Since the inception of liquid chromatography (LC) more than 100 years ago this separation technique has been developed into a powerful analytical tool that is frequently applied in life science research. To this end, unique insights into the interaction of metal species (throughout this manuscript “metal species” refers to “toxic metals, metalloid compounds, and metal-based drugs” and “toxic metals” to “toxic metals and metalloid compounds”) with endogenous ligands can be obtained by using LC approaches that involve their hyphenation with inductively coupled plasma-based element specific detectors. This review aims to provide a synopsis of the different LC approaches which may be employed to advance our understanding of these interactions either in a “bottom-up” or a “top-down” manner. In the “bottom-up” LC-configuration, endogenous ligands are introduced into a physiologically relevant mobile phase buffer, and the metal species of interest is injected. Subsequent “interrogation” of the on-column formed complex(es) by employing a suitable separation mechanism (e.g., size exclusion chromatography or reversed-phase LC) while changing the ligand concentration(s), the column temperature or the pH can provide valuable insight into the formation of complexes under near physiological conditions. This approach allows to establish the relative stability and hydrophobicity of metal-ligand complexes as well as the dynamic coordination of a metal species (injected) to two ligands (dissolved in the mobile phase). Conversely, the “top-down” analysis of a biological fluid (e.g., blood plasma) by LC (e.g., using size exclusion chromatography) can be used to determine the size distribution of endogenous metalloproteins which are collectively referred to as the “metalloproteome”. This approach can provide unique insight into the metabolism and the plasma protein binding of metal species, and can simultaneously visualize the dose-dependent perturbation of the metalloproteome by a particular metal species. The concerted application of these LC approaches is destined to provide new insight into biochemical processes which represent an important starting point to advance human health in the 21st century. 1. Introduction Based on his work on plant pigments, M.S. Tswett serendipitously invented adsorption chromatography as a universal analytical method to separate chemical compounds in 1906 [1]. After an interregnum of ~40 years, the discovery of liquid-liquid partition chromatography by A.J.P. Martin and R.L.M. Synge breathed new life into this ground-breaking separation
References
[1]
M. Tswett, “Physical chemical studies on chlorophyll adsorptions,” Berichte der Deutschen botanischen Gesellschaft, vol. 24, pp. 316–326, 1906.
[2]
A. J. P. Martin and R. L. M. Synge, “A new form of chromatogram employing two liquid phases,” Biochemical Journal, vol. 35, pp. 1358–1368, 1941.
[3]
L. S. Ettre and K. I. Sakodynskii, “M.S. Tswett and the discovery of chromatography. I: early work (1899–1903),” Chromatographia, vol. 35, no. 3-4, pp. 223–231, 1993.
[4]
A. Zotou, “An overview of recent advances in HPLC instrumentation,” Central European Journal of Chemistry, vol. 10, no. 3, pp. 554–569, 2012.
[5]
K. K. Unger and A. I. Liapis, “Adsorbents and columns in analytical high-performance liquid chromatography: a perspective with regard to development and understanding,” Journal of Separation Science, vol. 35, no. 10-11, pp. 1201–1212, 2012.
[6]
D. Ishii, K. Asai, K. Hibi, T. Jonokuchi, and M. Nagaya, “A study of micro-high-performance liquid chromatography. I. Development of technique for miniaturization of high-performance liquid chromatography,” Journal of Chromatography, vol. 144, no. 2, pp. 157–168, 1977.
[7]
C. G. Horvath, B. A. Preiss, and S. R. Lipsky, “Fast liquid chromatography: an investigation of operating parameters and the separation of nucleotides on pellicular ion exchangers,” Analytical Chemistry, vol. 39, no. 12, pp. 1422–1428, 1967.
[8]
P. D. McDonald and D. Patrick, “Fifty years of innovation in analysis and purification,” Chemical Heritage, vol. 26, no. 2, pp. 32–37, 2008.
[9]
M. E. Swartz, “UPLC: an introduction and review,” Journal of Liquid Chromatography & Related Technologies, vol. 28, no. 7-8, pp. 1253–1263, 2005.
[10]
L. Anderson and C. L. Hunter, “Quantitative mass spectrometric multiple reaction monitoring assays for major plasma proteins,” Molecular & Cellular Proteomics, vol. 5, no. 4, pp. 573–588, 2006.
[11]
D. J. States, G. S. Omenn, T. W. Blackwell et al., “Challenges in deriving high-confidence protein identifications from data gathered by a HUPO plasma proteome collaborative study,” Nature Biotechnology, vol. 24, no. 3, pp. 333–338, 2006.
[12]
S. P. Gygi, B. Rist, T. J. Griffin, et al., “Proteome analysis of low-abundance proteins using multidimensional chromatography and isotope-coded affinity tags,” Journal of Proteome Research, vol. 1, no. 1, pp. 47–54, 2002.
[13]
D. S. Wishart, “Advances in metabolite identification,” Bioanalysis, vol. 3, no. 15, pp. 1769–1782, 2011.
[14]
K. A. Francesconi and F. Pannier, “Selenium metabolites in urine: a critical overview of past work and current status,” Clinical Chemistry, vol. 50, no. 12, pp. 2240–2253, 2004.
[15]
R. F. Service, “Proteomics: ponders prime time,” Science, vol. 321, no. 5897, pp. 1758–1761, 2008.
[16]
J. K. Nicholson and J. C. Lindon, “Systems biology: metabonomics,” Nature, vol. 455, no. 7216, pp. 1054–1056, 2008.
[17]
S. C. Booth, M. L. Workentine, A. M. Weljie, and R. J. Turner, “Metabolomics and its application to studying metal toxicity,” Metallomics, vol. 3, pp. 1142–1152, 2011.
[18]
J. van der Greef, P. Stroobant, and R. van der Heijden, “The role of analytical sciences in medical systems biology,” Current Opinion in Chemical Biology, vol. 8, no. 5, pp. 559–565, 2004.
[19]
L. Hood, “A systems approach to medicine will transform healthcare,” in Physical Biology from Atoms to Medicine, A. H. Zewail, Ed., pp. 337–366, Imperial College Press, London, UK, 2008.
[20]
G. Xindu and W. Lili, “Liquid chromatography of recombinant proteins and protein drugs,” Journal of Chromatography B, vol. 866, no. 1-2, pp. 133–153, 2008.
[21]
R. J. McGorrin, “One hundred years of progress in food analysis,” Journal of Agricultural and Food Chemistry, vol. 57, no. 18, pp. 8076–8088, 2009.
[22]
L. S. Jackson, “Chemical food safety issues in the United States: past, present, and future,” Journal of Agricultural and Food Chemistry, vol. 57, no. 18, pp. 8161–8170, 2009.
[23]
S. J. Hill, A. Fisher, and M. Foulkes, “Basic concepts and instrumentation for plasma spectrometry,” in Inductively Coupled Plasma Spectrometry and Its Applications, S. J. Hill, Ed., pp. 61–97, Blackwell Publishing, Ames, Iowa, USA, 2007.
[24]
F. van Heuveln, H. Meijering, and J. Wieling, “Inductively coupled plasma-MS in drug development: bioanalytical aspects and applications,” Bioanalysis, vol. 4, no. 15, pp. 1933–1965, 2012.
[25]
S. Mounicou, J. Szpunar, and R. Lobinski, “Metallomics: the concept and methodology,” Chemical Society Reviews, vol. 38, no. 4, pp. 1119–1138, 2009.
[26]
J. L. Gómez-Ariza, E. Z. Jahromi, M. González-Fernández, T. García-Barrera, and J. Gailer, “Liquid chromatography-inductively coupled plasma-based metallomic approaches to probe health-relevant interactions between xenobiotics and mammalian organisms,” Metallomics, vol. 3, no. 6, pp. 566–577, 2011.
[27]
J. P. Barnett, D. J. Scanlan, and C. A. Blindauer, “Protein fractionation and detection for metalloproteomics: challenges and approaches,” Analytical and Bioanalytical Chemistry, vol. 402, no. 10, pp. 3311–3322, 2012.
[28]
J. Szpunar, “Advances in analytical methodology for bioinorganic speciation analysis: metallomics, metalloproteomics and heteroatom-tagged proteomics and metabolomics,” Analyst, vol. 130, no. 4, pp. 442–465, 2005.
[29]
R. Dufault, B. LeBlanc, R. Schnoll, et al., “Mercury from chlor-alkali plants: measured concentrations in food product sugar,” Environmental Health, vol. 8, article 2, 2009.
[30]
D. Gilbert-Diamond, K. L. Cottingham, J. F. Gruber et al., “Rice consumption contributes to arsenic exposure in US women,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 51, pp. 20656–20660, 2011.
[31]
H. H. Harris, I. J. Pickering, and G. N. George, “The chemical form of mercury in fish,” Science, vol. 301, no. 5637, p. 1203, 2003.
[32]
J. Gailer, “Arsenic-selenium and mercury-selenium bonds in biology,” Coordination Chemistry Reviews, vol. 251, no. 1-2, pp. 234–254, 2007.
[33]
A. Bhatnagar, “Environmental cardiology: studying mechanistic links between pollution and heart disease,” Circulation Research, vol. 99, no. 7, pp. 692–705, 2006.
[34]
J. Gailer, “Probing the bioinorganic chemistry of toxic metals in the mammalian bloodstream to advance human health,” Journal of Inorganic Biochemistry, vol. 108, pp. 128–132, 2012.
[35]
E. Z. Jahromi and J. Gailer, “Probing bioinorganic chemistry processes in the bloodstream to gain new insights into the origin of human diseases,” Dalton Transactions, vol. 39, no. 2, pp. 329–336, 2010.
[36]
E. Z. Jahromi and J. Gailer, “In vitro assessment of chelating agents with regard to their abstraction efficiency of Cd2+ bound to plasma proteins,” Metallomics, vol. 4, no. 9, pp. 995–1003, 2012.
[37]
M. Sooriyaarachchi, A. Narendran, and J. Gailer, “The effect of sodium thiosulfate on the metabolism of cis-platin in human plasma in vitro,” Metallomics, vol. 4, no. 9, pp. 960–967, 2012.
[38]
M. Montes-Bayón, K. DeNicola, and J. A. Caruso, “Liquid chromatography-inductively coupled plasma mass spectrometry,” Journal of Chromatography A, vol. 1000, no. 1-2, pp. 457–476, 2003.
[39]
J. López-Barea and J. L. Gómez-Ariza, “Environmental proteomics and metallomics,” Proteomics, vol. 6, supplement 1, pp. S51–S62, 2006.
[40]
M. A. O. da Silva, A. Sussulini, and M. A. Z. Arruda, “Metalloproteomics as an interdisciplinary area involving proteins and metals,” Expert Review of Proteomics, vol. 7, no. 3, pp. 387–400, 2010.
[41]
W. Shi and M. R. Chance, “Metalloproteomics: forward and reverse approaches in metalloprotein structural and functional characterization,” Current Opinion in Chemical Biology, vol. 15, no. 1, pp. 144–148, 2011.
[42]
W. Mertz, “The essential trace elements,” Science, vol. 213, no. 4514, pp. 1332–1338, 1981.
[43]
N. C. Andrews and P. J. Schmidt, “Iron homeostasis,” Annual Reviews of Physiology, vol. 69, pp. 69–85, 2007.
[44]
W. Maret and Y. Li, “Coordination dynamics of zinc in proteins,” Chemical Reviews, vol. 109, no. 10, pp. 4682–4707, 2009.
[45]
J. T. Rubino and K. J. Franz, “Coordination chemistry of copper proteins: how nature handles a toxic cargo for essential function,” Journal of Inorganic Biochemistry, vol. 107, no. 1, pp. 129–143, 2012.
[46]
S. M. Yannone, S. Hartung, A. L. Menon, M. W. W. Adams, and J. A. Tainer, “Metals in biology: defining metalloproteomes,” Current Opinion in Biotechnology, vol. 23, no. 1, pp. 89–95, 2012.
[47]
S. L. Sensi, P. Paoletti, A. I. Bush, and I. Sekler, “Zinc in the physiology and pathology of the CNS,” Nature Reviews Neuroscience, vol. 10, no. 11, pp. 780–791, 2009.
[48]
K. J. Waldron, J. C. Rutherford, D. Ford, and N. J. Robinson, “Metalloproteins and metal sensing,” Nature, vol. 460, no. 7257, pp. 823–830, 2009.
[49]
W. Maret, “Metalloproteomics, metalloproteomes, and the annotation of metalloproteins,” Metallomics, vol. 2, no. 2, pp. 117–125, 2010.
[50]
J. K. Nicholson and I. D. Wilson, “Understanding 'global' systems biology: metabonomics and the continuum of metabolism,” Nature Reviews Drug Discovery, vol. 2, no. 8, pp. 668–676, 2003.
[51]
J. L. Peters, T. S. Perlstein, M. J. Perry, E. McNeely, and J. Weuve, “Cadmium exposure in association with history of stroke and heart failure,” Environmental Research, vol. 110, no. 2, pp. 199–206, 2010.
[52]
O. Andersen, “Chemical and biological considerations in the treatment of metal intoxications by chelating agents,” Mini-Reviews in Medicinal Chemistry, vol. 4, no. 1, pp. 11–21, 2004.
[53]
M. Sooriyaarachchi, A. Narendran, and J. Gailer, “Comparative hydrolysis and plasma protein binding of cis-platin and carboplatin in human plasma in vitro,” Metallomics, vol. 3, no. 1, pp. 49–55, 2011.
[54]
T. W. Hambley, “Developing new metal-based therapeutics: challenges and opportunities,” Dalton Transactions, no. 43, pp. 4929–4937, 2007.
[55]
J. Reedijk, “Why does cisplatin reach guanine-N7 with competing S-donor ligands available in the cell?” Chemical Reviews, vol. 99, no. 9, pp. 2499–2510, 1999.
[56]
B. H. Ali and M. S. Al Moundhri, “Agents ameliorating or augmenting the nephrotoxicity of cisplatin and other platinum compounds: a review of some recent research,” Food and Chemical Toxicology, vol. 44, no. 8, pp. 1173–1183, 2006.
[57]
K. C. M. Campbell, R. P. Meech, J. J. Klemens et al., “Prevention of noise- and drug-induced hearing loss with d-methionine,” Hearing Research, vol. 226, no. 1-2, pp. 92–103, 2007.
[58]
G. D. Zeevalk and W. J. Nicklas, “Mechanisms underlying initiation of excitotoxicity associated with metabolic inhibition,” Journal of Pharmacology and Experimental Therapeutics, vol. 257, no. 2, pp. 870–878, 1991.
[59]
S. Cestèle and W. A. Catterall, “Molecular mechanisms of neurotoxin action on voltage-gated sodium channels,” Biochimie, vol. 82, no. 9-10, pp. 883–892, 2000.
[60]
K. Cottingham, “Systems biology: a boon for analytical chemists?” Analytical Chemistry, vol. 77, no. 9, pp. 197A–200A, 2005.
[61]
J. Gailer, G. N. George, I. J. Pickering et al., “Structural basis of the antagonism between inorganic mercury and selenium in mammals,” Chemical Research in Toxicology, vol. 13, no. 11, pp. 1135–1142, 2000.
[62]
J. Gailer, G. N. George, I. J. Pickering et al., “A metabolic link between arsenite and selenite: the seleno-bis(S- glutathionyl) arsinium ion,” Journal of the American Chemical Society, vol. 122, no. 19, pp. 4637–4639, 2000.
[63]
J. Gailer, S. Madden, G. A. Buttigieg, M. B. Denton, and H. S. Younis, “Identification of [(GS)2AsSe]? in rabbit bile by size-exclusion chromatography and simultaneous multielement-specific detection by inductively coupled plasma atomic emission spectroscopy,” Applied Organometallic Chemistry, vol. 16, no. 2, pp. 72–75, 2002.
[64]
J. Gailer, L. Ruprecht, P. Reitmeir, B. Benker, and P. Schramel, “Mobilization of exogenous and endogenous selenium to bile after the intravenous administration of environmentally relevant doses of arsenite to rabbits,” Applied Organometallic Chemistry, vol. 18, no. 12, pp. 670–675, 2004.
[65]
J. Gailer, G. N. George, I. J. Pickering, R. C. Prince, H. S. Younis, and J. J. Winzerling, “Biliary excretion of [(GS)2AsSe]- after intravenous injection of rabbits with arsenite and selenate,” Chemical Research in Toxicology, vol. 15, no. 11, pp. 1466–1471, 2002.
[66]
P. Hunter, P. Robbins, and D. Noble, “The IUPS human physiome project,” Pflügers Archiv, vol. 445, no. 1, pp. 1–9, 2002.
[67]
L. Hood, J. R. Heath, M. E. Phelps, and B. Lin, “Systems biology and new technologies enable predictive and preventative medicine,” Science, vol. 306, no. 5696, pp. 640–643, 2004.
[68]
L. M. Gierasch and A. Gershenson, “Post-reductionist protein science, or putting Humpty Dumpty back together again,” Nature Chemical Biology, vol. 5, pp. 774–777, 2009.
[69]
D. Noble, The Music of Life, Biology Beyond Genes, Oxford University Press, New York, NY, USA, 2006.
[70]
F. R. Luo, S. D. Wyrick, and S. G. Chaney, “Biotransformations of oxaliplatin in rat blood in vitro,” Journal of Biochemical and Molecular Toxicology, vol. 13, no. 3-4, pp. 159–169, 1999.
[71]
C. Jumarie, C. Fortin, M. Houde, P. G. C. Campbell, and F. Denizeau, “Cadmium uptake by Caco-2 cells: effects of Cd complexation by chloride, glutathione, and phytochelatins,” Toxicology and Applied Pharmacology, vol. 170, no. 1, pp. 29–38, 2001.
[72]
R. K. Zalups, “Molecular interactions with mercury in the kidney,” Pharmacological Reviews, vol. 52, no. 1, pp. 113–143, 2000.
[73]
R. K. Zalups and S. Ahmad, “Molecular handling of cadmium in transporting epithelia,” Toxicology and Applied Pharmacology, vol. 186, no. 3, pp. 163–188, 2003.
[74]
J. L. Webb, “Arsenicals,” in Enzyme and Metabolic Inhibitors, J. L. Webb, Ed., vol. 3, pp. 595–793, Academic Press, London, UK, 1966.
[75]
S. A. Manley and J. Gailer, “Analysis of the plasma metalloproteome by SEC-ICP-AES: bridging proteomics and metabolomics,” Expert Review of Proteomics, vol. 6, no. 3, pp. 251–265, 2009.
[76]
G. Chillemi, G. Mancini, N. Sanna et al., “Evidence for sevenfold coordination in the first solvation shell of Hg(II) aqua ion,” Journal of the American Chemical Society, vol. 129, no. 17, pp. 5430–5436, 2007.
[77]
G. M. Whitesides, P. W. Snyder, D. T. Moustakas, and K. A. Mirica, “Designing ligands to bind tightly to proteins,” in Physical Biology, from Atoms to Medicine, A. H. Zewail, Ed., pp. 189–215, Imperial College Press, London, UK, 2008.
[78]
J. P. K. Rooney, “The role of thiols, dithiols, nutritional factors and interacting ligands in the toxicology of mercury,” Toxicology, vol. 234, no. 3, pp. 145–156, 2007.
[79]
A. Casini and J. Reedijk, “Interactions of anticancer Pt compounds with proteins: an overlooked topic in medicinal inorganic chemistry?” Chemical Science, vol. 3, no. 11, pp. 3135–3144, 2012.
[80]
A. R. Timerbaev, C. G. Hartinger, S. S. Aleksenko, and B. K. Keppler, “Interactions of antitumor metallodrugs with serum proteins: advances in characterization using modern analytical methodology,” Chemical Reviews, vol. 106, no. 6, pp. 2224–2248, 2006.
[81]
M. Knipp, “Metallothioneins and platinum(II) anti-tumor compounds,” Current Medicinal Chemistry, vol. 16, pp. 522–537, 2009.
[82]
J. Gailer, “Reactive selenium metabolites as targets of toxic metals/metalloids in mammals: a molecular toxicological perspective,” Applied Organometallic Chemistry, vol. 16, no. 12, pp. 701–707, 2002.
[83]
W. Hu, Q. Luo, K. Wu et al., “The anticancer drug cisplatin can cross-link the interdomain zinc site on human albumin,” Chemical Communications, vol. 47, no. 21, pp. 6006–6008, 2011.
[84]
J. Gailer, “Chronic toxicity of in mammals: the role of (GS)2AsSe-,” Biochimie, vol. 91, no. 10, pp. 1268–1272, 2009.
[85]
M. T. Le, J. Gailer, and E. J. Prenner, “Hg2+ and Cd2+ interact differently with biomimetic erythrocyte membranes,” BioMetals, vol. 22, no. 2, pp. 261–274, 2009.
[86]
N. Ballatori, “Transport of toxic metals by molecular mimicry,” Environmental Health Perspectives, vol. 110, no. 5, pp. 689–694, 2002.
[87]
R. H. Holm, P. Kennepohl, and E. I. Solomon, “Structural and functional aspects of metal sites in biology,” Chemical Reviews, vol. 96, no. 7, pp. 2239–2314, 1996.
[88]
B. P. Esposito and R. Najjar, “Interactions of antitumoral platinum-group metallodrugs with albumin,” Coordination Chemistry Reviews, vol. 232, no. 1-2, pp. 137–149, 2002.
[89]
N. Ballatori and A. T. Truong, “Mechanisms of hepatic methylmercury uptake,” Journal of Toxicology and Environmental Health, vol. 46, no. 3, pp. 343–353, 1995.
[90]
N. Shimojo, Y. Kumagai, and J. Nagafune, “Difference between kidney and liver in decreased manganese superoxide dismutase activity caused by exposure of mice to mercuric chloride,” Archives of Toxicology, vol. 76, no. 7, pp. 383–387, 2002.
[91]
S. A. Manley, G. N. George, I. J. Pickering et al., “The seleno bis(S-glutathionyl) arsinium ion is assembled in erythrocyte lysate,” Chemical Research in Toxicology, vol. 19, no. 4, pp. 601–607, 2006.
[92]
A. Meister, “Glutathione metabolism and its selective modification,” Journal of Biological Chemistry, vol. 263, no. 33, pp. 17205–17208, 1988.
[93]
J. M. Mates, J. A. Segura, and F. J. Alonso, “Roles of dioxins and heavy metals in cancer and neurological diseases using ROS-mediated mechanisms,” Free Radical Biology and Medicine, vol. 49, no. 9, pp. 1328–1341, 2010.
[94]
T. J. Hagele, J. N. Mazerik, A. Gregory et al., “Mercury activates vascular endothelial cell phospholipase D through thiols and oxidative stress,” International Journal of Toxicology, vol. 26, no. 1, pp. 57–69, 2007.
[95]
S. Bhattacharya, S. Bose, B. Mukhopadhyay et al., “Specific binding of inorganic mercury to Na- K-ATPase in rat liver plasma membrane and signal transduction,” BioMetals, vol. 10, no. 3, pp. 157–162, 1997.
[96]
D. R. Green and G. Kroemer, “The pathophysiology of mitochondrial cell death,” Science, vol. 305, no. 5684, pp. 626–629, 2004.
[97]
S. Orrenius and B. Zhivotovsky, “The future of toxicology—does it matter how cells die?” Chemical Research in Toxicology, vol. 19, pp. 729–733, 2006.
[98]
J. D. Robertson and S. Orrenius, “Molecular mechanisms of apoptosis Induced by cytotoxic chemicals,” Critical Reviews in Toxicology, vol. 30, no. 5, pp. 609–627, 2000.
[99]
U. Landegren, J. V?nelid, M. Hammond et al., “Opportunities for sensitive plasma proteome analysis,” Analytical Chemistry, vol. 84, no. 4, pp. 1824–1830, 2012.
[100]
S. A. Manley, S. Byrns, A. W. Lyon, P. Brown, and J. Gailer, “Simultaneous Cu-, Fe-, and Zn-specific detection of metalloproteins contained in rabbit plasma by size-exclusion chromatography-inductively coupled plasma atomic emission spectroscopy,” JBIC Journal of Biological Inorganic Chemistry, vol. 14, no. 1, pp. 61–74, 2009.
[101]
P. J. Sadler and J. H. Viles, “1H and 113Cd NMR investigations of Cd2+ and Zn2+ binding sites on serum albumin: competition with Ca2+, Ni2+, Cu2+, and Zn2+,” Inorganic Chemistry, vol. 35, no. 15, pp. 4490–4496, 1996.
[102]
D. C. Carter and J. X. Ho, “Structure of serum albumin,” Advances in Protein Chemistry, vol. 45, pp. 153–203, 1994.
[103]
V. Sahni, D. Choudhury, and Z. Ahmed, “Chemotherapy-associated renal dysfunction,” Nature Reviews Nephrology, vol. 5, pp. 450–462, 2009.
[104]
D. Wang and S. J. Lippard, “Cellular processing of platinum anticancer drugs,” Nature Reviews Drug Discovery, vol. 4, pp. 307–320, 2005.
[105]
F. Caurant, M. Navarro, and J. C. Amiard, “Mercury in pilot whales: possible limits to the detoxification process,” Science of the Total Environment, vol. 186, no. 1-2, pp. 95–104, 1996.
[106]
M. Korbas, J. L. O. 'Donoghue, G. E. Watson et al., “The chemical nature of mercury in human brain following poisoning or environmental exposure,” ACS Chemical Neuroscience, vol. 1, no. 12, pp. 810–818, 2010.
[107]
J. P. Berry and P. Galle, “Selenium-arsenic interaction in renal cells: role of lysosomes. Electron microprobe study,” Journal of Submicroscopic Cytology and Pathology, vol. 26, no. 2, pp. 203–210, 1994.
[108]
S. Hann, G. Koellensperger, Z. Stefánka et al., “Application of HPLC-ICP-MS to speciation of cisplatin and its degradation products in water containing different chloride concentrations and in human urine,” Journal of Analytical Atomic Spectrometry, vol. 18, pp. 1391–1395, 2003.
[109]
Z. Gregus and C. D. Klaassen, “Disposition of metals in rats: a comparative study of fecal, urinary, and biliary excretion and tissue distribution of eighteen metals,” Toxicology and Applied Pharmacology, vol. 85, no. 1, pp. 24–38, 1986.
[110]
K. T. Suzuki, “Direct connection of high-speed liquid chromatograph (equipped with gel permeation column) to atomic absorption spectrophotometer for metalloprotein analysis: metallothionein,” Analytical Biochemistry, vol. 102, no. 1, pp. 31–34, 1980.
[111]
K. T. Suzuki, H. Sunaga, E. Kobayashi, and N. Shimojo, “Mercaptalbumin as a selective cadmium-binding protein in rat serum,” Toxicology and Applied Pharmacology, vol. 86, no. 3, pp. 466–473, 1986.
[112]
S. J. Berners-Price and P. J. Sadler, “Coordination chemistry of metallodrugs: insights into biological speciation from NMR spectroscopy,” Coordination Chemistry Reviews, vol. 151, pp. 1–40, 1996.
[113]
M. Groessl and P. J. Dyson, “Bioanalytical and biophysical techniques for the elucidation of the mode of action of metal-based drugs,” Current Topics in Medicinal Chemistry, vol. 11, no. 21, pp. 2632–2646, 2011.
[114]
V. Mah and F. Jalilehvand, “Cadmium(II) complex formation with glutathione,” JBIC Journal of Biological Inorganic Chemistry, vol. 15, no. 3, pp. 441–458, 2010.
[115]
R. Ortega, A. Carmona, I. Llorens, and P. L. Solari, “X-ray absorption spectroscopy of biological samples. A tutorial,” Journal of Analytical Atomic Spectrometry, vol. 27, pp. 2054–2065, 2012.
[116]
N. Cetinbas, M. I. Webb, J. A. Dubland, and C. J. Walsby, “Serum-protein interactions with anticancer Ru(III) complexes KP1019 and KP418 characterized by EPR,” JBIC Journal of Biological Inorganic Chemistry, vol. 15, no. 2, pp. 131–145, 2010.
[117]
Y. Kasherman, S. Sturup, and D. Gibson, “Is glutathione the major cellular target of cisplatin? A study of the interactions of cisplatin with cancer cell extracts,” Journal of Medicinal Chemistry, vol. 52, no. 14, pp. 4319–4328, 2009.
[118]
D. Gibson, “The mechanism of action of platinum anticancer agents—what do we really know about it?” Dalton Transactions, no. 48, pp. 10681–10689, 2009.
[119]
E. Z. Jahromi, W. White, Q. Wu, R. Yamdagni, and J. Gailer, “Remarkable effect of mobile phase buffer on the SEC-ICP-AES derived Cu, Fe and Zn-metalloproteome pattern of rabbit blood plasma,” Metallomics, vol. 2, no. 7, pp. 460–468, 2010.
[120]
C. C. Wu, P. H. Peng, Y. T. Chang et al., “Identification of potential serum markers for nasopharyngeal carcinoma from a xenografted mouse model using Cy-dye labeling combined with three-dimensional fractionation,” Proteomics, vol. 8, no. 17, pp. 3605–3620, 2008.
[121]
K. L. Pei, M. Sooriyaarachchi, D. A. Sherrell, G. N. George, and J. Gailer, “Probing the coordination behavior of Hg2+, CH3Hg+, and Cd2+ towards mixtures of two biological thiols by HPLC-ICP-AES,” Journal of Inorganic Biochemistry, vol. 105, no. 3, pp. 375–381, 2011.
[122]
E. K. M. Ueda, P. W. Gout, and L. Morganti, “Current and prospective applications of metal ion-protein binding,” Journal of Chromatography A, vol. 988, no. 1, pp. 1–23, 2003.
[123]
L. Andersson, E. Sulkowski, and J. Porath, “Immobilized metal ion affinity chromatography of serum albumins.,” Bioseparation, vol. 2, no. 1, pp. 15–22, 1991.
[124]
L. Gelunaite, V. Luk?a, O. Sud?iuviene, V. Bumelis, and H. Pesliakas, “Chelated mercury as a ligand in immobilized metal ion affinity chromatography of proteins,” Journal of Chromatography A, vol. 904, no. 2, pp. 131–143, 2000.
[125]
A. Meister and M. E. Anderson, “Glutathione,” Annual Review of Biochemistry, vol. 52, pp. 711–760, 1983.
[126]
N. Kaplowitz, T. Y. Aw, and M. Ookhtens, “The regulation of hepatic glutathione,” Annual Review of Pharmacology and Toxicology, vol. 25, pp. 715–744, 1985.
[127]
K. L. Haas and K. F. Franz, “Application of metal coordination chemistry to explore and manipulate cell biology,” Chemical Reviews, vol. 109, no. 10, pp. 4921–4960.
[128]
R. K. Singhal, M. E. Anderson, and A. Meister, “Glutathione, a first line of defense against cadmium toxicity,” FASEB Journal, vol. 1, no. 3, pp. 220–223, 1987.
[129]
A. Naganuma, M. E. Anderson, and A. Meister, “Cellular glutathione as a determinant of sensitivity to mercuric chloride toxicity: prevention of toxicity by giving glutathione monoester,” Biochemical Pharmacology, vol. 40, no. 4, pp. 693–697, 1990.
[130]
D. L. Rabenstein, “Metal complexes of glutathione and their biological significance,” in Glutathione: Chemical, Biochemical, and Medical Aspects, D. Dolphin, O. Avramovic, and R. Poulson, Eds., pp. 147–186, John Wiley & Sons, New York, NY, USA, 1989.
[131]
J. Gailer, G. N. George, I. J. Pickering, G. A. Buttigieg, M. B. Denton, and R. S. Glass, “Synthesis, X-ray absorption spectroscopy and purification of the seleno-bis (S-glutathionyl) arsinium anion from selenide, arsenite and glutathione,” Journal of Organometallic Chemistry, vol. 650, no. 1-2, pp. 108–113, 2002.
[132]
L. H. Lash and D. P. Jones, “Distribution of oxidized and reduced forms of glutathione and cysteine in rat plasma,” Archives of Biochemistry and Biophysics, vol. 240, no. 2, pp. 583–592, 1985.
[133]
B. Sebille, N. Thuaud, and J. P. Tillement, “Retention data methods for the determination of drug-protein binding parameters by high-performance liquid chromatography,” Journal of Chromatography, vol. 204, pp. 285–291, 1981.
[134]
B. Sebille and N. Thuaud, “Determination of tryptophan-human serum albumin binding from retention data and separation of tryptophan enantiomer by high performance liquid chromatography,” Journal of Liquid Chromatography, vol. 3, no. 2, pp. 299–308, 1980.
[135]
J. Gailer and W. Lindner, “On-column formation of arsenic-glutathione species detected by size-exclusion chromatography in conjunction with arsenic-specific detectors,” Journal of Chromatography B, vol. 716, pp. 83–93, 1998.
[136]
A. J. Percy and J. Gailer, “Methylated trivalent arsenic-glutathione complexes are more stable than their arsenite analog,” Bioinorganic Chemistry and Applications, vol. 2008, Article ID 539082, 8 pages, 2008.
[137]
K. Rehman and H. Naranmandura, “Arsenic metabolism and thioarsenicals,” Metallomics, vol. 4, no. 9, pp. 881–892, 2012.
[138]
S. V. Kala, M. W. Neely, G. Kala et al., “The MRP2/cMOAT transporter and arsenic-glutathione complex formation are required for biliary excretion of arsenic,” Journal of Biological Chemistry, vol. 275, no. 43, pp. 33404–33408, 2000.
[139]
E. M. Leslie, “Arsenic-glutathione conjugate transport by the human multidrug resistance proteins (MRPs/ABCCs),” Journal of Inorganic Biochemistry, vol. 108, pp. 141–149, 2012.
[140]
E. M. Leslie, R. G. Deely, and S. P. C. Cole, “Toxicological relevance of the multidrug resistance protein 1, MRP1 (ABCC1) and related transporters,” Toxicology, vol. 167, no. 1, pp. 3–23, 2001.
[141]
N. Ballatori, S. M. Krance, R. Marchan, and C. L. Hammond, “Plasma membrane glutathione transporters and their roles in cell physiology and pathophysiology,” Molecular Aspects of Medicine, vol. 30, no. 1-2, pp. 13–28, 2009.
[142]
R. S. Braman and C. C. Foreback, “Methylated forms of arsenic in the environment,” Science, vol. 182, no. 4118, pp. 1247–1249, 1973.
[143]
A. J. Percy, M. Korbas, G. N. George, and J. Gailer, “Reversed-phase high-performance liquid chromatographic separation of inorganic mercury and methylmercury driven by their different coordination chemistry towards thiols,” Journal of Chromatography A, vol. 1156, no. 1-2, pp. 331–339, 2007.
[144]
?. Balá?, M. Wiese, and J. K. Seydel, “A time hierarchy-based model for kinetics of drug disposition and its use in quantitative structure-activity relationships,” Journal of Pharmaceutical Sciences, vol. 81, no. 9, pp. 849–857, 1992.
[145]
J. D. Meers, E. Z. Jahromi, B. Heyne, and J. Gailer, “Improved RP-HPLC separation of Hg2+ and CH3Hg+ using a mixture of thiol-based mobile phase additives,” Journal of Environmental Science and Health A, vol. 47, no. 1, pp. 149–154, 2012.
[146]
G. Girardi and M. M. Elias, “Effectiveness of N-acetylcysteine in protecting against mercuric chloride-induced nephrotoxicity,” Toxicology, vol. 67, no. 2, pp. 155–164, 1991.
[147]
N. Ballatori, M. W. Lieberman, and W. Wang, “N-acetylcysteine as an antidote in methylmercury poisoning,” Environmental Health Perspectives, vol. 106, no. 5, pp. 267–271, 1998.
[148]
A. S. Koh, T. A. Simmons-Willis, J. B. Pritchard, S. M. Grassl, and N. Ballatori, “Identification of a mechanism by which the methylmercury antidotes N-acetylcysteine and dimercaptopropanesulfonate enhance urinary metal excretion: transport by the renal organic anion transporter-1,” Molecular Pharmacology, vol. 62, no. 4, pp. 921–926, 2002.
[149]
E. Hjorts?, J. S. Fomsgaard, and N. Fogh-Andersen, “Does N-acetylcysteine increase the excretion of trace metals (calcium, magnesium, iron, zinc and copper) when given orally?” European Journal of Clinical Pharmacology, vol. 39, no. 1, pp. 29–31, 1990.
[150]
L. Chapman and H. M. Chan, “The influence of nutrition on methyl mercury intoxication,” Environmental Health Perspectives, vol. 108, no. 1, pp. 29–56, 2000.
[151]
R. K. Zalups and L. H. Lash, “Interactions between glutathione and mercury in the kidney, liver, and blood,” in Toxicology of Metals, L. W. Chang, Ed., pp. 145–163, CRC Lewis Publishers, Boca Raton, Fla, USA, 1996.
[152]
E. Mitchell, S. Frisbie, and B. Sarkar, “Exposure to multiple metals from groundwater-a global crisis: geology, climate change, health effects, testing, and mitigation,” Metallomics, vol. 3, no. 9, pp. 874–908, 2011.
[153]
K. P. DuBois, A. L. Moxon, and O. E. Olson, “Further studies on the effective ness of arsenic in preventing selenium poisoning,” Journal of Nutrition, vol. 19, pp. 477–482, 1940.
[154]
B. Michalke, “Selenspeziation mit SAX-ICP-MS und RPLC-ICP-MS,” in Moderne Techniken Der Ionenanalyse, K. Fischer and D. Jensen, Eds., pp. 50–60, ECOMED Verlagsgesellschaft, Landsberg, Germany, 2002.
[155]
J. Gailer, S. Madden, M. F. Burke, M. B. Denton, and H. V. Aposhian, “Simultaneous multielement-specific detection of a novel glutathione-arsenic-selenium ion [(GS)2AsSe]? by ICP AES after micellar size- exclusion chromatography,” Applied Organometallic Chemistry, vol. 14, no. 7, pp. 355–363, 2000.
[156]
J. Parizek and I. Ostadalova, “The protective effect of small amounts of selenite in sublimate intoxication,” Experientia, vol. 23, no. 2, pp. 142–143, 1967.
[157]
S. Yoneda and K. T. Suzuki, “Equimolar Hg-Se complex binds to selenoprotein P,” Biochemical and Biophysical Research Communications, vol. 231, no. 1, pp. 7–11, 1997.
[158]
M. M. El-Begearmi, H. E. Ganther, and M. L. Sunde, “Dietary interaction between methylmercury, selenium, arsenic, and sulfur amino acids in Japanese quail,” Poultry Science, vol. 61, no. 2, pp. 272–279, 1982.
[159]
A. Naganuma and N. Imura, “Methylmercury binds to a low molecular weight substance in rabbit and human erythrocytes,” Toxicology and Applied Pharmacology, vol. 47, no. 3, pp. 613–616, 1979.
[160]
Y. Sugiura, Y. Tamai, and H. Tanaka, “Selenium protection against mercury toxicity: high binding affinity of methylmercury by selenium-containing ligands in comparison with sulfur-containing ligands,” Bioinorganic Chemistry, vol. 9, no. 2, pp. 167–180, 1978.
[161]
M. Korbas, A. J. Percy, J. Gailer, and G. N. George, “A possible molecular link between the toxicological effects of arsenic, selenium and methylmercury: methylmercury(II) seleno bis(S-glutathionyl) arsenic(III),” JBIC Journal of Biological Inorganic Chemistry, vol. 13, no. 3, pp. 461–470, 2008.
[162]
T. W. Hambley, “Chemistry: metal-based therapeutics,” Science, vol. 318, no. 5855, pp. 1392–1393, 2007.
[163]
K. H. Thompson and C. Orvig, “Boon and bane of metal ions in medicine,” Science, vol. 300, no. 5621, pp. 936–939, 2003.
[164]
M. J. Abrams and B. A. Murrer, “Metal compounds in therapy and diagnosis,” Science, vol. 261, no. 5122, pp. 725–730, 1993.
[165]
A. M. Pizarro and P. J. Sadler, “Unusual DNA binding modes for metal anticancer complexes,” Biochimie, vol. 91, no. 10, pp. 1198–1211, 2009.
[166]
S. Van Zutphen and J. Reedijk, “Targeting platinum anti-tumour drugs: overview of strategies employed to reduce systemic toxicity,” Coordination Chemistry Reviews, vol. 249, no. 24, pp. 2845–2853, 2005.
[167]
T. W. Hambley, “The influence of structure on the activity and toxicity of Pt anti-cancer drugs,” Coordination Chemistry Reviews, vol. 166, pp. 181–223, 1997.
[168]
B. W. Harper, A. M. Krause-Heuer, M. P. Grant, M. Manohar, K. B. Garbutcheon-Singh, and J. R. Aldrich-Wright, “Advances in platinum chemotherapeutics,” Chemistry, vol. 16, no. 24, pp. 7064–7077, 2010.
[169]
J. C. Dabrowiak, J. Goodisman, and A. K. Souid, “Kinetic study of the reaction of cisplatin with thiols,” Drug Metabolism and Disposition, vol. 30, no. 12, pp. 1378–1384, 2002.
[170]
T. Ishikawa and F. Ali-Osman, “Glutathione-associated cis-diamminedichloroplatinum(II) metabolism and ATP-dependent efflux from leukemia cells. Molecular characterization of glutathione-platinum complex and its biological significance,” Journal of Biological Chemistry, vol. 268, no. 27, pp. 20116–20125, 1993.
[171]
K. W. Lee and D. S. J. Martin, “Cis-dichlorodiammineplatinum(II). Aquation equilibria and isotopic exchange of chloride ligands with free chloride and tetrachloroplatinate(II),” Inorganica Chimica Acta, vol. 17, pp. 105–110, 1976.
[172]
A. V. Klein and T. W. Hambley, “Platinum drug distribution in cancer cells and tumors,” Chemical Reviews, vol. 109, no. 10, pp. 4911–4920, 2009.
[173]
X. Wang and Z. Guo, “The role of sulfur in platinum anticancer chemotherapy,” Anti-Cancer Agents in Medicinal Chemistry, vol. 7, no. 1, pp. 19–34, 2007.
[174]
A. I. I. Ivanov, J. Christodoulou, J. A. Parkinson et al., “Cisplatin binding sites on human albumin,” The Journal of Biological Chemistry, vol. 273, no. 24, pp. 14721–14730, 1998.
[175]
L. Trynda-Lemiesz, H. Kozlowski, and B. K. Keppler, “Effect of cis-, trans-diamminedichloroplatinum(II) and DBP on human serum albumin,” Journal of Inorganic Biochemistry, vol. 77, no. 3-4, pp. 141–146, 1999.
[176]
N. Ohta, D. Chen, S. Ito, T. Futo, T. Yotsuyanagi, and K. Ikeda, “Effect of trans-diamminedichloroplatinum(II) on human serum albumin: conformational changes through partial disulfide bond cleavage,” International Journal of Pharmaceutics, vol. 118, no. 1, pp. 85–93, 1995.
[177]
D. Esteban-Fernández, M. Montes-Bayón, E. B. González, M. M. Gómez-Gómez, M. A. Palacios, and A. Sanz-Medel, “Atomic (HPLC-ICP-MS) and molecular mass spectrometry (ESI-Q-TOF) to study cis-platin interactions with serum proteins,” Journal of Analytical Atomic Spectrometry, vol. 23, pp. 378–384, 2008.
[178]
H. Sun, H. Li, and P. J. Sadler, “Transferrin as a metal ion mediator,” Chemical Reviews, vol. 99, no. 9, pp. 2817–2842, 1999.
[179]
J. M. El Hage Chahine, M. Hemadi, and N. T. Ha-Duong, “Uptake and release of metal ions by transferrin and interaction with receptor,” Biochimica et Biophysica Acta, vol. 1820, no. 3, pp. 334–347, 2012.
[180]
M. Groessl, M. Terenghi, A. Casini, L. Elviri, R. Lobinski, and P. J. Dyson, “Reactivity of anticancer metallodrugs with serum proteins: new insights from size exclusion chromatography-ICP-MS and ESI-MS,” Journal of Analytical Atomic Spectrometry, vol. 25, no. 3, pp. 305–313, 2010.
[181]
A. J. Stewart, C. A. Blindauer, S. Berezenko, D. Sleep, and P. J. Sadler, “Interdomain zinc site on human albumin,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 7, pp. 3701–3706, 2003.
[182]
C. B. Kissinger and P. T. Kissinger, “Your bioanalytical data are only as good as your samples,” Bioanalysis, vol. 4, no. 12, pp. 1411–1415, 2012.
[183]
S. D?ker, S. Mounicou, M. Do?an, and R. Lobinski, “Probing the metal-homeostatis effects of the administration of chromium(vi) to mice by ICP MS and size-exclusion chromatography-ICP MS,” Metallomics, vol. 2, no. 8, pp. 549–555, 2010.
[184]
M. Gonzalez-Fernández, M. A. García-Sevilliano, R. Jara-Biedma et al., “Size characterization of metal species in liver and brain from free-living (Mus spretus) and laboratory (Mus Musculus) mice by SEC-ICP-MS: application to environmental contamination assessment,” Journal of Analytical Atomic Spectrometry, vol. 26, no. 1, pp. 141–149, 2011.
[185]
R. Montes-Nieto, C. A. Fuentes-Almagro, D. Bonilla-Valverde et al., “Proteomics in free-living Mus spretus to monitor terrestrial ecosystems,” Proteomics, vol. 7, no. 23, pp. 4376–4387, 2007.
[186]
J. Riuz-Laguna, N. Abril, T. García-Barrera, J. L. Gómez-Ariza, J. López-Barea, and C. Pueyo, “Absolute transcript expression signatures of Cyp and Gst genes in Mus spretus to detect environmental contamination,” Environmental Science & Technology, vol. 40, no. 11, pp. 3646–3652, 2006.
[187]
Y. H. Huang, C. M. Shih, C. J. Huang et al., “Effects of cadmium on structure and enzymatic activity of Cu, Zn-SOD and oxidative status in neural cells,” Journal of Cellular Biochemistry, vol. 98, no. 3, pp. 577–589, 2006.
[188]
L. E. Scott and C. Orvig, “Medicinal inorganic chemistry approaches to passivation and removal of aberrant metal ions in disease,” Chemical Reviews, vol. 109, no. 10, pp. 4885–4910, 2009.
[189]
W. B. Cannon, “Organization for physiological homeostasis,” Physiological Reviews, vol. 9, no. 3, pp. 399–431, 1929.
[190]
W. J. Crinnion and J. Q. Tran, “Organic foods contain higher levels of certain nutrients, lower levels of pesticides, and may provide health benefits for the consumer,” Alternative Medicine Review, vol. 15, pp. 303–310, 2010.
[191]
L. Charlet, Y. Chapron, P. Faller, R. Kirsch, A. T. Stone, and P. C. Baveye, “Neurodegenerative diseases and exposure to the environmental metals Mn, Pb, and Hg,” Coordination Chemistry Reviews, vol. 256, no. 19-20, pp. 2147–2163, 2012.
[192]
G. N. George, R. C. Prince, J. Gailer et al., “Mercury binding to the chelation therapy agents DMSA and DMPS and the rational design of custom chelators for mercury,” Chemical Research in Toxicology, vol. 17, no. 8, pp. 999–1006, 2004.
[193]
J. W. Scannell, A. Blanckley, H. Boldon, and B. Warrington, “Diagnosing the decline in pharmaceutical R&D efficiency,” Nature Reviews Drug Discovery, vol. 11, pp. 191–200, 2012.
[194]
D. Malakoff, “Can treatment costs be tamed?” Science, vol. 331, no. 6024, pp. 1545–1547, 2011.
[195]
S. Castellino, “MALDI imaging MS analysis of drug distribution in tissue: the right time!(?),” Bioanalysis, vol. 4, no. 21, pp. 2549–2551, 2012.
[196]
S. Tuncel, F. Dumoulin, J. Gailer et al., “A set of highly water-soluble tetraethyleneglycol-substituted Zn(II) phthalocyanines: synthesis, photochemical and photophysical properties, interaction with plasma proteins and in vitro phototoxicity,” Dalton Transactions, vol. 40, no. 16, pp. 4067–4079, 2011.
[197]
J. Moretto, B. Chauffert, F. Ghiringhelli, J. R. Aldrich-Wright, and F. Bouyer, “Discrepancy between in vitro and in vivo antitumor effect of a new platinum(II) metallointercalator,” Investigational New Drugs, vol. 29, no. 6, pp. 1164–1176, 2011.
[198]
J. Szpunar, A. Makarov, T. Pieper, B. K. Keppler, and R. Lobinski, “Investigation of metallodrug-protein interactions by size-exclusion chromatography coupled with inductively coupled plasma mass spectrometry (ICP-MS),” Analytica Chimica Acta, vol. 387, no. 2, pp. 135–144, 1999.
[199]
R. Mandal, R. Kalke, and X. F. Li, “Interaction of oxaliplatin, cisplatin, and carboplatin with hemoglobin and the resulting release of a heme group,” Chemical Research in Toxicology, vol. 17, no. 10, pp. 1391–1397, 2004.
[200]
R. Baliga, Z. Zhang, M. Baliga, N. Ueda, and S. V. Shah, “In vitro and in vivo evidence suggesting a role for iron in cisplatin-induced nephrotoxicity,” Kidney International, vol. 53, no. 2, pp. 394–401, 1998.
[201]
V. Calderone, A. Casini, S. Mangani, L. Messori, and P. L. Orioli, “Structural investigation of cisplatin-protein interactions: selective platination of His19 in a cuprozinc superoxide dismutase,” Angewandte Chemie, vol. 45, no. 8, pp. 1267–1269, 2006.
[202]
X. Sun, C. N. Tsang, and H. Sun, “Identification and characterization of metallodrug binding proteins by (metallo) proteomics,” Metallomics, vol. 1, no. 1, pp. 25–31, 2009.
[203]
K. L. Pei and J. Gailer, “Probing the interaction of arsenobetaine with blood plasma constituents in vitro: an SEC-ICP-AES study,” Metallomics, vol. 1, no. 5, pp. 403–408, 2009.
[204]
D. A. Smith, L. Di, and E. H. Kerns, “The effect of plasma protein binding on in vivo efficacy: misconceptions in drug discovery,” Nature Reviews Drug Discovery, vol. 9, pp. 929–939, 2010.
[205]
M. Kirberger and J. J. Yang, “Structural differences between Pb2+- and Ca2+-binding sites in proteins: implications with respect to toxicity,” Journal of Inorganic Biochemistry, vol. 102, no. 10, pp. 1901–1909, 2008.
[206]
J. M. Swanson, S. Entringer, C. Buss, and P. D. Wadhwa, “Developmental origins of health and disease: environmental exposures,” Seminars in Reproductive Medicine, vol. 27, no. 5, pp. 391–402, 2009.
[207]
L. Trasande, P. J. Landrigan, and C. Schechter, “Public health and economic consequences of methyl mercury toxicity to the developing brain,” Environmental Health Perspectives, vol. 113, no. 5, pp. 590–596, 2005.
[208]
L. D. Hylander and M. E. Goodsite, “Environmental costs of mercury pollution,” Science of the Total Environment, vol. 368, no. 1, pp. 352–370, 2006.
[209]
J. J. Wirth and R. S. Mijal, “Adverse effects of low level heavy metal exposure on male reproductive function,” Systems Biology in Reproductive Medicine, vol. 56, no. 2, pp. 147–167, 2010.
[210]
L. Jarup, “Hazards of heavy metal contamination,” British Medical Bulletin, vol. 68, no. 1, pp. 167–182, 2003.
[211]
B. H. Robinson, “E-waste: an assessment of global production and environmental impacts,” Science of the Total Environment, vol. 408, no. 2, pp. 183–191, 2009.
[212]
G. Zheng, X. Xu, K. Wu, T. A. Yekeen, and X. Huo, “Association between lung function in school children and exposure to three transition metals from an e-waste recycling area,” Journal of Exposure Science and Environmental Epidemiology, vol. 23, pp. 67–72, 2013.
[213]
S.-R. Lim, D. Kang, O. A. Ogunseitan, and J. M. Schoenung, “Potential environmental impacts from the metals in incandescent, compact fluorescent lamp (CFL), and light-emitting diode (LED) bulbs,” Environmental Science and Technology, vol. 47, no. 2, pp. 1040–1047, 2013.
[214]
Y. Liu, C. Wen, and X. Liu, “China’s food security soiled by contamination,” Science, vol. 339, pp. 1382–1383, 2013.