Zucco F (1991) Evaluation of metabolic end-points of acute cytotoxicity in V79-fibroblasts. Toxicol In Vitro 5(5–6):549–553. doi:10.1016/0887-2333(91)90091-Q

    CAS  PubMed  Google Scholar 

  • Andrew AS, Warren AJ, Barchowsky A, Temple KA, Klei L, Soucy NV, O’Hara KA, Hamilton JW (2003) Genomic and proteomic profiling of responses to toxic metals in human lung cells. Environ Health Perspect 111(6):825–835. doi:10.1289/ehp.6249

    CAS  PubMed Central  PubMed  Google Scholar 

  • Applegate MA, Humphries KM, Szweda LI (2008) Reversible inhibition of alpha-ketoglutarate dehydrogenase by hydrogen peroxide: glutathionylation and protection of lipoic acid. Biochemistry 47(1):473–478. doi:10.1021/bi7017464

    CAS  PubMed  Google Scholar 

  • Arakawa H, Wu F, Costa M, Rom W, Tang MS (2006) Sequence specificity of Cr(III)-DNA adduct formation in the p53 gene: NGG sequences are preferential adduct-forming sites. Carcinogenesis 27(3):639–645. doi:10.1093/carcin/bgi249

    CAS  PubMed  Google Scholar 

  • Arita A, Costa M (2009) Epigenetics in metal carcinogenesis: nickel, arsenic, chromium and cadmium. Metallomics 1(3):222–228. doi:10.1039/b903049b

    CAS  PubMed Central  PubMed  Google Scholar 

  • Arner ES, Holmgren A (2000) Physiological functions of thioredoxin and thioredoxin reductase. Eur J Biochem 267(20):6102–6109

    CAS  PubMed  Google Scholar 

  • Atkinson DE (1968) The energy charge of the adenylate pool as a regulatory parameter. Interaction with feedback modifiers. Biochemistry 7(11):4030–4034. doi:10.1021/bi00851a033

    CAS  PubMed  Google Scholar 

  • Bagchi D, Bagchi M, Stohs SJ (2001) Chromium (VI)-induced oxidative stress, apoptotic cell death and modulation of p53 tumor suppressor gene. Mol Cell Biochem 222(1–2):149–158. doi:10.1007/978-1-4615-0793-2_18

    CAS  PubMed  Google Scholar 

  • Barceloux DG (1999) Chromium. J Toxicol Clin Toxicol 37(2):173–194. doi:10.1081/CLT-100102418

    CAS  PubMed  Google Scholar 

  • Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, Gottlieb E, Vousden KH (2006) TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 126(1):107–120. doi:10.1016/j.cell.2006.05.036

    CAS  PubMed  Google Scholar 

  • Berg JM, Tymoczko JL, Stryer L (2012) Biochemistry, 7th edn. W. H. Freeman, New York

    Google Scholar 

  • Bianchi V, Dal Toso R, Debetto P, Levis AG, Luciani S, Majone F, Tamino G (1980) Mechanisms of chromium toxicity in mammalian cell cultures. Toxicology 17(2):219–224. doi:10.1016/0300-483X(80)90097-9

    CAS  PubMed  Google Scholar 

  • Bianchi V, Debetto P, Zantedeschi A, Levis AG (1982) Effects of hexavalent chromium on the adenylate pool of hamster fibroblasts. Toxicology 25(1):19–30. doi:10.1016/0300-483X(82)90081-6

    CAS  PubMed  Google Scholar 

  • Biswas S, Chida AS, Rahman I (2006) Redox modifications of protein-thiols: emerging roles in cell signaling. Biochem Pharmacol 71(5):551–564. doi:10.1016/j.bcp.2005.10.044

    CAS  PubMed  Google Scholar 

  • Borthiry GR, Antholine WE, Myers JM, Myers CR (2008) Reductive activation of hexavalent chromium by human lung epithelial cells: generation of Cr(V) and Cr(V)-thiol species. J Inorg Biochem 102(7):1449–1462. doi:10.1016/j.jinorgbio.2007.12.030

    CAS  PubMed Central  PubMed  Google Scholar 

  • Bruick RK, McKnight SL (2001) A conserved family of prolyl-4-hydroxylases that modify HIF. Science 294(5545):1337–1340. doi:10.1126/science.1066373

    CAS  PubMed  Google Scholar 

  • Bushdid PB, Brantley DM, Yull FE, Blaeuer GL, Hoffman LH, Niswander L, Kerr LD (1998) Inhibition of NF-kappaB activity results in disruption of the apical ectodermal ridge and aberrant limb morphogenesis. Nature 392(6676):615–618. doi:10.1038/33435

    CAS  PubMed  Google Scholar 

  • Caglieri A, Goldoni M, De Palma G, Mozzoni P, Gemma S, Vichi S, Testai E, Panico F, Corradi M, Tagliaferri S, Costa LG (2008) Exposure to low levels of hexavalent chromium: target doses and comparative effects on two human pulmonary cell lines. Acta Biomed 79(Suppl 1):104–115

    PubMed  Google Scholar 

  • Carlisle DL, Pritchard DE, Singh J, Owens BM, Blankenship LJ, Orenstein JM, Patierno SR (2000a) Apoptosis and P53 induction in human lung fibroblasts exposed to chromium (VI): effect of ascorbate and tocopherol. Toxicol Sci 55(1):60–68. doi:10.1093/toxsci/55.1.60

    CAS  PubMed  Google Scholar 

  • Carlisle DL, Pritchard DE, Singh J, Patierno SR (2000b) Chromium(VI) induces p53-dependent apoptosis in diploid human lung and mouse dermal fibroblasts. Mol Carcinog 28(2):111–118. doi:10.1002/1098-2744(200006)28:2<111:AID-MC7>3.0.CO;2-Y

    CAS  PubMed  Google Scholar 

  • Casadevall M, da Cruz Fresco P, Kortenkamp A (1999) Chromium(VI)-mediated DNA damage: oxidative pathways resulting in the formation of DNA breaks and abasic sites. Chem Biol Interact 123(2):117–132

    CAS  PubMed  Google Scholar 

  • Cerveira JF, Sánchez-Aragó M, Urbano AM, Cuezva JM (2013) Metabolic Reprogramming elicited by carcinogenic chromium(VI) in human bronchial epithelial cells. Rev Port Pneumol 19:11

    Google Scholar 

  • Chen F, Ding M, Lu Y, Leonard SS, Vallyathan V, Castranova V, Shi X (2000) Participation of MAP kinase p38 and IkappaB kinase in chromium (VI)-induced NF-kappaB and AP-1 activation. J Environ Pathol Toxicol Oncol 19(3):231–238

    PubMed  Google Scholar 

  • Chen F, Bower J, Leonard SS, Ding M, Lu Y, Rojanasakul Y, Kung HF, Vallyathan V, Castranova V, Shi X (2002) Protective roles of NF-kappa B for chromium(VI)-induced cytotoxicity is revealed by expression of Ikappa B kinase-beta mutant. J Biol Chem 277(5):3342–3349. doi:10.1074/jbc.M101089200

    CAS  PubMed  Google Scholar 

  • Colell A, Green DR, Ricci JE (2009) Novel roles for GAPDH in cell death and carcinogenesis. Cell Death Differ 16(12):1573–1581. doi:10.1038/cdd.2009.137

    CAS  PubMed  Google Scholar 

  • Cooks T, Pateras IS, Tarcic O, Solomon H, Schetter AJ, Wilder S, Lozano G, Pikarsky E, Forshew T, Rosenfeld N, Harpaz N, Itzkowitz S, Harris CC, Rotter V, Gorgoulis VG, Oren M (2013) Mutant p53 prolongs NF-kappaB activation and promotes chronic inflammation and inflammation-associated colorectal cancer. Cancer Cell 23(5):634–646. doi:10.1016/j.ccr.2013.03.022

    CAS  PubMed  Google Scholar 

  • Costa M, Salnikow K, Sutherland JE, Broday L, Peng W, Zhang Q, Kluz T (2002) The role of oxidative stress in nickel and chromate genotoxicity. Mol Cell Biochem 234–235(1–2):265–275

    PubMed  Google Scholar 

  • Costa AN, Moreno V, Prieto MJ, Urbano AM, Alpoim MC (2010) Induction of morphological changes in BEAS-2B human bronchial epithelial cells following chronic sub-cytotoxic and mildly cytotoxic hexavalent chromium exposures. Mol Carcinog 49(6):582–591. doi:10.1002/mc.20624

    CAS  PubMed  Google Scholar 

  • Dalle-Donne I, Rossi R, Giustarini D, Milzani A, Colombo R (2003) Protein carbonyl groups as biomarkers of oxidative stress. Clin Chim Acta 329(1–2):23–38

    CAS  PubMed  Google Scholar 

  • Dalle-Donne I, Rossi R, Colombo G, Giustarini D, Milzani A (2009) Protein S-glutathionylation: a regulatory device from bacteria to humans. Trends Biochem Sci 34(2):85–96. doi:10.1016/j.tibs.2008.11.002

    CAS  PubMed  Google Scholar 

  • Dang CV (2012) MYC on the path to cancer. Cell 149(1):22–35. doi:10.1016/j.cell.2012.03.003

    CAS  PubMed Central  PubMed  Google Scholar 

  • Debetto P, Luciani S (1988) Toxic effect of chromium on cellular metabolism. Sci Total Environ 71(3):365–377. doi:10.1016/0048-9697(88)90209-4

    CAS  PubMed  Google Scholar 

  • Debetto P, Dal Toso R, Varotto R, Bianchi V, Luciani S (1982) Effects of potassium dichromate on ATP content of mammalian cells cultured in vitro. Chem Biol Interact 41(1):15–24. doi:10.1016/0009-2797(82)90013-8

    CAS  PubMed  Google Scholar 

  • DeFlora SW, DeFlora KE (1989) Mechanism of chromium metabolism and genotoxicity. Life Chem Rep 7:169–244

    CAS  Google Scholar 

  • Dlugosz A, Rembacz KP, Pruss A, Durlak M, Lembas-Bogaczyk J (2012) Influence of chromium on the natural antioxidant barrier. Pol J Environ Stud 21(2):331–335

    CAS  Google Scholar 

  • Dubrovskaya VA, Wetterhahn KE (1998) Effects of Cr(VI) on the expression of the oxidative stress genes in human lung cells. Carcinogenesis 19(8):1401–1407. doi:10.1093/carcin/19.8.1401

    CAS  PubMed  Google Scholar 

  • Elstrom RL, Bauer DE, Buzzai M, Karnauskas R, Harris MH, Plas DR, Zhuang H, Cinalli RM, Alavi A, Rudin CM, Thompson CB (2004) Akt stimulates aerobic glycolysis in cancer cells. Cancer Res 64(11):3892–3899. doi:10.1158/0008-5472.CAN-03-2904

    CAS  PubMed  Google Scholar 

  • Fatima S, Mahmood R (2007) Vitamin C attenuates potassium dichromate-induced nephrotoxicity and alterations in renal brush border membrane enzymes and phosphate transport in rats. Clin Chim Acta 386(1–2):94–99. doi:10.1016/j.cca.2007.08.006

    CAS  PubMed  Google Scholar 

  • Fearon ER, Vogelstein B (1990) A genetic model for colorectal tumorigenesis. Cell 61(5):759–767. doi:10.1016/0092-8674(90)90186-I

    CAS  PubMed  Google Scholar 

  • Feng Z, Levine AJ (2010) The regulation of energy metabolism and the IGF-1/mTOR pathways by the p53 protein. Trends Cell Biol 20(7):427–434. doi:10.1016/j.tcb.2010.03.004

    CAS  PubMed Central  PubMed  Google Scholar 

  • Fernandes MA, Santos MS, Alpoim MC, Madeira VM, Vicente JA (2002) Chromium(VI) interaction with plant and animal mitochondrial bioenergetics: a comparative study. J Biochem Mol Toxicol 16(2):53–63. doi:10.1002/jbt.10025

    CAS  PubMed  Google Scholar 

  • Ferreira LM (2010) Cancer metabolism: the Warburg effect today. Exp Mol Pathol 89(3):372–380. doi:10.1016/j.yexmp.2010.08.006

    CAS  PubMed  Google Scholar 

  • Ferreira LMR, Guiomar AJ, Santos MS, Alpoim MC, Urbano AM (2011) Impact of carcinogenic chromium(VI) on the energy metabolism of human bronchial epithelial cells. Acta Med Port 24(Suppl 1):P45

    Google Scholar 

  • Fletcher JW, Djulbegovic B, Soares HP, Siegel BA, Lowe VJ, Lyman GH, Coleman RE, Wahl R, Paschold JC, Avril N, Einhorn LH, Suh WW, Samson D, Delbeke D, Gorman M, Shields AF (2008) Recommendations on the use of 18F-FDG PET in oncology. J Nucl Med 49(3):480–508. doi:10.2967/jnumed.107.047787

    PubMed  Google Scholar 

  • Fu J, Liang X, Chen Y, Tang L, Zhang QH, Dong Q (2008) Oxidative stress as a component of chromium-induced cytotoxicity in rat calvarial osteoblasts. Cell Biol Toxicol 24(3):201–212. doi:10.1007/s10565-007-9029-7

    CAS  PubMed  Google Scholar 

  • Gao N, Jiang BH, Leonard SS, Corum L, Zhang Z, Roberts JR, Antonini J, Zheng JZ, Flynn DC, Castranova V, Shi X (2002) p38 Signaling-mediated hypoxia-inducible factor 1alpha and vascular endothelial growth factor induction by Cr(VI) in DU145 human prostate carcinoma cells. J Biol Chem 277(47):45041–45048. doi:10.1074/jbc.M202775200

    CAS  PubMed  Google Scholar 

  • Garcia-Nino WR, Tapia E, Zazueta C, Zatarain-Barron ZL, Hernandez-Pando R, Vega-Garcia CC, Pedraza-Chaverri J (2013) Curcumin pretreatment prevents potassium dichromate-induced hepatotoxicity, oxidative stress, decreased respiratory complex I activity, and membrane permeability transition pore opening. Evid Based Complement Alternat Med 2013:424692. doi:10.1155/2013/424692

    PubMed Central  PubMed  Google Scholar 

  • Gavin IM, Gillis B, Arbieva Z, Prabhakar BS (2007) Identification of human cell responses to hexavalent chromium. Environ Mol Mutagen 48(8):650–657. doi:10.1002/em.20331

    CAS  PubMed  Google Scholar 

  • Gerhauser C (2012) Cancer cell metabolism, epigenetics and the potential influence of dietary components: a perspective. Biomed Res India 23:69–89

    Google Scholar 

  • Goldenthal MJ, Marin-Garcia J (2004) Mitochondrial signaling pathways: a receiver/integrator organelle. Mol Cell Biochem 262(1–2):1–16

    CAS  PubMed  Google Scholar 

  • Goncalves MJ, Santos ACC, Rodrigues CFD, Coelho P, Costa AN, Guiomar AJ, Santos MS, Alpoim MC, Urbano AM (2011) Changes in glucose uptake rate and in the energy status of PC-12 cells acutely exposed to hexavalent chromium, an established human carcinogen. Toxicol Environ Chem 93(6):1202–1211. doi:10.1080/02772248.2011.581340

    CAS  Google Scholar 

  • Hahn WC, Weinberg RA (2002) Modelling the molecular circuitry of cancer. Nat Rev Cancer 2(5):331–341. doi:10.1038/nrc795

    CAS  PubMed  Google Scholar 

  • Hamilton JW, Kaltreider RC, Bajenova OV, Ihnat MA, McCaffrey J, Turpie BW, Rowell EE, Oh J, Nemeth MJ, Pesce CA, Lariviere JP (1998) Molecular basis for effects of carcinogenic heavy metals on inducible gene expression. Environ Health Perspect 106(Suppl 4):1005–1015. doi:10.2307/3434145

    CAS  PubMed Central  PubMed  Google Scholar 

  • Hammond EM, Giaccia AJ (2005) The role of p53 in hypoxia-induced apoptosis. Biochem Biophys Res Commun 331(3):718–725. doi:10.1016/j.bbrc.2005.03.154

    CAS  PubMed  Google Scholar 

  • Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–674. doi:10.1016/j.cell.2011.02.013

    CAS  PubMed  Google Scholar 

  • Hanaoka T, Yamano Y, Katsuno N, Kagawa J, Ishizu S (1997) Elevated serum levels of pantropic p53 proteins in chromium workers. Scand J Work Environ Health 23(1):37–40. doi:10.5271/sjweh.176

    CAS  PubMed  Google Scholar 

  • Harty LC, Guinee DG Jr, Travis WD, Bennett WP, Jett J, Colby TV, Tazelaar H, Trastek V, Pairolero P, Liotta LA, Harris CC, Caporaso NE (1996) p53 mutations and occupational exposures in a surgical series of lung cancers. Cancer Epidemiol Biomarkers Prev 5(12):997–1003

    CAS  PubMed  Google Scholar 

  • Hirose T, Kondo K, Takahashi Y, Ishikura H, Fujino H, Tsuyuguchi M, Hashimoto M, Yokose T, Mukai K, Kodama T, Monden Y (2002) Frequent microsatellite instability in lung cancer from chromate-exposed workers. Mol Carcinog 33(3):172–180. doi:10.1002/mc.10035

    CAS  PubMed  Google Scholar 

  • Hollstein M, Sidransky D, Vogelstein B, Harris CC (1991) p53 mutations in human cancers. Science 253(5015):49–53. doi:10.1126/science.1905840

    CAS  PubMed  Google Scholar 

  • Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z (2010) Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc Natl Acad Sci USA 107(16):7455–7460. doi:10.1073/pnas.1001006107

    CAS  PubMed Central  PubMed  Google Scholar 

  • Hu S, Balakrishnan A, Bok RA, Anderton B, Larson PE, Nelson SJ, Kurhanewicz J, Vigneron DB, Goga A (2011) 13C-pyruvate imaging reveals alterations in glycolysis that precede c-Myc-induced tumor formation and regression. Cell Metab 14(1):131–142. doi:10.1016/j.cmet.2011.04.012

    CAS  PubMed  Google Scholar 

  • Hwang TL, Liang Y, Chien KY, Yu JS (2006) Overexpression and elevated serum levels of phosphoglycerate kinase 1 in pancreatic ductal adenocarcinoma. Proteomics 6(7):2259–2272. doi:10.1002/pmic.200500345

    CAS  PubMed  Google Scholar 

  • IARC (1990) Chromium, nickel and welding. In: IARC monographs on the evaluation of carcinogenic risks to humans, vol 49. IARC Scientific Publications, Lyon

  • Ishii T, Uchida K (2004) Induction of reversible cysteine-targeted protein oxidation by an endogenous electrophile 15-deoxy-delta12,14-prostaglandin J2. Chem Res Toxicol 17(10):1313–1322. doi:10.1021/tx049860+

    CAS  PubMed  Google Scholar 

  • Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, Kriegsheim A, Hebestreit HF, Mukherji M, Schofield CJ, Maxwell PH, Pugh CW, Ratcliffe PJ (2001) Targeting of HIF-alpha to the von Hippel–Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292(5516):468–472. doi:10.1126/science.1059796

    CAS  PubMed  Google Scholar 

  • Jannetto PJ, Antholine WE, Myers CR (2001) Cytochrome b(5) plays a key role in human microsomal chromium(VI) reduction. Toxicology 159(3):119–133

    CAS  PubMed  Google Scholar 

  • Janson CA, Cleland WW (1974a) The inhibition of acetate, pyruvate, and 3-phosphogylcerate kinases by chromium adenosine triphosphate. J Biol Chem 249(8):2567–2571

    CAS  PubMed  Google Scholar 

  • Janson CA, Cleland WW (1974b) The specificity of chromium nucleotides as inhibitors of selected kinases. J Biol Chem 249(8):2572–2574

    CAS  PubMed  Google Scholar 

  • Jennette KW (1982) Microsomal reduction of the carcinogen chromate produces chromium(V). J Am Chem Soc 104(3):874–875. doi:10.1021/ja00367a050

    CAS  Google Scholar 

  • Jones RG, Thompson CB (2009) Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev 23(5):537–548. doi:10.1101/gad.1756509

    CAS  PubMed Central  PubMed  Google Scholar 

  • Kabe Y, Ando K, Hirao S, Yoshida M, Handa H (2005) Redox regulation of NF-kappaB activation: distinct redox regulation between the cytoplasm and the nucleus. Antioxid Redox Signal 7(3–4):395–403. doi:10.1089/ars.2005.7.395

    CAS  PubMed  Google Scholar 

  • Kaczmarek M, Timofeeva OA, Karaczyn A, Malyguine A, Kasprzak KS, Salnikow K (2007) The role of ascorbate in the modulation of HIF-1alpha protein and HIF-dependent transcription by chromium(VI) and nickel(II). Free Radic Biol Med 42(8):1246–1257. doi:10.1016/j.freeradbiomed.2007.01.026

    CAS  PubMed Central  PubMed  Google Scholar 

  • Kalyanaraman B, Darley-Usmar V, Davies KJ, Dennery PA, Forman HJ, Grisham MB, Mann GE, Moore K, Roberts LJ 2nd, Ischiropoulos H (2012) Measuring reactive oxygen and nitrogen species with fluorescent probes: challenges and limitations. Free Radic Biol Med 52(1):1–6. doi:10.1016/j.freeradbiomed.2011.09.030

    CAS  PubMed Central  PubMed  Google Scholar 

  • Katada S, Imhof A, Sassone-Corsi P (2012) Connecting threads: epigenetics and metabolism. Cell 148(1–2):24–28. doi:10.1016/j.cell.2012.01.001

    CAS  PubMed  Google Scholar 

  • Kawauchi K, Araki K, Tobiume K, Tanaka N (2008) p53 regulates glucose metabolism through an IKK-NF-kappaB pathway and inhibits cell transformation. Nat Cell Biol 10(5):611–618. doi:10.1038/ncb1724

    CAS  PubMed  Google Scholar 

  • Khoury MP, Bourdon JC (2010) The isoforms of the p53 protein. Cold Spring Harb Perspect Biol 2(3):a000927. doi:10.1101/cshperspect.a000927

    PubMed Central  PubMed  Google Scholar 

  • Kilburn DG, Lilly MD, Webb FC (1969) The energetics of mammalian cell growth. J Cell Sci 4(3):645–654

    CAS  PubMed  Google Scholar 

  • Kim G, Yurkow EJ (1996) Chromium induces a persistent activation of mitogen-activated protein kinases by a redox-sensitive mechanism in H4 rat hepatoma cells. Cancer Res 56(9):2045–2051

    CAS  PubMed  Google Scholar 

  • Kim YD, An SC, Oyama T, Kawamoto T, Kim H (2003) Oxidative stress, hogg1 expression and NF-kappaB activity in cells exposed to low level chromium. J Occup Health 45(5):271–277

    CAS  PubMed  Google Scholar 

  • Kim JW, Gao P, Liu YC, Semenza GL, Dang CV (2007) Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1. Mol Cell Biol 27(21):7381–7393. doi:10.1128/MCB.00440-07

    CAS  PubMed Central  PubMed  Google Scholar 

  • Klein CB, Su L, Bowser D, Leszczynska J (2002) Chromate-induced epimutations in mammalian cells. Environ Health Perspect 110(Suppl 5):739–743. doi:10.1289/ehp.02110s5739

    CAS  PubMed Central  PubMed  Google Scholar 

  • Klimova T, Chandel NS (2008) Mitochondrial complex III regulates hypoxic activation of HIF. Cell Death Differ 15(4):660–666. doi:10.1038/sj.cdd.4402307

    CAS  PubMed  Google Scholar 

  • Kondo K, Hino N, Sasa M, Kamamura Y, Sakiyama S, Tsuyuguchi M, Hashimoto M, Uyama T, Monden Y (1997) Mutations of the p53 gene in human lung cancer from chromate-exposed workers. Biochem Biophys Res Commun 239(1):95–100. doi:10.1006/bbrc.1997.7425

    CAS  PubMed  Google Scholar 

  • Kondo K, Takahashi Y, Hirose Y, Nagao T, Tsuyuguchi M, Hashimoto M, Ochiai A, Monden Y, Tangoku A (2006) The reduced expression and aberrant methylation of p16(INK4a) in chromate workers with lung cancer. Lung Cancer 53(3):295–302. doi:10.1016/j.lungcan.2006.05.022

    PubMed  Google Scholar 

  • Koutras GA, Hattori M, Schneider AS, Ebaugh FG Jr, Valentine WN (1964) Studies on chromated erythrocytes. Effect of sodium chromate on erythrocyte glutathione reductase. J Clin Invest 43:323–331. doi:10.1172/JCI104917

    CAS  PubMed Central  PubMed  Google Scholar 

  • Kregel KC, Zhang HJ (2007) An integrated view of oxidative stress in aging: basic mechanisms, functional effects, and pathological considerations. Am J Physiol Regul Integr Comp Physiol 292(1):R18–R36. doi:10.1152/ajpregu.00327.2006

    CAS  PubMed  Google Scholar 

  • Landolph JR (1994) Molecular mechanisms of transformation of C3H/10T1/2 C1 8 mouse embryo cells and diploid human fibroblasts by carcinogenic metal compounds. Environ Health Perspect 102(Suppl 3):119–125. doi:10.1289/ehp.94102s3119

    CAS  PubMed Central  PubMed  Google Scholar 

  • Lane DP (1992) p53, guardian of the genome. Nature 358(6381):15–16. doi:10.1038/358015a0

    CAS  PubMed  Google Scholar 

  • Lazzarini A, Luciani S, Beltrame M, Arslan P (1985) Effects of chromium(VI) and chromium(III) on energy charge and oxygen consumption in rat thymocytes. Chem Biol Interact 53(3):273–281. doi:10.1016/S0009-2797(85)80104-6

    CAS  PubMed  Google Scholar 

  • Lei T, He QY, Cai Z, Zhou Y, Wang YL, Si LS, Chiu JF (2008) Proteomic analysis of chromium cytotoxicity in cultured rat lung epithelial cells. Proteomics 8(12):2420–2429. doi:10.1002/pmic.200701050

    CAS  PubMed  Google Scholar 

  • Levine AJ (1997) p53, the cellular gatekeeper for growth and division. Cell 88(3):323–331. doi:10.1016/S0092-8674(00)81871-1

    CAS  PubMed  Google Scholar 

  • Levine AJ, Oren M (2009) The first 30 years of p53: growing ever more complex. Nat Rev Cancer 9(10):749–758. doi:10.1038/nrc2723

    CAS  PubMed Central  PubMed  Google Scholar 

  • Levine AJ, Puzio-Kuter AM (2010) The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science 330(6009):1340–1344. doi:10.1126/science.1193494

    CAS  PubMed  Google Scholar 

  • Li Q, Chen H, Huang X, Costa M (2006) Effects of 12 metal ions on iron regulatory protein 1 (IRP-1) and hypoxia-inducible factor-1 alpha (HIF-1alpha) and HIF-regulated genes. Toxicol Appl Pharmacol 213(3):245–255. doi:10.1016/j.taap.2005.11.006

    CAS  PubMed Central  PubMed  Google Scholar 

  • Liebross RH, Wetterhahn KE (1992) In vivo formation of chromium(V) in chick embryo liver and red blood cells. Carcinogenesis 13(11):2113–2120

    CAS  PubMed  Google Scholar 

  • Lin CY, Loven J, Rahl PB, Paranal RM, Burge CB, Bradner JE, Lee TI, Young RA (2012) Transcriptional amplification in tumor cells with elevated c-Myc. Cell 151(1):56–67. doi:10.1016/j.cell.2012.08.026

    CAS  PubMed Central  PubMed  Google Scholar 

  • Lippard SJ, Berg JM (1997) Principles of bioinorganic chemistry. University Science Books, Herdon

    Google Scholar 

  • Liu KJ, Shi X (2001) In vivo reduction of chromium (VI) and its related free radical generation. Mol Cell Biochem 222(1–2):41–47

    CAS  PubMed  Google Scholar 

  • Liu KJ, Jiang J, Swartz HM, Shi X (1994) Low-frequency EPR detection of chromium(V) formation by chromium(VI) reduction in whole live mice. Arch Biochem Biophys 313(2):248–252

    CAS  PubMed  Google Scholar 

  • Liu KJ, Shi X, Jiang JJ, Goda F, Dalal N, Swartz HM (1995) Chromate-induced chromium(V) formation in live mice and its control by cellular antioxidants: an L-band electron paramagnetic resonance study. Arch Biochem Biophys 323(1):33–39

    CAS  PubMed  Google Scholar 

  • Liu B, Chen Y, St Clair DK (2008) ROS and p53: a versatile partnership. Free Radic Biol Med 44(8):1529–1535. doi:10.1016/j.freeradbiomed.2008.01.011

    CAS  PubMed Central  PubMed  Google Scholar 

  • Liu W, Chaspoul F, Botta C, De Meo M, Gallice P (2010) Bioenergetics and DNA alteration of normal human fibroblasts by hexavalent chromium. Environ Toxicol Pharmacol 29(1):58–63. doi:10.1016/j.etap.2009.10.001

    CAS  PubMed  Google Scholar 

  • Lu J, Holmgren A (2013) The thioredoxin antioxidant system. Free Radic Biol Med. doi:10.1016/j.freeradbiomed.2013.07.036

    Google Scholar 

  • Lubin R, Zalcman G, Bouchet L, Tredanel J, Legros Y, Cazals D, Hirsch A, Soussi T (1995) Serum p53 antibodies as early markers of lung cancer. Nat Med 1(7):701–702. doi:10.1038/nm0795-701

    CAS  PubMed  Google Scholar 

  • Luciani S, Dal Toso R, Rebellato AM, Levis AG (1979) Effects of chromium compounds on plasma membrane Mg2+-ATPase activity of BHK cells. Chem Biol Interact 27(1):59–67. doi:10.1016/0009-2797(79)90149-2

    CAS  PubMed  Google Scholar 

  • Luo J, Kuo MK (2009) Linking nutrient metabolism to epigenetics. Cell Sci Rev 6(1):49–54

    Google Scholar 

  • Martin BD, Schoenhard JA, Sugden KD (1998) Hypervalent chromium mimics reactive oxygen species as measured by the oxidant-sensitive dyes 2′,7′-dichlorofluorescin and dihydrorhodamine. Chem Res Toxicol 11(12):1402–1410. doi:10.1021/tx9801559

    CAS  PubMed  Google Scholar 

  • Matoba S, Kang JG, Patino WD, Wragg A, Boehm M, Gavrilova O, Hurley PJ, Bunz F, Hwang PM (2006) p53 regulates mitochondrial respiration. Science 312(5780):1650–1653. doi:10.1126/science.1126863

    CAS  PubMed  Google Scholar 

  • Meister A, Anderson ME (1983) Glutathione. Annu Rev Biochem 52:711–760. doi:10.1146/annurev.bi.52.070183.003431

    CAS  PubMed  Google Scholar 

  • Messer RL, Lucas LC (1999) Evaluations of metabolic activities as biocompatibility tools: a study of individual ions’ effects on fibroblasts. Dent Mater 15(1):1–6. doi:10.1016/S0109-5641(99)90023-4

    CAS  PubMed  Google Scholar 

  • Messer RL, Doeller JE, Kraus DW, Lucas LC (2000) An investigation of fibroblast mitochondria enzyme activity and respiration in response to metallic ions released from dental alloys. J Biomed Mater Res 50(4):598–604. doi:10.1002/(SICI)1097-4636(20000615)50:4<598:AID-JBM16>3.3.CO;2-R

    CAS  PubMed  Google Scholar 

  • Mitsuuchi Y, Testa JR (2002) Cytogenetics and molecular genetics of lung cancer. Am J Med Genet 115(3):183–188. doi:10.1002/ajmg.10692

    PubMed  Google Scholar 

  • Molina-Jijon E, Tapia E, Zazueta C, El Hafidi M, Zatarain-Barron ZL, Hernandez-Pando R, Medina-Campos ON, Zarco-Marquez G, Torres I, Pedraza-Chaverri J (2011) Curcumin prevents Cr(VI)-induced renal oxidant damage by a mitochondrial pathway. Free Radic Biol Med 51(8):1543–1557. doi:10.1016/j.freeradbiomed.2011.07.018

    CAS  PubMed  Google Scholar 

  • Molina-Jijon E, Zarco-Marquez G, Medina-Campos ON, Zatarain-Barron ZL, Hernandez-Pando R, Pinzon E, Zavaleta RM, Tapia E, Pedraza-Chaverri J (2012) Deferoxamine pretreatment prevents Cr(VI)-induced nephrotoxicity and oxidant stress: role of Cr(VI) chelation. Toxicology 291(1–3):93–101. doi:10.1016/j.tox.2011.11.003

    CAS  PubMed  Google Scholar 

  • Myers CR (2012) The effects of chromium(VI) on the thioredoxin system: implications for redox regulation. Free Radic Biol Med 52(10):2091–2107. doi:10.1016/j.freeradbiomed.2012.03.013

    CAS  PubMed Central  PubMed  Google Scholar 

  • Myers JM, Myers CR (2009) The effects of hexavalent chromium on thioredoxin reductase and peroxiredoxins in human bronchial epithelial cells. Free Radic Biol Med 47(10):1477–1485. doi:10.1016/j.freeradbiomed.2009.08.015

    CAS  PubMed Central  PubMed  Google Scholar 

  • Myers CR, Myers JM, Carstens BP, Antholine WE (2000) Reduction of chromium(VI) to chromium(V) by human microsomal enzymes: effects of iron and quinones. Toxic Subst Mech 19(1):25–51. doi:10.1080/10769180051125734

    CAS  Google Scholar 

  • Myers JM, Antholine WE, Myers CR (2008) Hexavalent chromium causes the oxidation of thioredoxin in human bronchial epithelial cells. Toxicology 246(2–3):222–233. doi:10.1016/j.tox.2008.01.017

    CAS  PubMed Central  PubMed  Google Scholar 

  • Myers CR, Antholine WE, Myers JM (2010) The pro-oxidant chromium(VI) inhibits mitochondrial complex I, complex II, and aconitase in the bronchial epithelium: EPR markers for Fe-S proteins. Free Radic Biol Med 49(12):1903–1915. doi:10.1016/j.freeradbiomed.2010.09.020

    CAS  PubMed Central  PubMed  Google Scholar 

  • Myers JM, Antholine WE, Myers CR (2011) The intracellular redox stress caused by hexavalent chromium is selective for proteins that have key roles in cell survival and thiol redox control. Toxicology 281(1–3):37–47. doi:10.1016/j.tox.2011.01.001

    CAS  PubMed Central  PubMed  Google Scholar 

  • Nair S, Singh SV, Krishan A (1991) Flow cytometric monitoring of glutathione content and anthracycline retention in tumor cells. Cytometry 12(4):336–342. doi:10.1002/cyto.990120408

    CAS  PubMed  Google Scholar 

  • Nemec AA, Barchowsky A (2009) Signal transducer and activator of transcription 1 (STAT1) is essential for chromium silencing of gene induction in human airway epithelial cells. Toxicol Sci 110(1):212–223. doi:10.1093/toxsci/kfp084

    CAS  PubMed Central  PubMed  Google Scholar 

  • Nickens KP, Han Y, Shandilya H, Larrimore A, Gerard GF, Kaldjian E, Patierno SR, Ceryak S (2012) Acquisition of mitochondrial dysregulation and resistance to mitochondrial-mediated apoptosis after genotoxic insult in normal human fibroblasts: a possible model for early stage carcinogenesis. Biochim Biophys Acta 1823(2):264–272. doi:10.1016/j.bbamcr.2011.10.005

    CAS  PubMed Central  PubMed  Google Scholar 

  • Nie Z, Hu G, Wei G, Cui K, Yamane A, Resch W, Wang R, Green DR, Tessarollo L, Casellas R, Zhao K, Levens D (2012) c-Myc is a universal amplifier of expressed genes in lymphocytes and embryonic stem cells. Cell 151(1):68–79. doi:10.1016/j.cell.2012.08.033

    CAS  PubMed Central  PubMed  Google Scholar 

  • Nudler SI, Quinteros FA, Miler EA, Cabilla JP, Ronchetti SA, Duvilanski BH (2009) Chromium VI administration induces oxidative stress in hypothalamus and anterior pituitary gland from male rats. Toxicol Lett 185(3):187–192. doi:10.1016/j.toxlet.2009.01.003

    CAS  PubMed  Google Scholar 

  • O’Brien TJ, Ceryak S, Patierno SR (2003) Complexities of chromium carcinogenesis: role of cellular response, repair and recovery mechanisms. Mutat Res Fundam Mol Mech Mutagen 533(1–2):3–36. doi:10.1016/j.mrfmmm.2003.09.006

    Google Scholar 

  • Pan TL, Wang PW, Chen CC, Fang JY, Sintupisut N (2012) Functional proteomics reveals hepatotoxicity and the molecular mechanisms of different forms of chromium delivered by skin administration. Proteomics 12(3):477–489. doi:10.1002/pmic.201100055

    CAS  PubMed  Google Scholar 

  • Pani G, Galeotti T, Chiarugi P (2010) Metastasis: cancer cell’s escape from oxidative stress. Cancer Metastasis Rev 29(2):351–378. doi:10.1007/s10555-010-9225-4

    CAS  PubMed  Google Scholar 

  • Patlolla AK, Barnes C, Yedjou C, Velma VR, Tchounwou PB (2009) Oxidative stress, DNA damage, and antioxidant enzyme activity induced by hexavalent chromium in Sprague-Dawley rats. Environ Toxico 24(1):66–73. doi:10.1002/tox.20395

    CAS  Google Scholar 

  • Pauls H, Serpersu EH, Kirch U, Schoner W (1986) Chromium(III)ATP inactivating (Na+ + K+)-ATPase supports Na+ − Na+ and Rb+ − Rb+ exchanges in everted red blood cells but not Na+, K+ transport. Eur J Biochem 157(3):585–595

    CAS  PubMed  Google Scholar 

  • Permenter MG, Lewis JA, Jackson DA (2011) Exposure to nickel, chromium, or cadmium causes distinct changes in the gene expression patterns of a rat liver derived cell line. PLoS One 6(11):e27730. doi:10.1371/journal.pone.0027730

    CAS  PubMed Central  PubMed  Google Scholar 

  • Pouyssegur J, Mechta-Grigoriou F (2006) Redox regulation of the hypoxia-inducible factor. Biol Chem 387(10–11):1337–1346. doi:10.1515/BC.2006.167

    CAS  PubMed  Google Scholar 

  • Pouyssegur J, Dayan F, Mazure NM (2006) Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 441(7092):437–443. doi:10.1038/nature04871

    CAS  PubMed  Google Scholar 

  • Powis G, Montfort WR (2001) Properties and biological activities of thioredoxins. Annu Rev Biophys Biomol Struct 30:421–455. doi:10.1146/annurev.biophys.30.1.421

    CAS  PubMed  Google Scholar 

  • Poynton RA, Hampton MB (2014) Peroxiredoxins as biomarkers of oxidative stress. Biochim Biophys Acta 1840(2):906–912. doi:10.1016/j.bbagen.2013.08.001

    CAS  PubMed  Google Scholar 

  • Pritchard DE, Ceryak S, Ramsey KE, O’Brien TJ, Ha L, Fornsaglio JL, Stephan DA, Patierno SR (2005) Resistance to apoptosis, increased growth potential, and altered gene expression in cells that survived genotoxic hexavalent chromium [Cr(VI)] exposure. Mol Cell Biochem 279(1–2):169–181. doi:10.1007/s11010-005-8292-2

    CAS  PubMed Central  PubMed  Google Scholar 

  • Quievryn G, Goulart M, Messer J, Zhitkovich A (2001) Reduction of Cr(VI) by cysteine: significance in human lymphocytes and formation of DNA damage in reactions with variable reduction rates. Mol Cell Biochem 222(1–2):107–118. doi:10.1007/978-1-4615-0793-2_13

    CAS  PubMed  Google Scholar 

  • Quinteros FA, Machiavelli LI, Miler EA, Cabilla JP, Duvilanski BH (2008) Mechanisms of chromium (VI)-induced apoptosis in anterior pituitary cells. Toxicology 249(2–3):109–115. doi:10.1016/j.tox.2008.04.012

    CAS  PubMed  Google Scholar 

  • Raghunathan VK, Tettey JN, Ellis EM, Grant MH (2009) Comparative chronic in vitro toxicity of hexavalent chromium to osteoblasts and monocytes. J Biomed Mater Res A 88(2):543–550. doi:10.1002/jbm.a.31893

    PubMed  Google Scholar 

  • Raghunathan VK, Grant MH, Ellis EM (2010) Changes in protein expression associated with chronic in vitro exposure of hexavalent chromium to osteoblasts and monocytes: a proteomic approach. J Biomed Mater Res A 92(2):615–625. doi:10.1002/jbm.a.32396

    PubMed  Google Scholar 

  • Ramanathan A, Wang C, Schreiber SL (2005) Perturbational profiling of a cell-line model of tumorigenesis by using metabolic measurements. Proc Natl Acad Sci USA 102(17):5992–5997. doi:10.1073/pnas.0502267102

    CAS  PubMed Central  PubMed  Google Scholar 

  • Ramos-Montoya A, Lee WN, Bassilian S, Lim S, Trebukhina RV, Kazhyna MV, Ciudad CJ, Noe V, Centelles JJ, Cascante M (2006) Pentose phosphate cycle oxidative and nonoxidative balance: a new vulnerable target for overcoming drug resistance in cancer. Int J Cancer 119(12):2733–2741. doi:10.1002/ijc.22227

    CAS  PubMed  Google Scholar 

  • Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, Zeng Q, Dillehay LE, Madan A, Semenza GL, Bedi A (2000) Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1alpha. Genes Dev 14(1):34–44. doi:10.1101/gad.14.1.34

    CAS  PubMed Central  PubMed  Google Scholar 

  • Reitman ZJ, Yan H (2010) Isocitrate dehydrogenase 1 and 2 mutations in cancer: alterations at a crossroads of cellular metabolism. J Natl Cancer Inst 102(13):932–941. doi:10.1093/jnci/djq187

    CAS  PubMed Central  PubMed  Google Scholar 

  • Reynolds M, Zhitkovich A (2007) Cellular vitamin C increases chromate toxicity via a death program requiring mismatch repair but not p53. Carcinogenesis 28(7):1613–1620. doi:10.1093/carcin/bgm031

    CAS  PubMed  Google Scholar 

  • Rodrigues CF, Urbano AM, Matoso E, Carreira I, Almeida A, Santos P, Botelho F, Carvalho L, Alves M, Monteiro C, Costa AN, Moreno V, Alpoim MC (2009) Human bronchial epithelial cells malignantly transformed by hexavalent chromium exhibit an aneuploid phenotype but no microsatellite instability. Mutat Res Fundam Mol Mech Mutagen 670(1–2):42–52. doi:10.1016/j.mrfmmm.2009.07.004

    CAS  Google Scholar 

  • Rossi SC, Gorman N, Wetterhahn KE (1988) Mitochondrial reduction of the carcinogen chromate: formation of chromium(V). Chem Res Toxicol 1(2):101–107. doi:10.1021/tx00002a003

    CAS  PubMed  Google Scholar 

  • Ryberg D, Alexander J (1984) Inhibitory action of hexavalent chromium (Cr(VI)) on the mitochondrial respiration and a possible coupling to the reduction of Cr(VI). Biochem Pharmacol 33(15):2461–2466. doi:10.1016/0006-2952(84)90718-4

    CAS  PubMed  Google Scholar 

  • Ryberg D, Alexander J (1990) Mechanisms of chromium toxicity in mitochondria. Chem Biol Interact 75(2):141–151. doi:10.1016/0009-2797(90)90114-3

    CAS  PubMed  Google Scholar 

  • Salmon SE, Hamburger AW, Soehnlen B, Durie BG, Alberts DS, Moon TE (1978) Quantitation of differential sensitivity of human-tumor stem cells to anticancer drugs. N Engl J Med 298(24):1321–1327. doi:10.1056/NEJM197806152982401

    CAS  PubMed  Google Scholar 

  • Salnikow K, Zhitkovich A (2008) Genetic and epigenetic mechanisms in metal carcinogenesis and cocarcinogenesis: nickel, arsenic, and chromium. Chem Res Toxicol 21(1):28–44. doi:10.1021/tx700198a

    PubMed Central  PubMed  Google Scholar 

  • Schafer FQ, Buettner GR (2001) Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med 30(11):1191–1212

    CAS  PubMed  Google Scholar 

  • Schulz GE, Schirmer RH, Sachsenheimer W, Pai EF (1978) The structure of the flavoenzyme glutathione reductase. Nature 273(5658):120–124. doi:10.1038/273120a0

    CAS  PubMed  Google Scholar 

  • Sedoris KC, Thomas SD, Miller DM (2010) Hypoxia induces differential translation of enolase/MBP-1. BMC Cancer 10:157. doi:10.1186/1471-2407-10-157

    PubMed Central  PubMed  Google Scholar 

  • Semenza GL (2003) Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3(10):721–732. doi:10.1038/nrc1187

    CAS  PubMed  Google Scholar 

  • Semenza GL (2009) Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin Cancer Biol 19(1):12–16. doi:10.1016/j.semcancer.2008.11.009

    CAS  PubMed  Google Scholar 

  • Semenza GL (2010) HIF-1: upstream and downstream of cancer metabolism. Curr Opin Genet Dev 20(1):51–56. doi:10.1016/j.gde.2009.10.009

    CAS  PubMed Central  PubMed  Google Scholar 

  • Shi X, Ding M, Ye J, Wang S, Leonard SS, Zang L, Castranova V, Vallyathan V, Chiu A, Dalal N, Liu K (1999) Cr(IV) causes activation of nuclear transcription factor-kappa B, DNA strand breaks and dG hydroxylation via free radical reactions. J Inorg Biochem 75(1):37–44. doi:10.1016/S0162-0134(99)00030-6

    CAS  PubMed  Google Scholar 

  • Shim H, Dolde C, Lewis BC, Wu CS, Dang G, Jungmann RA, Dalla-Favera R, Dang CV (1997) c-Myc transactivation of LDH-A: implications for tumor metabolism and growth. Proc Natl Acad Sci USA 94(13):6658–6663

    CAS  PubMed Central  PubMed  Google Scholar 

  • Shumilla JA, Wetterhahn KE, Barchowsky A (1998) Inhibition of NF-kappa B binding to DNA by chromium, cadmium, mercury, zinc, and arsenite in vitro: evidence of a thiol mechanism. Arch Biochem Biophys 349(2):356–362. doi:10.1006/abbi.1997.0470

    CAS  PubMed  Google Scholar 

  • Shumilla JA, Broderick RJ, Wang Y, Barchowsky A (1999) Chromium(VI) inhibits the transcriptional activity of nuclear factor-kappaB by decreasing the interaction of p65 with cAMP-responsive element-binding protein-binding protein. J Biol Chem 274(51):36207–36212. doi:10.1074/jbc.274.51.36207

    CAS  PubMed  Google Scholar 

  • Skrede S, Bremer J, Eldjarn L (1965) The effect of disulphides on mitochondrial oxidations. Biochem J 95:838–846

    CAS  PubMed Central  PubMed  Google Scholar 

  • Slade PG, Hailer MK, Martin BD, Sugden KD (2005) Guanine-specific oxidation of double-stranded DNA by Cr(VI) and ascorbic acid forms spiroiminodihydantoin and 8-oxo-2′-deoxyguanosine. Chem Res Toxicol 18(7):1140–1149. doi:10.1021/tx050033y

    CAS  PubMed Central  PubMed  Google Scholar 

  • Stickland LH (1949) The activation of phosphoglucomutase by metal ions. Biochem J 44(2):190–197

    CAS  PubMed Central  PubMed  Google Scholar 

  • Sugiyama M, Tsuzuki K, Haramaki N (1993) Influence of o-phenanthroline on DNA single-strand breaks, alkali-labile sites, glutathione reductase, and formation of chromium(V) in Chinese hamster V-79 cells treated with sodium chromate (VI). Arch Biochem Biophys 305(2):261–266. doi:10.1006/abbi.1993.1420

    CAS  PubMed  Google Scholar 

  • Sumner ER, Shanmuganathan A, Sideri TC, Willetts SA, Houghton JE, Avery SV (2005) Oxidative protein damage causes chromium toxicity in yeast. Microbiology 151(Pt 6):1939–1948. doi:10.1099/mic.0.27945-0

    CAS  PubMed  Google Scholar 

  • Sun H, Zhou X, Chen H, Li Q, Costa M (2009) Modulation of histone methylation and MLH1 gene silencing by hexavalent chromium. Toxicol Appl Pharmacol 237(3):258–266. doi:10.1016/j.taap.2009.04.008

    CAS  PubMed Central  PubMed  Google Scholar 

  • Sun H, Clancy HA, Kluz T, Zavadil J, Costa M (2011) Comparison of gene expression profiles in chromate transformed BEAS-2B cells. PLoS One 6(3):e17982. doi:10.1371/journal.pone.0017982

    CAS  PubMed Central  PubMed  Google Scholar 

  • Tajima H, Yoshida T, Ohnuma A, Fukuyama T, Hayashi K, Yamaguchi S, Ohtsuka R, Sasaki J, Tomita M, Kojima S, Takahashi N, Kashimoto Y, Kuwahara M, Takeda M, Kosaka T, Nakashima N, Harada T (2010) Pulmonary injury and antioxidant response in mice exposed to arsenate and hexavalent chromium and their combination. Toxicology 267(1–3):118–124. doi:10.1016/j.tox.2009.10.032

    CAS  PubMed  Google Scholar 

  • Takahashi Y, Kondo K, Hirose T, Nakagawa H, Tsuyuguchi M, Hashimoto M, Sano T, Ochiai A, Monden Y (2005) Microsatellite instability and protein expression of the DNA mismatch repair gene, hMLH1, of lung cancer in chromate-exposed workers. Mol Carcinog 42(3):150–158. doi:10.1002/mc.20073

    CAS  PubMed  Google Scholar 

  • Tampo Y, Kotamraju S, Chitambar CR, Kalivendi SV, Keszler A, Joseph J, Kalyanaraman B (2003) Oxidative stress-induced iron signaling is responsible for peroxide-dependent oxidation of dichlorodihydrofluorescein in endothelial cells: role of transferrin receptor-dependent iron uptake in apoptosis. Circ Res 92(1):56–63

    CAS  PubMed  Google Scholar 

  • Thompson CB (2011) Rethinking the regulation of cellular metabolism. Cold Spring Harb Symp Quant Biol 76:23–29. doi:10.1101/sqb.2012.76.010496

    CAS  PubMed  Google Scholar 

  • Tsao DA, Tseng WC, Chang HR (2011) The expression of RKIP, RhoGDI, galectin, c-Myc and p53 in gastrointestinal system of Cr(VI)-exposed rats. J Appl Toxicol 31(8):730–740. doi:10.1002/jat.1621

    CAS  PubMed  Google Scholar 

  • Ueno S, Kashimoto T, Susa N, Furukawa Y, Ishii M, Yokoi K, Yasuno M, Sasaki YF, Ueda J, Nishimura Y, Sugiyama M (2001) Detection of dichromate (VI)-induced DNA strand breaks and formation of paramagnetic chromium in multiple mouse organs. Toxicol Appl Pharmacol 170(1):56–62. doi:10.1006/taap.2000.9081

    CAS  PubMed  Google Scholar 

  • Urbano AM, Rodrigues CFD, Alpoim MC (2008) Hexavalent chromium exposure, genomic instability and lung cancer. Gene Ther Mol Biol 12B:219–238

    Google Scholar 

  • Urbano AM, Ferreira LMR, Cerveira JF, Rodrigues CF, Alpoim MC (2011) DNA damage, DNA repair and human health. Repair and misrepair in cancer and in cancer therapy. InTech, Rijeka. doi:10.5772/23190

    Google Scholar 

  • Urbano AM, Ferreira LMR, Alpoim MC (2012) Molecular and cellular mechanisms of hexavalent chromium-induced lung cancer: an updated perspective. Curr Drug Metab 13(3):284–305

    CAS  PubMed  Google Scholar 

  • Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M (2006) Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact 160(1):1–40. doi:10.1016/j.cbi.2005.12.009

    CAS  PubMed  Google Scholar 

  • Vaupel P, Harrison L (2004) Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist 9(Suppl 5):4–9. doi:10.1634/theoncologist.9-90005-4

    PubMed  Google Scholar 

  • Vogelstein B, Kinzler KW (2004) Cancer genes and the pathways they control. Nat Med 10(8):789–799. doi:10.1038/nm1087

    CAS  PubMed  Google Scholar 

  • Wallace DC (2005) A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet 39:359–407. doi:10.1146/annurev.genet.39.110304.095751

    CAS  PubMed Central  PubMed  Google Scholar 

  • Wang S, Shi X (2001) Mechanisms of Cr(VI)-induced p53 activation: the role of phosphorylation, mdm2 and ERK. Carcinogenesis 22(5):757–762

    CAS  PubMed  Google Scholar 

  • Wang S, Chen F, Zhang Z, Jiang BH, Jia L, Shi X (2004) NF-kappaB prevents cells from undergoing Cr(VI)-induced apoptosis. Mol Cell Biochem 255(1–2):129–137

    CAS  PubMed  Google Scholar 

  • Wang XF, Xing ML, Shen Y, Zhu X, Xu LH (2006) Oral administration of Cr(VI) induced oxidative stress, DNA damage and apoptotic cell death in mice. Toxicology 228(1):16–23. doi:10.1016/j.tox.2006.08.005

    CAS  PubMed  Google Scholar 

  • Warburg O (1930) The metabolism of tumors. Constable & Co. Ltd., London

    Google Scholar 

  • Wellen KE, Thompson CB (2012) A two-way street: reciprocal regulation of metabolism and signalling. Nat Rev Mol Cell Biol 13(4):270–276. doi:10.1038/nrm3305

    CAS  PubMed  Google Scholar 

  • Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB (2009) ATP-citrate lyase links cellular metabolism to histone acetylation. Science 324(5930):1076–1080. doi:10.1126/science.1164097

    CAS  PubMed Central  PubMed  Google Scholar 

  • Wiegand HJ, Ottenwalder H, Bolt HM (1986) Disposition of a soluble chromate in the isolated perfused rat liver. Xenobiotica 16(9):839–844. doi:10.3109/00498258609038965

    CAS  PubMed  Google Scholar 

  • Williams AC, Collard TJ, Paraskeva C (1999) An acidic environment leads to p53 dependent induction of apoptosis in human adenoma and carcinoma cell lines: implications for clonal selection during colorectal carcinogenesis. Oncogene 18(21):3199–3204. doi:10.1038/sj.onc.1202660

    CAS  PubMed  Google Scholar 

  • Winterbourn CC (2008) Reconciling the chemistry and biology of reactive oxygen species. Nat Chem Biol 4(5):278–286. doi:10.1038/nchembio.85

    CAS  PubMed  Google Scholar 

  • Xiao F, Feng X, Zeng M, Guan L, Hu Q, Zhong C (2012a) Hexavalent chromium induces energy metabolism disturbance and p53-dependent cell cycle arrest via reactive oxygen species in L-02 hepatocytes. Mol Cell Biochem. doi:10.1007/s11010-012-1423-7

    Google Scholar 

  • Xiao F, Li Y, Dai L, Deng Y, Zou Y, Li P, Yang Y, Zhong C (2012b) Hexavalent chromium targets mitochondrial respiratory chain complex I to induce reactive oxygen species-dependent caspase-3 activation in L-02 hepatocytes. Int J Mol Med 30(3):629–635. doi:10.3892/ijmm.2012.1031

    CAS  PubMed  Google Scholar 

  • Xie Y, Zhong C, Zeng M, Guan L, Luo L (2013) Effect of hexavalent chromium on electron leakage of respiratory chain in mitochondria isolated from rat liver. Cell Physiol Biochem 31(2–3):473–485. doi:10.1159/000350062

    CAS  PubMed  Google Scholar 

  • Xu J, Bubley GJ, Detrick B, Blankenship LJ, Patierno SR (1996) Chromium(VI) treatment of normal human lung cells results in guanine-specific DNA polymerase arrest, DNA–DNA cross-links and S-phase blockade of cell cycle. Carcinogenesis 17(7):1511–1517. doi:10.1093/carcin/17.7.1511

    CAS  PubMed  Google Scholar 

  • Yamamoto M, Taguchi Y, Ito-Kureha T, Semba K, Yamaguchi N, Inoue J (2013) NF-kappaB non-cell-autonomously regulates cancer stem cell populations in the basal-like breast cancer subtype. Nat Commun 4:2299. doi:10.1038/ncomms3299

    PubMed  Google Scholar 

  • Ye J, Shi X (2001) Gene expression profile in response to chromium-induced cell stress in A549 cells. Mol Cell Biochem 222(1–2):189–197. doi:10.1007/978-1-4615-0793-2_22

    CAS  PubMed  Google Scholar 

  • Ye J, Zhang X, Young HA, Mao Y, Shi X (1995) Chromium(VI)-induced nuclear factor-kappa B activation in intact cells via free radical reactions. Carcinogenesis 16(10):2401–2405. doi:10.1093/carcin/16.10.2401

    CAS  PubMed  Google Scholar 

  • Yuan Y, Ming Z, Gong-Hua H, Lan G, Lu D, Peng L, Feng J, Cai-Gao Z (2012) Cr(VI) induces the decrease of ATP level and the increase of apoptosis rate mediated by ROS or VDAC1 in L-02 hepatocytes. Environ Toxicol Pharmacol 34(2):579–587. doi:10.1016/j.etap.2012.06.016

    CAS  PubMed  Google Scholar 

  • Yun JW, Kim SK, Kim H (2011) Prolonged protein turnover of glyceraldehyde-3-phosphate dehydrogenase by phospholipase C-gamma 1 is critical for anchorage-independent growth and ATP synthesis in transformed cells. Cancer Invest 29(2):93–101. doi:10.3109/07357907.2010.535062

    CAS  PubMed  Google Scholar 

  • Zhang H, Bosch-Marce M, Shimoda LA, Tan YS, Baek JH, Wesley JB, Gonzalez FJ, Semenza GL (2008) Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J Biol Chem 283(16):10892–10903. doi:10.1074/jbc.M800102200

    CAS  PubMed Central  PubMed  Google Scholar 

  • Zhitkovich A (2005) Importance of chromium-DNA adducts in mutagenicity and toxicity of chromium(VI). Chem Res Toxicol 18(1):3–11. doi:10.1021/tx049774+

    CAS  PubMed  Google Scholar 

  • Zornig M, Evan GI (1996) Cell cycle: on target with Myc. Curr Biol 6(12):1553–1556. doi:10.1016/S0960-9822(02)70769-0

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

Investigations from the authors’ laboratories described here were supported by Centro de Investigação em Meio Ambiente, Genética e Oncobiologia (CIMAGO), Portugal (Grants 26/07, to AMU, and 16/06, to MCA) and by Fundação para a Ciência e a Tecnologia, Portugal (Grant PEst-OE/QUI/UI0070/2011, to Unidade de Química Física Molecular). The authors apologize to all colleagues whose relevant work could not be mentioned and/or cited owing to space limitations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to A. M. Urbano.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Abreu, P.L., Ferreira, L.M.R., Alpoim, M.C. et al. Impact of hexavalent chromium on mammalian cell bioenergetics: phenotypic changes, molecular basis and potential relevance to chromate-induced lung cancer. Biometals 27, 409–443 (2014). https://doi.org/10.1007/s10534-014-9726-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10534-014-9726-7

Keywords

Navigation