Sei sulla pagina 1di 45

C H A P T E R

56

Tin
ELENA A. OSTRAKHOVITCH

ABSTRACT its deposition in lungs and may cause “stannosis,” a


benign pneumoconiosis. Some tin salts are irritating or
Inorganic tin compounds occur naturally in the can liberate toxic fumes during decomposition. Gas-
Earth’s crust. In the environment, tin can be found in trointestinal absorption of soluble tin salts is only a
both inorganic and organic forms. Inorganic tin com- few percent of the ingested dose. In chronic exposure,
pounds are released from natural and anthropogenic tin tends to accumulate in kidney, liver, and bones.
sources. The conversion of metallic tin forms to com- Bones comprise a major site of deposition of tin after
pounds that may be more soluble increases the risk of long-term exposure and intramuscular injection. The
exposure and toxicity. Organotin compounds in the biological half-life of tin in bone is approximately 100
environment largely originate from anthropogenic days. Exposure to high concentrations of inorganic tin
activities. However, chemical and biochemical meth- may cause gastrointestinal illness, as well as liver and
ylation reactions convert inorganic tin compounds into kidney problems. No noteworthy histopathological
methyltin form. Biomethylation of alkyltins results in observations of nonneoplastic nature were reported in
the accumulation of more toxic organotin compounds. a long-term study.
Tin is not regarded an essential nutrient for humans. Short-chain alkyls are easily absorbed from gastro-
However, tin is considered an essential for the growth intestinal tract. Some alkyltin compounds, particularly
of rats, with a requirement for tin of between 1 and tributyltin and triphenyltin, have high toxicity. Chronic
2 mg/kg in the diet. Elemental tin, inorganic tin com- exposure to butyltin compounds causes imposex,
pounds, and long-chain alkyltins are poorly absorbed a pathological condition characterized by develop-
when ingested, which accounts for their relatively ment of male sex characteristics in female gastropods.
low toxicity. The toxicity of organotin compounds Toxicity of organotins in humans is most frequently
depends on the length of alkyl chain, hydrophobicity, reported as loss of memory and seizure, as well as
and other physicochemical properties. Hydrophobic other symptoms including death. Short-chain alkyl-
organotins are toxic to a wide variety of organisms tin, particularly the trialkyl derivatives, and aromatic
owing to their high solubility in cell membranes. The tin compounds are neurotoxic. Hydrophobic tri-
alkyl tins are more toxic than the aryl tins, whereas methyltin and triethyltin compounds readily diffuse
the toxicity of short-chain alkyltin compounds such as into richly lipophilic tissues such as brain and cause
trimethyl- and triethyltin is higher than that of long- encephalopathy, cerebral edema, and severe seizures.
chain compounds: toxicity increases with number of Tetraalkyltins are enzymatically converted to the tri-
alkyl groups. The excretory routes of tin compounds substituted form and exert delayed but similar neuro-
may vary depending of the type of compounds and the toxic effects. Tributyltin compounds are less toxic than
mode of exposure. trimethyl- and triethyltins. They may be strongly irri-
Insoluble inorganic tin compounds are largely tating to the skin in humans. It was reported that ship-
nontoxic. However, inhalation of tin dust results in yard workers exposed to tributyltin oxide developed

Handbook on the Toxicology of Metals 4E


http://dx.doi.org/10.1016/B978-0-444-59453-2.00056-1 1241 Copyright © 2015 Elsevier B.V. All rights reserved.
1242 Elena A. Ostrakhovitch

severe dermatitis, difficulty in breathing, and flu-like organic alkyl or aryl group and L is an organic or inor-
symptoms. Trisubstituted organotin derivatives are ganic ligand. The noncarbon-bonded anionic group is
implicated in hepatotoxicity, immunodeficiency, endo- commonly halide, oxide, hydroxide, carboxylate, or
crine disruption, and both reproductive anomalies mercaptide (Kirk-Othmer Encyclopedia of Chemical
and infertility in laboratory animals. Monobutyltin Technology, 2000). Mono-, di-, tri-, and tetraorganotin
and dibutyltin show genotoxicity, while mono- and compounds can be formed (Alzieu et al., 1989; Alzieu
dimethyltin are not genotoxic. Organotin compounds et al., 1986; Errecalde et al., 1995). Many diorganotin
penetrate the cell membrane and interrupt oxidative oxides (R2SnO)n are oligomeric. The chemical proper-
phosphorylation, disturb calcium homeostasis, dam- ties of organotin compounds depend upon their struc-
age mitochondria, and induce apoptosis. tures; however, water solubility across the group is
very low. Tetraalkyl- and tetraaryltin compounds exist
under all conditions as tetrahedral monomers; all are
1  PHYSICAL AND CHEMICAL PROPERTIES insoluble in water but are soluble in various organic
solvents. The tin-carbon bond in tetraorganotins is eas-
Tin [from the Latin word stannum (Sn)] is a chemi- ily cleaved by mineral acids. For example, tetrabutyl-
cal element of Group 14 (previously Group IVA) of tin is stable, combustible, and colorless liquid at room
the Periodic Table, with atomic number 50 and atomic temperature. Trisubstituted compounds usually give
weight 118.71. Its abbreviated electron configuration five-coordinated complexes R3SnL. The triorganotin
is [Kr]4d105s25p2. The melting point of tin is 231.93°C halides are insoluble in water, except for trimethyltin
(449.474°F) and the boiling point is 2586°C (4686.8°F) chloride, which is completely water soluble. Tributyl-
(CRC Handbook, 2011). Tin has 10 stable isotopes and tin is unstable unless it forms an oxide [bis(tributyltin)
exists in three allotropic forms known as gray tin (alpha oxide], which is a colorless to pale yellow liquid with
tin), with a cubic structure, white metallic tetragonal tin a melting point of −45°C, a boiling point of 180°C,
(beta tin), and metallic rhombic tin (gamma tin or tin and low water solubility (20 mg/kg). The diorganotin
brittle). When white tin cools below 13.2°C, it changes chlorides, bromides, and iodides are soluble in many
from a white to a gray form, a change commonly called organic solvents, whereas dimethyltin dichloride is
“tin pest.” Tin is insoluble in water and is resistant to soluble in water. Commercial grades of diorganotin
oxygen. Because of its low electropositivity, tin may carboxylates frequently have wider melting ranges
be obtained from cassiterite (SnO2) by reduction with because of the use of less pure grades of carboxylic
carbon. Tin compounds exist in two main oxidation acids. Unlike diorganotins, monoorganotins are hygro-
states, +2 and +4, with the +4 state being more stable. scopic, low-melting point solids or liquids, which can
Tin forms inorganic (without a tin-carbon bond) be hydrolyzed in water that contains enough acid to
and organic compounds (with tin-carbon bonding). retard hydrolysis. Among the organotin compounds,
Inorganic tin has four oxidation states: −2, 0, +2, and the most widely investigated are tributyltin oxide
+4. The Chemical Abstracts Service (CAS) registry (CAS registry number, 56-35-9), triphenyltin (CAS reg-
number for tin inorganic compounds (except oxides) istry number, 76-87-9), dibutyltin dilaurate (CAS regis-
is 7440-31-5. Inorganic tin compounds are divided into try number, 77-58-7), and tetramethyl tin (CAS registry
two classes: stannous, or tin(II), compounds; and stan- number, 594-27-4).
nic, or tin(IV), compounds. Stannous compounds are There are many naturally occurring organotin com-
readily oxidized at high pH. Both stannous and stan- pounds (mainly methylated forms) that are mostly
nic compounds tend to hydrolyze in solution (Pettine products of the methylation of inorganic tin observed in
et al., 1981). Tin forms stannic acid, H2SnO4, when estuarine, sewage waters, and sediment (Quevauviller
heated in air or oxygen at high temperature and pro- et al., 1989). However, most organotins entering the
duces metastannic acid, H2SnO3, in concentrated nitric environment are introduced deliberately as man-made
acid. Tin dissolves in hydrochloric acid to form stan- products (e.g. biocides or stabilizers).
nous chloride (SnCl2). Among the widely used inor-
ganic tin compounds are stannous chloride (SnCl2),
stannous oxide (SnO), stannous fluoride (SnF2), and 2  METHODS AND ANALYSIS
stannic chloride (SnCl4). The CAS registry numbers
for stannous oxide (SnO) and stannic oxide (SnO2) are A range of analytical methods, from the simple to
21651-19-4 and 18282-10-5, respectively. the more sophisticated, have been used to determine
Organotin compounds are generally classified inorganic tin and organotin compounds. Detection
according to the tin oxidation state. They are rep- methods include anodic stripping voltammetry, colo-
resented by the formula RxSn(L)(4 - x), where R is an rimetry, spectrophotometry, fluorescence spectrometry,
56 Tin 1243

ion chromatography, polarography, titration, atomic a glassy carbon electrode with bismuth and a film of
absorption spectrometry (AAS), gas chromatography poly(bromophenol blue) allows one to measure tin in
(GC), mass spectrometry (MS), and inductively cou- canned food with a detection limit of 7 nM (Yang et al.,
pled plasma (ICP) techniques (Boa Morte et al., 2009; 2012). A linear sweep voltammetric approach was pro-
Detcheva and Grobecker, 2006; Ozcan and Akman, posed for gaining rapid information on tin release in
2000; Yu et al., 2010; Aghaie et al., 2005; Li et al., processed canned food (Toniolo et al., 2009). Although
1999). Numerous techniques have been developed to this method does not provide quantitative data on
improve sample preparation and detection methods metal release, it determines the thermodynamic condi-
to achieve better sensitivity, accuracy, precision, and tions controlling a specific tinplate corrosion process
detection limits through the enhancement of features and allows one to establish whether epoxyphenolic
such as miniaturization and the acceleration of ana- lacquered tinplates should be employed.
lytical processes. However, despite great strides being An AAS analytical method for the detection of tin
made, the detection of organotin compounds remains was widely used in the past for the detection of inor-
a challenging task. A major challenge is the develop- ganic tin in metallurgical samples and biological mate-
ment of effective diluted sample preparation strategies rials. However, in routine operations where tin level is
that must be implemented before samples are actually below 3-5 μg, flame AAS has an unsatisfactory detec-
subjected to detection analysis. Therefore, the search tion power (Engberg, 1973). The replacement of flame
continues for sensitive analytical methods capable of with an electrically heated graphite tube increases the
providing quantitative analysis for various classes of atom density and residence time, which improves
organotin compounds at very low limits of detection. graphite furnace (GF)-AAS detection limits by a fac-
In the past, many colorimetric methods have been tor of up to 1000 times compared to flame AAS, thus
applied for the detection of tin, using dithiol, catechol bringing the detection limits down to the 0.1-05 μg/L
violet, phenylfluorone, oxine (8-quinolinol), dithi- range. Furthermore, GF-AAS allows the direct deter-
zone, and quercetin (Adcock and Hope, 1970; Corbin, mination of tin in solid samples (Hirano et al., 2001)
1973). However, these methods are not very sensitive However, because of the temperature limitation, the
and they do not distinguish between the various tin refractory element performance is still somewhat lim-
compounds that may be present in the sample. Thus, ited. Furthermore, limitations of GF-AAS are the inter-
for catechol violet, sensitivity was given as 0.1 μg tin action of tin with graphite during the atomization step
in the sample (Corbin, 1973). Compared with the con- and matrix interference. In electrothermal atomizers
ventional spectrophotometer, charge-coupled device (ET-AAS), which include GF, increased light absorp-
diode array detectors can improve precision and the tion lowers the detection limits. This technique was
signal-to-noise ratio and rapidly scan the absorption developed in the 1990s and was widely used for the
spectrum. Application of the diode array detector in detection of tin in biological materials and sediments.
the sequential injection analysis methodology devel- ET-AAS can be used for the analysis of tin in water
oped for the determination of Sn in canned food gen- (NEMI, Standard Method 3113B; APHA, 1998a,b).
erated data in good agreement with electrothermal Combined with high-performance liquid chromatog-
(ET)-AAS (Boa Morte et al., 2009). raphy (HPLC) separation, ET-AAS showed higher
Anodic stripping voltammetric analysis has shown selectivity and sensitivity (Quevauviller et al., 1993).
good sensitivity, for micromolar level detection of tri- In combination with a hydride generation system GF-
phenyltin (Booth and Fleet, 1970; Florence and Farrar, AAS technique (HG-GF-AAS), tin is reduced to its
1974). However, the critical step in stripping experi- volatile hydride, which allows direct analysis of solid
ments is the preconcentration (or deposition) step, samples with higher sensitivity (Hirano Y. et al., 2001).
when analyte compounds are deposited on the elec- However, the dependence of the hydride species on
trode from the solution, as well as the problem of the acidity of the sample solution makes this technique
overlapping stripping signals (for example, stripping less reliable. The development of ICP atomic emission
signals for tin and lead) in multianalyte determina- spectroscopy (ICP-AES) reduced the interference of the
tion. The overlap problem can be overcome either sample matrix, enhanced productivity, and improved
by chemical separation of tin or by using a stripping the detection limits and dynamic ranges for quantita-
voltammetric procedure at the bismuth film electrode tive multielement analysis. As with emission spectros-
(in place of a mercury film electrode) in the presence copy, the samples are atomized in this method. The U.S.
of a supporting electrolyte (Prior, 2010; Hutton et al., Environmental Protection Agency (USEPA) approved
2006; Frena et al., 2011). This enhanced procedure ICP-AES for the determination of metals in environ-
improves the detection limits for tin (to 1.9 μg/L) and mental samples and drinking water analysis (USEPA,
enables multielement detection. The modification of 1994a,b). ICP-AES environmental applications require
1244 Elena A. Ostrakhovitch

that (1) samples must be in a liquid form so as to be was developed to measure tin isotopic composition
aspired through a nebulizer to increase the mass trans- with high precision: multicollector ICP-MS (MC-ICP-
fer efficiency and (2) suspended soils and sediments MS) on a Nu Plasma 500 (Moynier et al., 2009). Tin
must be first digested to extract the metal. Drinking isotopes have been fractionated by liquid-liquid tin
water with turbidity higher than one normal turbidity isotope extraction with a crown ether, dicyclohexano-
unit must also be digested prior to analysis for the suc- 18-crown-6 (or DC18C6).
cessful separation of the analyte from the matrix. The Where determination of specific organotin com-
detection limit was determined as 30 μg/L (Perring and pounds is required, a separation technique such as GC
Basic-Dvorzak, 2002). ICP-AES has been used for the with a flame-ionization detector (FID), a flame pho-
detection of tin in water, sediments, plants, animal tis- tometric detector (FPD), a pulsed flame photometric
sue, air filters, soil, leachates, effluents, food samples, detector (PFPD), or a thermal conductivity detector
and biominerals (Yokoi et al., 1990a; Elekes et al., 2010; (TCD) is useful. The GC-FPD or GC-FID techniques
Afonso et al., 2010; Perring and Basic-Dvorzak, 2002; cannot provide structural information. However, GC-
Boutakhrit et al., 2011). ICP-AES is the preferred method PFPD offers a compromise between analytical cost and
for the routine analysis of tin in canned food due to its satisfactory levels of sensitivity, selectivity, and reli-
good accuracy, reliability, and ease of use. The method ability for the detection of organotin compounds in a
was recommended for measurements of airborne broad range of soils (Jing and Amirav, 1998; H ­ eroult
inorganic tin and its compounds, except for oxides et al., 2008). A method combining the advantages
(NIOSH, 1994). In 2002, a combined nebulizer/gas- of both GC and MS (i.e. GC-MS) has been accepted
liquid separator design was introduced (McLaughlin as a “gold standard” technique for the detection of
and Brindle, 2002). A multimode sample introduction volatile and semivolatile organic compounds. Meth-
system (MSIS) was patented in 2005; it is capable of ods most frequently used are GC-MS and HPLC-­
operating as a conventional spray chamber, as a gas- coupled MS (Kawaguchi et al., 2004; Devos et al., 2005;
liquid separator, or both (McLaughlin et al., 2006). Gas Lopez de Alda et al., 2001). GC-coupled ICP-MS (GC-
from the nebulizer strips the volatile species and deliv- ICP-MS) provides the possibility of applying species-
ers them to the plasma. The MSIS diminishes the sig- specific isotope dilution analysis for the quantification
nal noise and improves detection limits. The Canadian organometallic compounds (Monperrus et al., 2003;
Association for Environmental Analytical Laboratories Pinel-Raffaitin et al., 2007). The detection limits of
has approved the MSIS application for the detection alkylated tin compounds were in the range of 0.01 to
of elements in sludges, biosolids, soils, drinking water, 0.04 μg Sn/L, with the best detection limits for dibutyl-
and wastewater. and tributyltin compounds. A comparison of ICP-MS,
ICP-MS also uses an ICP plasma source to dissoci- HG-ICP-AES, ETA-AAS, and ICP-AES techniques for
ate the sample into atoms and ions. However, ICP-MS the routine analysis of tin in canned food demonstrated
detects ions separated on the basis of their atomic mass- that ICP-AES is the preferred method for the range of
charge ratio rather than by the light they emit. ICP-MS 50-200 mg/kg due to its good analytical performance,
has been used for the detection of tin compounds in reliability, and ease of use (Boutakhrit et al., 2011).
urine samples, food samples, soils, waters, and sedi- GC-MS analysis with FID was found to provide
ments with high levels of precision and reproducibility adequate sensitivity for the detection of volatile alkyl-
(Sieniawska et al., 2012; Millour et al., 2011; Boutakhrit tins. The detection limit of this analytical procedure
et al., 2011; APHA, 1998c). The ICP-MS detection limit for alkyltins was calculated to be 4.8 pg, with the
for tin in canned food was 0.1 μg/kg, as compared to detection limit of the overall procedure (measured as
0.5 μg/kg for HG-ICP-AES and 2 mg/kg for ICP-AES mass per sample and expressed as equivalent air con-
(Boutakhrit et al., 2011). Tin is not specifically listed as centration) calculated as 0.21 μg/sample (4.3 μg/m3).
an analyte for ICP-MS (NEMI, Standard Method 3125). However, a major drawback of GC for tri-, di-, and
However, it can be quantified in water, canned milk, monosubstituted organotin detection is the require-
and in fruits by ICP-MS (ISO, 2003a,b, 2004). Laser ment of a derivatization step because of the poor
ablation ICP-MS (LA-ICP-MS) allows mulielemental volatility of these organotin compounds. GC-ICP-MS
analysis without extensive sample preparation with has been employed for the detection of organometal-
a shortened analysis time. LA-ICP-MS was shown to lic compounds of tin (Rajendran et al., 2000; Uveges
be an efficient method for the characterization of trace et al., 2007; Inagaki et al., 2007; Pinel-Raffaitin et al.,
metals in complex solid matrices such as heteroge- 2007). A major challenge of the GC-ICP-MS techniques
neous domestic wastes and polymer waste, as well as is to avoid thermal degradation of analyzed com-
compost (Motelica-Heino et al., 1998; Jimenez et al., pounds in the injector and column and during trans-
2007; Stehrer et al., 2010). Recently, a new technique port from the column to the ICP torch. Capillary GC
56 Tin 1245

coupled to a PFPD achieved a low ng/L detection level enabling the separation of nonvolatile species. HPLC
range for pesticidal and nonpesticidal organotin com- coupled to ICP-MS (HPLC-ICP-MS) or to ESI-MS, tan-
pounds (Evans et al., 2009). Derivatization of organo- dem MS/MS, and AAS increase its application range.
tin compounds with tetraethylborate was coupled to LC-MS/MS can be performed without complete sep-
liquid-liquid extraction. Capillary GC coupled to high aration of the different compounds, which makes it
resolution double-focusing magnetic sector MS detec- highly suitable for high-throughput analyses. The LC-
tion offers high resolution and sensitivity in the detec- ESI-MS/MS method of detection allows quantification
tion of tin species, bringing the detection limits below in the low range of 0.01-0.02 μg/L. HPLC-coupled MS
3 ng/L for inorganic tin, monophenyltin, diphenyltin, (HPLC-MS) was developed for speciation analysis of
triphenyltin, and monobutyltin. For dibutyltin and methylated tins, such as mono-, di- and trimethyl-
tributyltin, the detection limits were calculated to be ated tin species, dibutyltin and triphenyltin (Wahlen
3.4 and 7.3 ng/L, respectively (McSheehy et al., 2012). and Catterick, 2003; Wang et al., 2008; Yoo et al., 2007;
HR-ICP-MS detection shows a high resolving power, Suzuki et al., 2008). HPLC-ICP-MS was applied for the
high sensitivity, and an extremely low background, simultaneous detection of dibutyl-, tributyl-, diphenyl-,
exceeding the performance of a quadrupole instrument. and triphenyl tin species in shellfish samples with
An alternative to GC analysis is liquid chromatogra- detection limits ranging from 0.24 to 0.37 μg/L and
phy (LC), which does not require the prior derivatiza- recoveries of over 80% for dibutyl- and diphenyltins
tion of organotins. Ion exchange LC and reversed (Yu, Z.H. et al., 2010). The limits of ESI-MS detection
phase LC have been proposed to achieve the separa- were 0.4 μg/L for tributyltin and 20 μg/L for triphen-
tion of mono-, di-, and trialkyl tin derivatives. Origi- yltin compounds (Wang et al., 2008). However, MS/
nally, coupling GC with ultraviolet (UV) detection, MS was proven to be insufficient for the detection of
ICP optical emission spectrophotometry (ICP-OES), di- and trimethylated tin compounds in diluted sam-
flame atomic absorption spectrophotometry (FAAS), ples. Multiple reactions monitoring mode was applied
or GF-AAS was discounted because of poor sensitivity in order to achieve a higher sensitivity with MS/MS
(Gonzalez-Toledo et al., 2003). In ICP-OES, the radiation detection and to reach the detection limit of 0.1 mg/L
emitted is measured at a unique wavelength, whereas or less for trimethylated tin (Suzuki et al., 2008). An
in ICP-MS the tail flame of the plasma is extracted into HPLC-coupled HG-ICP-MS system was developed
a low-pressure interface containing the sampler and a for rapid, direct, and sensitive speciation of methyl-
skimmer. The ions are transmitted to a mass analyzer tins in seawater without any pretreatment step, with
via an ion lens system. Although ICP-OES allows one detection limits of 0.266 and 0.095 ng/mL for mono-
to simultaneously detect metal ions at different concen- dimethyltins, respectively, and as low as 0.039 ng/
tration, the detection limits are in the μg/L range. ICP- mL for trimethyltin (Zhai et al., 2009). However, some
MS offers the advantages of ICP-OES along with those basic analytical problems such as incomplete separa-
of mass spectral acquisition, such as excellent sensitiv- tion, column packing, and column and buffer solu-
ity, enhanced detection limits (reaching ng/L levels), tions competing for bioinorganic ligands remain to be
and isotopic analysis. LC-coupled electrospray ioniza- solved. The development of hydrophilic interaction
tion MS (ESI-MS) was proven to be a powerful method LC-ESI-MS analysis allows one to analyze the concen-
for detection of triorganotin compounds in water trations of tributyltin and triphenyltin compounds in
samples. ESI-MS (Q, quadrupole, quadrupole ion trap, seawater at parts-per-trillion levels (Sano et al., 2010).
QqQ, triple quadrupole, and QqTOF, and quadrupole- Detection methods such as ICP-MS and ESI-MS are
time-of-flight detection) is suitable for both polar and extremely sensitive. However, there are several dis-
ionic organometallic compounds and allows the analy- advantages. The presence of the organic phase and
sis of highly dilute solutions (Henderson and ­Taylor, the proportion of salt in the mobile phase during LC
1996; Henderson and McIndoe, 2005; Baul et al., 2008; separation may jeopardize accurate quantification by
Chalupa et al., 2008). The ­limits of detection for tribu- ICP-MS and ESI-MS as a result of increased spectral
tyltin and triphenyltin were 10 and 20 pg, respectively overlap, leading to positively biased results. There-
(Sano et al., 2010). However, ESI-MS detection also fore, restrictions are placed on the type of eluents and
imposes some restrictions related to chromatographic concentration of salts that can be employed. Capillary
separation. Fluorescence detection has found an electrophoresis is an alternative to chromatography
application in LC as a sensitive detection approach for the separation of organometallic compounds. Elec-
to tin speciation. However, organotin compounds trophoresis in conjunction with ICP-MS was devel-
are nonfluorescent, and derivatization reagents such oped for the separation and analysis of trimethyl-,
as ­flavone derivatives are required to allow their detec- tributyl-, dibutyl-, and monobutyltin compounds
tion. The application of HPLC has the advantage of (Sun et al., 2010). The authors applied a semipermanent
1246 Elena A. Ostrakhovitch

coating of dioctadecyl dimethyl ammonium chloride GC analysis can be accomplished using either the
to enhance methyltin separation. The detection limits Grignard reaction or alkylation with sodium alkylbo-
for these organotin compounds ranged from 0.037 to rate. However, nonaqueous alkylation, such as the use of
0.112 mg/L, with method accuracies of 8.6 and 7.8% for Grignard reagent, is less used due to complex multistep
tributyltin and dibutyltin, respectively. analytical procedure and the requirement for strictly
Samples are commonly complex and heteroge- anhydrous conditions. The advantage of using alkylbo-
neous, with low species concentration. Thus, for rate is its stability in water, compared to the hydrolytic
sediment samples, sieving is often used to limit the instability of Grignard reagents (Dabek-Zlotorzynska
heterogeneity of samples (Quevauviller and Donard, et al., 1998). However, the main disadvantage of using
1990). Acidification of filtered water samples and NaBEt4 is its short lifetime in the presence of oxygen
freezing followed by oven-drying of sediment samples and moisture. The rate of derivatization depends on
was shown to be suitable for preserving the stability the pH of the medium, metal ion concentration, and
of organotin compounds. The samples often require temperature. The working pH range for the derivatiza-
additional clean-up and preconcentration to obtain a tion reaction is between 4.0 and 5.0. Cai and Bayona
sufficiently high amount of sample for measurement. (1995) have optimized the pH range for the ethyl-
Four main methods of sample digestion are commonly ation reaction of organotins by sodium tetraethylbo-
reported by laboratories: dry ashing of the sample in a rate without any additional clean-up step. Tropolone
conventional oven; microwave digestion of the sample has been used as chelating agent for the derivatiza-
in a strong acid; acid digestion of the sample by heating tion of tin ions (Lu and Whang, 1995; Inagaki et al.,
in a pressure vessel; and dissolving the sample directly 2007). However, the most widely used methods for
into acid. Some laboratories also extract the metals in tin derivatization are ethylation and, to a lesser extent,
2-methylhexan-2-one isobutyl methyl ketone. Meth- propylation using sodium tetraalkylborates [sodium
ods of sampling and analysis for the official control of tetraethylborate (NaBEt4), tetrapropylborate (NaBPr4),
the level of inorganic tin was implemented by Euro- bromomagnesium tetrapropylborate (BrMgPr4B), and
pean Commission regulation No 836/2011 amend- bromomagnesium tetraethylborate (BrMgEt4B)] (de
ing European Commission regulation No 333/2007 Smaele et al., 1998; Ashby and Craig, 1988b; Bergmann
(European Commission, 2011). Microwave diges- and Neidhart, 2001; Nsengimana et al., 2009). Methods
tion was approved by the USEPA using closed-vessel of organometallic tin extraction include liquid-liquid
method, which employs hydrofluoric acid, for the extraction and solid-liquid extraction procedures.
solubilization of all suspended solids. It was speci- However, both of these methods are time-consuming
fied by Occupational Safety and Health Administra- and require high amounts of organic solvents in order
tion (OSHA) that specific organotins must be sampled to isolate and preconcentrate the derivatized ana-
and analyzed until a satisfactory procedure for sepa- lytes (Zachariadis and Rosenberg, 2009a,b). However,
rating the organic tin compounds as a group from the ­liquid-liquid extraction in hexane after derivatization
inorganic ones is found. The National Institute for by tetraethyl- or tetrapropylborate reagents was suc-
Occupational Safety and Health (NIOSH) method for cessfully applied for the fast speciation of tributyltin
organotin compounds (NIOSH, 1994) recommended compounds in human urine (Kapsimali et al., 2012).
sampling with a glass fiber filter/XAD-2 sorbent tube, With regard to the trend toward sample size miniatur-
desorption with acetic acid/acetonitrile, and analysis ization, avoiding hazardous solvents, and minimiz-
with HPLC and GF-AAS. In this procedure, specific ing the number of steps, solid phase microextraction
organotin compounds are used for calibration. How- methods have been developed in combination with a
ever, this method is not suitable for volatile tetraethyl- derivatization reaction for total element determination
tin and tetramethyltin. (Kaur et al., 2006; Centineo et al., 2004). The derivatized
Preconcentration may involve hot trapping or ion species are moved into the vapor phase, therefore mini-
exchange materials combined with an extraction or mizing interference from the sample matrix. The use of
derivatization step. Organometallic species in envi- tetraethylborate and solid phase microextraction, cou-
ronmental samples are mostly ionic and nonvola- pled with GC, were recommended for the speciation
tile. For organotin speciation analysis, especially for analysis of methyltin (Morcillo et al., 1995; Cui et al.,
GC separation, sample derivatization is crucial for 2011). NaBPr4 has been used for the detection of tribu-
organotin extraction from the original matrix into non- tyltin in sediments by solid phase microextraction com-
polar medium to facilitate preconcentration and subse- bined with isotope dilution GC-MS (Bancon-Montigny
quently provide volatile thermally stable compounds et al., 2002). Butyltin and tetramethyltin compounds
for separation (Centineo et al., 2004; Penalver et al., were detected by headspace solid phase microextrac-
2011). Alkylation of organotin salts for subsequent tion after derivatization with NaBEt4 combined with
56 Tin 1247

either GC-ion trap MS-MS, GC-PFPD, or capillary GC- extraction, separation, and analysis of various inor-
coupled microwave-induced plasma atomic emission ganic and organic tin compounds.
detection (Azenha and Vasconcelos, 2002; Carvalho
et al., 2007; Zachariadis and Rosenberg, 2009b; Le Gac 3  PRODUCTION AND USES
et al., 2003; Tzollas and Zachariadis, 2010). However,
despite the critical need for derivatization in GC, there
3.1 Production
is always risk of NaBEt4 interaction with components
of the sample and of producing by-product deriva- Tin is a relatively rare element, with an abundance
tives, which increases the complexity of their deter- in the Earth’s crust of about 2.2 parts per million or
mination. Molecular imprinted solid phase extraction 2.2 mg/kg, which accounts for approximately 0.00022%
was developed for organotin compounds (Puri et al., of the total mass of the Earth’s crust. Tin does not occur
2004; Gallego-Gallegos et al., 2006). Rahmi et al. (2010) as a free metal in nature. The major mineral source of
developed syringe-based solid phase microextraction, tin is cassiterite (SnO2) ore. However, small quantities
named “tip-in chelating monolith,” for trace elements of tin are recovered from sulfide minerals containing
including tin in natural waters. The “tip-in chelat- tin such as stannite (Cu2S-FeS-SnS2), cylindrite (Pb3Sn4
ing monolith” was prepared within a commercially FeSb2S14), frankeite (Fe(Pb,Sn)6Sn2Sb2S14), and teallite
available syringe filter tip by polymerization of 22 5% (PbSnS2). The world resources of tin, principally cassit-
glycidyl methacrylate, 7.5% ethylene glycol dimethac- erite, are located in western Africa, southeastern Asia,
rylate, 35% 1-propanol, and 28% 1,4-butanediol, and Australia, Bolivia, Brazil, China, and Russia, whereas
subsequent modification with iminodiacetate solution. at least half of the world’s supply of tin comes from
The elements were detected by ICP-MS, with a detec- Southeast Asia. The world tin reserves are estimated
tion limit for tin of 0.0015 μg/L. Tin speciation has been to be 5.2 million metric tons. In 2009, statistics for the
also performed using polydimethylsiloxane (100 μm) world’s major tin producing countries were estimated
as a preconcentration polymer. The thickness of the as China (44%), Indonesia (21%), Peru (14%), Bolivia
polymer coating plays an important role, since the (7%), and Brazil (5%). In response to higher tin prices
thicker the film employed, the slower the absorption in 2011, tin producers opened new tin mines and tin
process and the higher the desorption temperature is smelters and expanded the existing operations, includ-
required. ing those in Australia, Bolivia, Canada, and Thailand.
The point of primary importance is the validation The total world mine production of tin reached 265,000
of new or modified methods before their application. tons in 2010 and slightly decreased to 253,000 tons in
The sampling procedures and methods of tin analy- 2011 (USGS, 2010).
sis have been implemented by legislative acts of the The largest producers of recycled tin are France and
European Union No. 1881 and No. 333 (European the United States. According to the most recent sur-
Commission, 2006a, 2007) for the official control of vey conducted by ITRL Ltd (2011), global refined tin
the levels of tin. These legislative acts require valida- consumption was about 350,000 tons in 2011, whereas
tion of the methods through analysis of certified refer- about 12,500 tons of tin was recovered from second-
ence materials such as marine sediment, river water, ary (old and new scrap) metal. In 2010, about 16% of
and certified biological reference materials to prevent tin consumed in the United States was recovered from
scattering of values. This also allows the comparison secondary metal.
of data obtained from different sources. Furthermore, On 4 January, 2011, the “Reduction of Lead in Drink-
validation of the sample treatment step for specifica- ing Water Act” (Public Law 111-380) was passed by the
tion of tin species is crucial. A major problem is the U.S. Congress. The law lowers the national standard
insufficient number of matrices that match certified for lead in faucets, pipe fittings, and pipes from 8.0% to
reference materials. For tin species, available matri- 0.25%, which may lead to a greater use of tin in plumb-
ces are freshwater sediment (BCR 646), certified for ing fixtures (Plumbing Manufacturers International,
mono-, di- and tributyltin compounds, mono- and 2011). ITRL Ltd (2011) estimated that the global tin
diphenyltins; costal sediment (BCR 462), certified to demand in 2015 would be about 400,000 metric tons
dibutyl-, tributyl-, and triphenyltins; and harbor sedi- per year.
ment certified reference material (PACS-2) and mussel Organometallic tin compounds have been increas-
tissue (ERM-CE477), which were certified for mono-, ingly produced since 1950, reaching 50,000 tonnes
di-, and tributyltin compounds, as well as the recently pumped by the organotin industry each year between
added sewage sludge quality control material (Dietz 1990 and 2003 (Fent, 1996). Twenty-five organotin
et al., 2007; Zuliani et al., 2012). However, new meth- compounds are presently produced and used to any
ods still need to be developed for the quantitative great extent (Laughlin and Linden, 1985). Since the
1248 Elena A. Ostrakhovitch

ban on tributyltin-based, dibutyltin, and dioctyltin for- toothpastes because of its long-lasting inhibitory effect
mulations, the production of organotins has gradually on oral microflora, and SnCl2 in soft drinks. Inorganic
decreased. A major commercial application of organo- tin compounds are also used in the ceramic and glass
tin compounds is in the production of polyvinyl chlo- industries.
ride (PVC) plastic stabilizers. In Europe, the highest Organometallic tin compounds comprise mono-,
use of organotin stabilizers is in calendered film (10,000 di-, tri-, and tetrabutyl and triphenyl tin compounds.
tons) and sheet and profile (1500 tons), according to They are widely used as stabilizers for PVC (76%),
the European Council of Vinyl Manufacturers. In 2009, antifouling biocides (10%), agricultural biocides (8%),
8% of stabilizer production in the EU was organotin- and catalysts for the production of polyurethanes and
based (over 10,000 tons). However, dibutyltins will be silicones (5%). These uses account for approximately
prohibited by 2015 for use in soft PVC profiles, coated 20,000 tons of tin consumption per year. Mono- and
fabrics for outdoor use, outdoor pipes, and roofing dialkyltin compounds (RSnL3 and R2SnL2) are well-
materials. The use of dioctyltins has been restricted established PVC stabilizers that have been used to
since January 2012 in skin contact textiles and wall and reduce polymer backbone degradation and inhibit
floor coverings. heat- and light-induced deterioration of plastic prod-
ucts since the mid-twentieth century. There are three
major types of tin stabilizers: octyltin, butyltin, and
3.2 Uses
methyltin. In chemical synthesis, the organotins (pre-
Industrial applications of inorganic tin chemicals dominantly monobutyltin, dibutyltin, and dioctyltin)
are classified as direct or indirect. Indirect applications are used for esterification and transesterification of
include tin salts used as electrolytes for tinplate and tin mono- and polyesters. Alkyltin chlorides are the main
compounds used as intermediates in the manufacture industrial precursors of commercial organotin prod-
of other compounds. Stannous chloride is the most ucts. The major commercial application of organotin
commercially used inorganic tin compound in organic compounds is as stabilizers of rigid PVC to prevent
and inorganic synthesis and tin galvanizing. The major photochemical and thermal transformation (Kizlink,
uses of tin in 2003 were cans and containers, 27%; elec- 1996). Organotin stabilizers are mostly a mixture of
trical, 23%; construction, 10%; and others, 40% (Carlin, mono- and dialkyltins, ranging from 10 to 80% mono-
2004). Tin is used in tin-plated food cans and in aerosol alkyl. The main consumer uses of octyltin stabilizers
containers for protection against corrosion of the base are in the production of rigid packing materials such
metal. Tin alloys cover a wide range of compositions as bottles and pots for food and cosmetics. Dibutyltin
and many applications. Solder, an alloy of 63% tin and stabilizers were used in soft PVC profiles, coated fab-
lead, is primarily used for electronic applications and rics for outdoor use, and outdoor pipes and roofing
has consistently accounted for more than 50% of the tin materials, whereas dioctyltins were applied in textiles,
end-use market since 2005. Lead-free tin solders, con- and wall and floor coverings. However, the European
taining up to 5% silver or antimony, are used at high Commission banned dibutyltin compounds in mix-
temperatures. Tin alloys, including babbit (containing tures, and dioctyltin compounds from 2012. In 2008,
copper, antimony, tin and lead), brasses and bronzes Environment Canada implemented regulations to
(essentially tin-copper alloys), pewter (containing tin, improve management practice guidelines to minimize
1-8% bismuth, and 0.5-3% copper), are widely used the releases of organotin stabilizers. The compounds
(Bulten, 1991). Tin is also an important component have been used in PVC fabric coatings, shoe insoles,
of dental amalgams (silver, tin, and mercury alloy), and antimicrobial finishes for socks and sport clothes.
which has been used for centuries. However, today’s Monooctyltin stabilizers offer excellent transparency
dental alloys exclude mercury and consist of silver, tin, for rigid PVC, with almost no discoloration. A methylt-
copper, indium, palladium, and zinc. Titanium alloys invinyl stabilizer was designed for demanding foam
are used in aircraft components, whereas tin alloys extrusion applications, in particular rigid vinyl pipes.
with zinc, nickel, or copper are used in superconduct- Monoorganotins (predominantly monobutyltin) and
ing cables and magnets. Indium-tin oxide is used in diorganotins are used on glass containers.
spectroelectrochemistry, metallic photonic crystals and Trisubstituted organotin compounds (R3SnL) have
liquid crystal display, organic light-emitting diodes, biocidal properties, whereas monobutyl- and dibutyl-
and photovoltaic and other applications. Direct appli- tins do not exhibit these properties. Trimethyltins are
cations include the use of mixed tin oxide-metal oxide highly insecticidal, and the tripropyl-, tributyl-, and
systems as pigments. Furthermore, SnO2 is used in the tripentyltin compounds have high fungicide and bac-
manufacture of alabaster glass and enamels, dyeing tericide activities. Among the trisubstituted organo-
fabrics, and varnish, SnS2 in pseudogilding, SnF2 in tin compounds, the most important are tributyltin,
56 Tin 1249

triphenyltin, and tricyclohexyltin compounds. Tribu- contained tin at concentrations as high as 242 mg/kg,
tyltin was used from the 1960s onward as a biocide anti- with the highest concentration in the laundry area,
fouling agent in paints for underwater structures and suggesting occupational contamination of the cloth-
on the hulls of ships to prevent the growth of barnacles, ing (Rinehart and Yanagisawa, 1993). Organotins were
seaweed, and other organisms (Granmo et al., 2002). found in house dust at concentrations from 390 to
These antifouling paints contained approximately 20% 28,000 ng/g (Kannan et al., 2010). The relative abun-
(w/w) tributyltin, and released about 4 μg/cm2/day dances of organotin compounds in house dust were
tributyltin into the surrounding water (Matthias et al., ranked in the order:
1988; Swennen et al., 1997). The application of tribu-
monobutyltin → monooctyltin → dibutyltin → dioctyl-
tyltin in antifouling paints was restricted throughout
tin → tributyltin
the 1980s and early 1990s in many countries around
the world due to their high toxicity to aquatic organ- with monobutyltin accounting for 50% of the total
isms (Okoro et al., 2011). The International Maritime organotin concentration. The abundance of monobutyl-
Organization proposed a worldwide ban on tributyl- tin suggests that a major source of organotin compounds
tin-containing paints, commencing with a ban on its in household dust is heat- and light-stabilized PVC
application on vessels in 2003 and a total ban on its products. Concentrations of total organotin species var-
presence on vessel hulls by 2008 (Gipperth, 2009). The ied widely from 140 to 780,000 ng/g in wallpapers and
European Commission banned the use of trisubstituted from 3700 to 120,000 ng/g in floor tiles. The calculated
organic tin compounds in 2010. Tributyltin and triphe- maximum daily intake of organotin in children through
nyltin compounds cannot be used after 1 July, 2010, on dust ingestion was 186 ng/kg body weight (b.w.), which
articles where its concentration is greater than 0.1% by is below the tolerable daily intake of 250 ng/kg b.w. per
weight of tin (European Commission, 2010). The ban day. However, dietary intake combined with intake
on the use of organotin compounds as antifoulants and through the dust digestion could reach the tolerable
biocides worldwide has resulted in reduced release of daily intake value. Additional sources of tin include
these compounds into the environment. The measures mining, industrial production (foundries, smelters,
taken so far have reduced imposex (a marine snail oil refineries, petrochemical plants, pesticide produc-
disease) indices in the majority of location monitored. tion, chemical industry), untreated sewage sludge, and
However, the release of organotin compounds to water diverse sources such as PVC piping, traffic, and com-
continue as a result of redistribution and resuspension bustion by-products from coal-burning power stations.
of organotins through the dumping of contaminated An increasing global problem is the management of
sediments from docks removing contaminated coat- electronic waste (e-waste), particularly the disposal of
ings from ships. old computers and mobile phones (UNEP, 2006).
Other applications include tributyltin and triphe- Inorganic tin(II) can be methylated in the environ-
nyltin use as industrial disinfectants in circulating ment in the presence of methyl donors such as methyl-
industrial cooling waters, for controlling slime in cobalamine (Dizikes et al., 1978; Ashby and Craig, 1991;
paper mills, as a slimicide in masonry, as a biocide Craig and Rapsomanikis, 1985; Chen et al., 2006). Both
disinfectant for textile and leather processing, and in estuary microorganisms and marsh grass exhibited the
industrial wood treatment, paper mills, and the brew- ability to convert inorganic tin into methylated coun-
ing industry (Antizar-Ladislao, 2008). However, these terparts (Hallas et al., 1982; Falke and Weber, 1994a).
uses are minor and are declining. Triorganotins such Guard et al. (1981) also demonstrated that trimethyl-
as triphenyltin hydroxide, tricyclohexyltin hydroxide, tin compounds can be further methylated in estuarine
tricyclohexyltin triazole, trineophenyltin oxide, and sediments. Methylation of tin(II) in the presence of
triphenyltin acetate are used as agricultural fungicides organic methyl donors occurs through oxidative trans-
and acaricides (Isensee et al., 1994). fer of carbonium ethyl to yield monomethyltin, with
the highest yield obtained at pH 5 (Chen et al., 2012).
However, monomethyltin transforms to dimethyltin
4  ENVIRONMENTAL LEVELS AND
as the pH increases. In aqueous solutions with low
EXPOSURES
acidity, tin transforms into dimethyltin, as well as tri-
methyltin as the minor product.
4.1  General Environment
The widespread use of organotin antifouling paints
Tin is present in water, sediments, soils, dust and and biocides has introduced high levels of organo-
air. The presence of inorganic tin and organotin stabi- tin compounds into the aquatic system. Organotins
lizers was also documented in household dust. Dust are hydrophobic and only slightly soluble in water.
in homes of men employed as electric cable splicers Approximately 95% of tributyltin in water was found
1250 Elena A. Ostrakhovitch

to be bound to suspended particles and the remainder pH of the content, storage temperature and time, and
was associated with dissolved organic matter and both the presence of nitrates used as fertilizers (Blunden
organic and inorganic ligands (Gadd, 2000; Fent, 1996). and Wallace, 2003). Exposure of the general popula-
Sorption of organotin compounds onto particles, tion to tin is essentially dietary in origin, deriving
with sorption coefficients ranging from 100 to 10,000, particularly from the consumption of food stored in
is mostly followed by sedimentation (Sanderson cans. This may provoke gastric irritation in certain
et al., 2002; Weidenhaupt et al., 1997; Stewart and susceptible groups. European Commission regulation
Thompson, 1997; Stewart and Demora, 1992). Conse- No. 242/2004 (­ European Commission, 2004) intro-
quently, trisubstituted organotin compounds and their duces limits for inorganic tin in canned foods, canned
degradation products such as di- and monoorganotins beverages, and canned infant foods. For canned solid
accumulate in marine sediments. The aqueous degra- foods, the maximum levels are set at 200  mg/kg
dation rates of triorganotin are faster than those found and for canned beverages at 100 mg/kg (European
in aerobic sediments, and faster again than those in ­Commission 2006a; JECFA, 2011). The acceptable level
anaerobic sediments. of tin in cooked chopped meat, cooked ham, corned
Since the ban on the use of organotin com- beef, and luncheon meat is 50 mg/kg. For inorganic tin
pounds in the antifouling paint was introduced in in canned foods and canned beverages for children,
2008, remobilization of sediments with a high con- the maximum permissible level is 50 mg/kg of wet
tent of organotin compounds is the main source of weight, as covered by Directive 2003/13/EC, the latest
­contamination. Organotin toxicity is highest for trisub- amendments to Regulation (EC) No 466/2001. Guid-
stituted species, and the ecotoxicity increases in order: ance on food contaminants, including inorganic tin,
tributyltin → dibutyltin → monobutyltin. The high con- was published by the Food Standards Agency in 2009
centrations of organotins are extremely harmful to the in conjunction with the pertinent EU legislation (FSA,
marine environment and have been linked to mass 2009b; European Commission, 2006a, 2009).
mortalities of marine mammals, reproductive failure Tin in canned food is likely to be in the cationic form
related to imposex, and population decline in gastro- or in the form of inorganic tin salts in +2 and +4 oxida-
pods, mussels, oysters, and other species (Horiguchi tion states (stannous and stannic compounds), rather
et al., 2001a,b). Marine mollusks can accumulate up to than in covalently bound organometallic compounds.
5 μg/g tributyltin (Hoch, 2001). Crustaceans and fish The tin content of canned foods varies according to
accumulate much lower amounts of organotin due to whether the can is lacquered, the pH of the food in
their ability to degrade tributyltin. The no observed the can, the presence of plant pigments, the storage
effect level (NOEL) for oyster spat is about 20 ng/L, conditions (i.e. time and temperature) of the canned
whereas the development of imposex is associated foods, whether the food is stored in opened cans, and
with NOEL values as low as 1.5 ng/L (WHO, 1990). the presence of oxygen, reducible organic compounds,
Organic tin compounds accumulate in the tissue of and food additives. The most obvious influence on
filter-feeding zooplankton, grazing vertebrates, fish, internal corrosion in plain tinplate cans is the chemis-
water birds, and mammals (Berge et al., 2004; Borghi try of the food product. It should be noted that fruits
and Porte, 2002; Harino et al., 2000; Ohji et al., 2007). and vegetables (e.g. tomatoes) will have significant
Organotin compounds were reported to negatively natural variation in pH and acid type and concentra-
affect the immune system of fish and seals (Allner tion, depending on the variety, maturity, time, place,
et al., 2010; Frouin et al., 2008). and conditions of harvest, soil chemistry, and agri-
cultural practices. Acidic foods are more damaging to
the tin coating in metal cans, and canned acidic foods
4.1.1  Food and Daily Intake
have higher tin contents (Blunden and Wallace, 2003).
Food is the main source of tin in humans. The Tomato-based products tend to have high levels of tin
amounts found in fresh meat, cereals, and vegetables because the presence of nitrates in the food acceler-
depend on the concentration of tin in the soil of the ates corrosion of the tin. Tin may be present in food
area in which the food is produced. Stannous chloride stored in incorrectly manufactured tins (i.e. when the
(SnCl2) is a permitted food additive (E512). Further- alloy layer is not compact and continuous), causing tin
more, tin is present in some multivitamin and mineral present in the interior coating of the can to leach into
supplements at levels up to 0.01 mg in the daily recom- the foodstuff. During food and beverage packaging in
mended dose. Larger amounts of tin may be found in tinplate cans, dissolution of tin into the food may occur
foods stored in plain cans and, occasionally, in foods (Boogaard et al., 2003; Shimbo et al., 2007). The addi-
stored in lacquered cans and drink cans. Canned food tion of essential onion oil to a tinplate can filled with
contains more or less dissolved tin, depending on the tomato puree (pH 4.3-4) prevents the corrosion process
56 Tin 1251

and leads to a significant decrease in the tin content data from the UK National Food survey (MAFF, 1998;
to 28 mg/kg, compared to 223 mg/kg in the absence FSA, 2009a). An estimate of the daily intake of tin by
of onion oil (Nincevic Grassino et al., 2009; Grassino French consumers was 2.32 mg in 2003; this was sig-
et al., 2010). However, essential onion oil stimulates the nificantly below the tolerable limits (Noel et al., 2003).
corrosion of iron, which results in an increase in iron In 2006, dietary exposure to tin was estimated for UK
content from 15 to 46 mg/kg in the canned food. The consumers: the concentration of tin was below the
reported concentration of tin in foods is also affected limit of detection in most food groups (e.g. bread, fish,
by the method of sample preparation and the analyti- and fruit) (Rose et al., 2010). Canned foods contained
cal method used. higher concentrations of tin, presumably as a result of
A provisional maximum tolerable daily intake for slow dissolution of the tin coating. Tin concentrations
tin of 2 mg/kg b.w. and a provisional tolerable weekly in canned vegetables and fruits were 36.1 mg/kg and
intake (PTWI) for tin of 14 mg/kg b.w. (expressed as 11.1 mg/kg, respectively. The concentration of tin in
Sn), equivalent to 12.0 mg/day for a 60 kg adult, were the canned vegetables group was found to be higher
determined by the Joint FAO/WHO Expert Commit- than the 2000 total diet study result (25.06 mg/kg) but
tee on Food Additives (JECFA, 2001). Only limited lower than the value reported for the 1997 total diet
data are available on the toxicological effects of inor- study survey (44 mg/kg) (FSA, 2002). The population
ganic tin present in canned food resulting from disso- dietary exposure was estimated to be 1.80-1.81 mg per
lution of the tin coating. The sixty-fourth JECFA (2006) day. The highest estimated daily dietary exposure to
concluded that the available data indicate that it is tin was for high-level consumers aged 1.5-4.5 years
inappropriate to establish an acute reference dose for (341.5 mg/kg b.w.). Although the daily intake of tin in
inorganic tin because irritation of the gastrointestinal this group exceeded the guidance level set by Expert
tract depends on the concentration and nature of tin in Group on Vitamins and Minerals (220 μg/kg b.w. per
the product, rather than on the dose ingested on a body day), the UK Government’s Scientific Committee on
weight basis. Short-term effects may occur in some Toxicity of Chemicals in Food, Consumer Products
individuals at concentrations greater than 150 mg/ and the Environment (COT) concluded that the small
kg in beverages and over 250 mg/kg in canned food, excess over the guidance level is within the area of
resulting in irritation of the mucosa of the gastroin- uncertainty and that the estimated dietary exposures
testinal tract, which may lead to vomiting, diarrhea, to tin were unlikely to be of toxicological concern
anorexia, depression, ataxia, and muscular weakness. (FSA, 2003; COT, 2008). In Japan, the daily intake of
Organic tin compounds, which differ considerably tin for children was estimated at 162 μg/day, and for
from inorganic tin with respect to toxicity, are consid- adults at 152 μg/day (Aung et al., 2006). However,
ered separately (JECFA, 1978). Based on limited data, when the daily intake was corrected for body weight,
preliminary short-term intakes of inorganic tin were the weekly intake for children was calculated as 61 μg/
estimated to be between 0.004 and 3.3 mg/kg b.w. per kg b.w., whereas for adults the weekly intake was
day, depending on the food considered (JECFA, 2006). around 19.8 μg/kg b.w. For both children and adults,
The total content of tin in canned pear nectar, the intake on a body weight basis was far below the
mango juice, and orange juice was below 70 mg/kg, PTWI of 14 mg/kg b.w. per week (JECFA, 1989). The
whereas the tin concentration was higher in pineap- study conducted by Shimbo and coauthors elucidated
ple juice (77.8 mg/kg) and over 100 mg/kg in canned the possible effect of canned food on tin consump-
fish, mango slices, fruit cocktail, fruit juice, and tomato tion between 1999 and 2004 in Japan. The intake of
puree (Manzoori et al., 2006; Boutakhrit et al., 2011). tin by people eating canned food was 35.7 μg per day,
The content of tin did not exceed the maximum limit eight times higher than the level of 4.5 μg per day for
for tin in canned food for adults. However, canned noncanned food consumers as measured by ICP-MS
pineapple juice and fish cannot be used for consump- (Shimbo et al., 2007). The distribution of tin varied, but
tion by children. The Sn content in the majority of the highest detected value was 2291 μg per day. Intake
samples of canned juices and fruits was high, in par- tended to be high among people consuming canned
ticular in canned papaya (269.8 mg/L) and apricot food such as peach, pineapple, clementines, tuna
(153.4 mg/L) juices (Boa Morte et al., 2009). The levels flakes, or mackerel. The tin intake by the adult popu-
of tin in canned rainbow trout and anchovies produced lation living in Tarragona County (Catalonia, Spain),
in Turkey were shown to average 0.023 and 0.14 mg/ as estimated by the duplicate method, was 69.2 μg/
kg, respectively (Mol, 2011). The estimated maximum day in 2010, which was two times higher than the
intake of tin in 1994 was in the range of 2.4-7.9 mg/ value found in 2003 (Domingo et al., 2012; Bocio et al.,
day, whereas intakes of tin were estimated to be 1.4 2005). In 2006, an estimated dietary intake of 37.9 μg/
mg/kg in 2000 and 1.8 in 2006 based on consumption day for an adult man of 70 kg was observed in various
1252 Elena A. Ostrakhovitch

localities near to a hazardous waste incinerator in concentration in fish collected from the market dur-
­Tarragona County (Marti-Cid et al., 2009). The daily ing 2005 ranged between 1.1 and 23 ng/g wet weight
intake was calculated as the sum of the tin concentra- (Guerin et al., 2007). In Portugal, the tributyltin con-
tion in each food item (such as meat, fish and seafood, centration in shellfish was in the range of 43.2-317 ng/g
cereals, vegetables, tubers, fruits, sugar, eggs, and wet weight (Santos et al., 2009). The estimated average
dairy products) multiplied by the weight of that food daily intake of organotin compounds from the Finnish
consumed by an average individual. Cereals were the market basket for the period 2000-2002 was determined
food group that made the highest contribution to tin to be 5.95 ng/kg b.w., 81% of which originated from
intake. These studies show that there is a trend toward fish (Rantakokko et al., 2006). The total daily intake of
an increased daily consumption of tin. monobutyltin, dibutyltin, and tributyltin was calcu-
Organotin compounds reach humans primarily lated to be 3.39, 0.61, and 1.30 ng/kg b.w., respectively.
through the diet, in particular through contaminated However, for consumers eating large quantities of fish,
water, meat, and fish products. Of about 100 existing the intake may be much higher. Imported Norwegian
organostannic compounds, mono-, di-, and tributyltin salmon contained a few ng/g fresh weight of tributyl-
and mono-, di-, and triphenyltin are those most fre- tin, which was below the limit of quantification. How-
quently found in fishery products. These compounds ever, in Finnish coastal areas with a high density of
are widely present throughout the aquatic environ- shipping, the level of organotins (mostly trisubstituted
ment, resulting in the accumulation of organotins in species) reaches several hundred ng/g fresh weight in
aquatic organisms and eventually in aquatic food costal fish such as pike, perch, and turbot. The content
products such as fish, oysters, and crabs. Octyltins are of organotin compounds in Baltic cod was within the
not detected in fishery products. A scientific panel has range of recommended permissible limits in force in
assessed the health risks to consumers associated with the European Union (European Union Commission,
exposure to organotin compounds in food products 2006a). The concentrations of tributyltin and triphen-
and the tolerable daily intake for the sum of tributyltin, yltin species were above the limit of quantitation: they
dibutyltin, triphenyltin, and di-n-octyltin was accepted were 1.10 μg Sn/kg and 3.21 μg Sn/kg in cod and her-
as 250 ng/kg b.w. by the World Health Organization ring, respectively. Organotin daily intake through fish
(WHO, 1999) and the European Food Safety Author- consumption in 2005-2007 decreased to 3.2 ng/kg b.w.,
ity (EFSA, 2004b). The PTWI of organotins (as the sum which is 1.3% of the tolerable daily intake (Airaksinen
of dibutyltin, tributyltin, triphenyltin, and dioctylodi- et al., 2010). In total, fish (in particular, perch, salmon,
etylotin) is 0.7 μg Sn/kg b.w., which, for an individual and rainbow trout) accounted for over 60% of the total
weighing 70 kg, is 49.0 μg Sn per week (EFSA, 2004a). organotin intake. These data indicate that the fish
The USEPA has established an oral reference dose of analyzed (especially Baltic salmon) do not pose a risk
300 ng/kg b.w. per day (USEPA, 1997). to consumer health as far as the presence of organo-
In Japan in the early 1990s, the daily intakes of tri- tin compounds is concerned. Because tributyltin was
butyltin and triphenyltin from a market food basket a common ingredient in antifouling paint and is still
were on average 6.8 and 3.3 μg per person, respectively. present in sediments in the Bay of Islands (Northland,
Seafood accounted for more than 95% of the intake of New Zealand), it was expected that trace amounts of
organotins (Tsuda et al., 1995). By the late 1990s, daily tin might still be detectable in shellfish (Coelho et al.,
intakes had decreased and were 1.7 μg for tributyl- 2002). However, as a result of restriction in 2003 and
tin, 0.09 μg for triphenyltin, and 0.45 μg for dibutyltin complete prohibition in 2008 on the use of organotin-
(Toyoda et al., 2000). The tributyltin concentration containing antifouling agents on ships, tin was not
measured in 11 species of fish from the port of Osaka detectable in mussels from this area; thus, that there is
and the Yodo River was in the range of 11-182 ng/g no risk to shellfish consumers among the general pub-
wet weight (Harino, et al., 2000). The estimated daily lic, and in particular, the Maori population and Pacific
intake of butyltin compounds from seafood consump- Island peoples whose diet is based primarily on fish
tion for the general population in Korea was assessed and seafood (Whyte et al., 2009). The levels of mono-,
as 17.2 ng/kg b.w. per day (Choi et al., 2012). Among di-, and tri butyltin compounds were significantly
the seafood groups, fish contributed to the highest lower than the tolerable level of 100 ng/g wet weight
intake of tributyltins (8.66 ng/kg b.w./day), followed in mussels and frequently consumed fish species from
by cephalopods (4.39 ng/kg b.w./day). High concen- Iranian coastal waters of the Caspian Sea (Rastkari
trations of butyltins were found in shellfish. How- et al., 2012). In China, high concentrations of butyltins
ever, the contribution of shellfish to the total butyltin (10-200 ng Sn/g) were detected in beans, vegetables,
intake was low (18%) because of the low consump- fruits, eggs, milk, sugar, meat, potatoes, and bever-
tion rate of shellfish (3.9 g/day). In France, tributyltin ages (Qunfang et al., 2004). In Brazil, the application of
56 Tin 1253

triphenyltin during rice sowing was shown to result in highest concentrations of organotin species. Organo-
the accumulation of triphenyltin and its degradation tin compounds are hydrophobic substances and only
products such as diphenyltin and monophenyltin in slightly soluble in water. Tributyltin is generally lipo-
the roots (Antes et al., 2011). However, the concentra- philic in character, and has a high specific gravity and
tion of these compounds in rice grains was lower than low solubility in seawater (less than 10 mg/L at 20°C
the detection limits. and pH 7.0) (Fent, 1996). It was suggested that the high
Furthermore, dietary exposure to organotin com- concentrations of tributyltins in seawater result from
pounds may result from the backing on silicon-backed their release from sediments, which act as a long-term
parchment, certain brands of gloves, sponges, sanitary source of dissolved-phase contamination in marine
napkins, food storage in containers made from PVC environments (Ritsema et al., 1998). Tributyltin is
polymers, and some brands of vine stored in PVC bot- reported to be highly adsorbed in sediments with fine
tles or barrels (Takahashi et al., 1999). Organotin species texture and abundant organic matter (Burton et al.,
also leach into water through PVC pipes (Sadiki and 2004). Organotins are relatively stable under anoxic
Williams, 1999); contamination levels of 291 ng/L for conditions in sediments: they remain in sediments for
monomethyltin, 49.1 ng/L for dimethyltin, 28.5 ng/L many years and slowly leach into surrounding water.
for monobutyltin, and 52.3 ng/L dimethyltin were The half-life of tributyltin was calculated to be 6 days
detected. The highest levels of monomethyltin, in summer for turbid water and up to 127 days in
dimethyltin, monobutyltin, and dibutyltin were winter for nonturbid water; the half-life is much lon-
detected in PVC pipes subjected to deionized water ger in sediments, and has been estimated to last sev-
exposure during the first 1-5 days (Impellitteri et al., eral years (Alzieu, 2000, 2006). Tributyltin degrades
2007). Monobutyltin was the most prevalent kind of to dibutyltin and monobutyltin species in the aquatic
organotin. The concentrations of organotin were 0.4- system via dealkylation by UV photolysis and micro-
2.4 μg/L for monobutyltin, 0.2-1.0 μg/L for dimethyl- bial degradation. Biodegradation of tributyltin is faster
tin, and 0.04-0.01 μg/L for tributyltin. A similar pattern in organic-rich sediments than in sandy sediments
of release from PVC pipes was found for dimethyltin with little organic material (Landmeyer et al., 2004).
and dibutyltin, with an initial rapid increase in con- Furthermore, the toxicity of tributyltin in salt water
centration (Adams et al., 2011). The diffusion coeffi- is less than in freshwater due to reduced microbial
cient for organotin in PVC pipe material was found degradation. The retention of trisubstituted organotin
to be 9 × 10−18 m2/s. Butyltin species were detected compounds depends on temperature, pH, and light.
in vinegar stored in plastic bags: the concentration of The retention time of tributyltin is approximately 2.1
tributyltins ranged from 0.012 to 14.1 μg (Sn)/L (Cui years, whereas the retention times of dibutyl- and
et al., 2012). monobutyltin are 1.9 and 1.1 years, respectively
(Sarradin et al., 1995). The half-life of methyltin and
butyltin compounds was estimated to be in the range
4.1.2  Water, Soil, and Air
of 0.5-15 years in organic and mineral forest soils
Tin compounds occur naturally in the soil and water. (Huang and Matzner, 2004a,b). Degradation rates
Trace quantities of tin can be measured in air. Compo- were: monosubstituted → ­disubstituted  → ­trisubstituted
nents of the soil may be released as dust and the wind organotin compounds. A similar half-life was deter-
may carry airborne particles for long distances before mined for dibutyltin in marine sediments (Almeida
deposition. In the air, tin largely associates with small et al., 2004). The half-life of dibutyltin in acti-
respirable particles 1-3 μm in size (WHO, 1980a). Inor- vated sludge was found to be 5.1 days (Stasinakis
ganic tin is considered to be relatively immobile in et al., 2005).
the environment. However, it may also undergo bio- Despite the global ban on organotin antifouling
methylation in sediments, and redistribution between paints, they are still common contaminants of marine
alkylated species of tin has been observed in decay- and freshwater suspended matter, sediments, biomass,
ing plant materials in the environment (Falke and and coastal organisms around the world. In India,
Weber, 1994a,b; Guard et al., 1981; Hallas, et al., 1982). there has been no legal ban on the use of tributyltin,
A large percentage of tin is present in particulate form and organotin compounds are still used in antifoul-
(­Langston et al., 1987). In soil-plant systems, trans- ing paints (Garg et al., 2010). The total levels of butyl-
port coefficients for inorganic tin were reported to be tins (tri-, di-, and monobutyltins) measured in 2007 in
0.01-0.1 (Kloke et al., 1984). the Mandovi and Zuari estuaries located on the west
Organotin compounds can degrade into inor- coast of India were up to 77 ng/L in water and up to
ganic forms or may remain unchanged for years in 120 ng/g in sediments. The highest concentration of
sediments. The marine biota tends to accumulate the tributyltin (119 ng/g dry weight) was detected in the
1254 Elena A. Ostrakhovitch

Zuari Estuary. High tributyltin pollution was reported The fate of organotin compounds depends on the soil
in sediments in the Bitung ferry port in Manado, Indo- composition, water pH, wetting and drying of the soil,
nesia, where the concentration of tributyltin reached and temperature (Huang and Matzner, 2004a; Hoch
a staggering 250 μg/kg dry weight and 4.25 μg/g wet et al., 2002, 2003; Hoch and Schwesig, 2004). The high-
weight (Harino et al., 2012; Undap et al., 2013a). Rela- est level of contamination by organotin compounds
tively high concentrations of organotin compounds in a freshwater river system was reported in central
were measured in sediments and organisms in many South Carolina in the United States in early 2000 (Land-
areas of the Brazilian coast, particularly in harbors and meyer et al., 2004). The concentrations of tetrabutyltin
shipyards (Liscio et al., 2009; Fernandez et al., 2005). and tributyltin were greater than 10 mg/kg in samples
Organotin pollution was detected in the Bahia Blanca of surface water bed sediments collected in 2000 and
Estuary in southwest Buenos Aires Province, Argen- 2001 in deposition areas. In bed sediments of a pond
tina (Delucchi et al., 2007). Tributyltin concentrations located about 1.6 km from the release of organotins, the
measured in the inner region of the estuary ranged concentration of total organotins reached 40 mg/kg.
from below the detection limit to 170.3 ng/g sedi- Contamination by organotin compounds was reported
ments, whereas the concentration near the dry dock at in surface and ground waters in the Hérault water-
Puerto Belgrano Naval Base was 3288 ng/g. The con- shed (in the Mediterranean Basin) (Bancon-Montigny
centration of dibutyltin was also the highest at Peuto et al., 2008). The total organotin content ranged from
Belgrano, and was estimated to be 1645 ng/g. In the below the detection limit to 0.51 ng/L in 2005; how-
Archipelago Sea in southwestern Finland, the concen- ever, the concentration of tributyltin, and occasionally
tration of tributyltin in sediments was estimated to be of phenyltin, in water was found to be up to 368 ng/L.
42 μg/kg, with higher levels found in the northern part Di- and trisubstituted compounds were prevalent in
(71 μg/g) and reaching 1443 μg/kg in the vicinity of a April, whereas monosubstituted compounds predomi-
repair shipyard (the largest individual source of tribu- nated in July. The degradation of organotin species in
tyltin) (Lilley et al., 2012b; Airaksinen et al., 2010). Stud- the sludge via dealkylation depends on the nature of
ies in chironomid species, which are an important food the parent compound, the concentration, and the par-
source for fish, birds, and bats, showed that increased ticulate phase. A number of studies have reported the
tributyltin concentration in sediment reduced spe- presence of octyltin in sewage sludge from wastewa-
cies diversity but did not markedly affect the number ter treatment. The highest concentration of dioctyl-
of individuals in the Archipelago Sea in southwest- tin reported in sludge was 0.56 mg/kg, whereas the
ern Finland (Lilley et al., 2012a). Similar effects were monooctyltin concentration was 0.715 mg/g (Summer
reported for marine periphyton (Sayer et al., 2006; et al., 2003). Organotin stabilizers in PVC pose a public
Smith et al., 2008). The reduced diversity of the spe- health risk involving the workplace, consumer prod-
cies along with the prevalence of tributyltin-resistant ucts, and the general environment.
species may lead to a transfer of organotin compounds The leaching of organotin compounds from PVC
between the aquatic and terrestrial ecosystems. pipes contributes to the accumulation of organotin
The agricultural and biocidal applications of organo- in the environment. On average, 35 mg/m3 tin were
tin compounds resulted in their accumulation in soil, released into the water from PVC pipes with a length
water, and air through spraying, leaching, and runoff. of 46 m after the first use. Analysis of environmental
Triphenyltin has been excessively used to control fun- samples such as water, sediments, plants, and algae
gal diseases of potato, sugar beets, carrots, onions, and collected in Johannesburg (South Africa) showed high
rice, as well as peanuts, pecans, coffee, and cocoa, and is concentrations of dibutyltin and monobutyltin in com-
currently approved for use as a fungicide (Jones-Lepp parison with tributyltin (Nsengimana et al., 2009). The
et al., 2004). Due to the volatility of triphenyltin, it can lower concentration of tributyltin (22.3 μg/L in surface
be transported long distances from treated fields. Accu- water and 14.6 μg/kg in sediments) was attributed to
mulation of triphenyltin and its degradation products in its degradation. The concentrations of monobutyltin
the soil and underground water poses a serious risk to and dibutyltin in surface water were 25.1 μg/L and
the environment. The environmental Serious Risk Con- 38.7 μg/L, respectively, and reached 39.2 μg/kg and
centrations (SRCs) for soil are 28, 0.052, and 0.24 mg/kg 42.4 μg/kg in sediments. The accumulation of dibu-
dry weight soil for dibutyltin, tributyltin, and triphen- tyltin and monobutyltin compounds was suggested to
yltin, respectively. For groundwater, the SRCs are 50, result from tributyltin degradation and the use of PVC
0.046, and 0.40 μg/L respectively. The maximum allow- materials in the supply of water for mining.
able concentration of tributyltin compounds in inland Tin can be found near to tin mines and is also natu-
surface waters, as proposed by the European Commis- rally present as a result of burning fossil fuels such as
sion, is 0.0015 μg/L (European Commission, 2006b). coal and oil or of burning organic and inorganic waste.
56 Tin 1255

The tin concentration also depends on the extent of 5 METABOLISM


industrial activity. The content of tin-containing par-
ticles in airborne ash from coal-burning power plants 5.1  Inorganic Tin
ranges from 7 to 19 mg/kg. According to the USEPA,
in 1997 more than 3.2 million tons of e-waste com- 5.1.1 Absorption
prising electrical and electronic equipment had been
dumped in U.S. landfills. Today, the estimated amount 5.1.1.1 Inhalation
of e-waste is 20-50 million tons/year worldwide. It Exposure to tin oxide dusts and fumes during fusion
has been estimated that, in the USA alone, over 30 mil- operations, when tin reaches its melting temperature,
lion computers and 100 million cell phones contribute may cause benign pneumoconiosis known as stanno-
more than 10,000 tons of e-waste annually (BAN, 2004; sis. Occupational stannosis was reported in nonmin-
Cobbing, 2008). Printed circuit boards contain metal- ing industries such as grinding, briquette-making, and
lic tin in the form of lead-tin alloy, and plastics, which casting processes. Long-term exposure to tin oxide
constitute more than 20% of e-waste, contain various dust and fumes results in the deposition of particles in
organotin species (Schlummer et al., 2007; IGES, 2009). lung phagocytes and lung tissue due to its insolubility
Due to the crude recycling process, tin may leak from and poor absorption (Robertson et al., 1961; Brown-
the residual ashes to the ground and to aquifers in the ing, 1969; Krigman and Silverman, 1984). Thus, lungs
surroundings (Bi et al., 2010). are the major target organ. Smooth muscle of the
It was reported that after 2 weeks of exposure to bronchiole was shown to remain intact. Chronic expo-
atmospheric fallouts in the courtyard of a secondary sure to tin dust causes benign pneumoconiosis and
lead smelter that recycles car batteries, the content of stannosis, without indications of fibrosis (Robertson
tin during the entire plant exposure period increased et al., 1961). Slius-Cremer and colleagues (1989) ques-
from 1 mg/kg to 4.9 mg/kg in parsley and from 2 mg/ tioned whether abnormalities associated with tin
kg to 13.3 mg/kg in ryegrass, reaching 7.9 mg/kg in exposure could be called pneumoconiosis at all since
parsley and 16.4 mg/kg in ryegrass in 4 weeks (Schreck these abnormalities were not associated with fibrosis
et al., 2012). and respiratory failure. Silicosis and pulmonary heart
disorders had been reported among Chinese workers
in tin mines in the late 1980s (Chen et al., 1992). An
4.2  Working Environment
increase of nearly sixfold in the death rate from pul-
The OSHA (2013) established workplace permis- monary heart disease was observed among workers
sible exposure limits of 0.1 mg/m3 air for organotin exposed to dust in mines. A 50% increase in mortality
compounds, defined as the maximum recommended was associated with nonmalignant respiratory dis-
worksite concentration for a conventional 8-h work- eases caused by respirable silica dust in hard rock tin
day, and 2 mg/m3 for all inorganic tin compounds mines (Chen et al., 2005). However, there was a trend
except oxides. A value of 0.2 mg/m3 was specified as toward increased cancer risk among tin miners. Later
the maximum worksite concentration for short period- findings indicated a positive dose-response relation
exposure by the American Conference of Industrial between exposure to cumulative dust in tin mines
Hygienists (ACGIH), providing that the daily limit and mortality from lung cancer (Chen et al., 2006).
value is not exceeded. NIOSH recommends workplace Some studies reported an increased risk of death from
exposure limits of 2 mg/m3 for inorganic tin com- gastric cancer among tin miners, especially in under-
pounds, except for SnH4, and 0.1 mg/m3 for organo- ground workers (Raj et al., 2003). However, high
tins, except for tricyclohexyltin hydroxide (NIOSH, arsenic, crystalline silica, and radon content in dust
2013). NIOSH states that a tricyclohexyltin hydroxide particles might be a cause of increased mortality from
concentration of 25 mg/m3 should be considered as lung cancer (Chen et al., 2006; Hodgson and Jones,
immediately dangerous to life or health. The current 1990; Raj et al., 2003).
OSHA standard for inorganic tin compounds (as Sn) is Inhalation of tin oxides in molten metal-refining
2 mg/m3 of air. In the Netherlands, the administrative furnaces can lead to the same conditions. Micro-
MAC value is 2 mg/m3 as Sn for tin and inorganic tin scopic analysis revealed dark dust foci in the lung tis-
compounds (Health Council of the Netherlands, 2005). sue of subjects employed in the tin-melting industry.
In Denmark, Finland, Iceland, Norway, the UK, and Recently, Yilmaz et al. (2009) described two cases of
the USA, 2 mg/m3 as Sn is recommended by ACGIH, stannosis associated with exposure to tin fume. Long-
NIOSH, and OSHA as the occupational exposure limit. term tin workers developed stannosis and died as a
In addition, in the UK a short-time value of 4 mg/m3 is result of respiratory failure. The subjects had numer-
available (ACGIH, 2003). ous small dense nodules in both lungs, with dense
1256 Elena A. Ostrakhovitch

nodular lesions. The authors suggested that tin fume of nitrate action, which act as a corrosion accelerator
exposure may not be as benign as commonly believed. (Nincevic Grassino et al., 2009). The addition of essen-
Two cases of pulmonary alveolar proteinosis, includ- tial onion oil as a potential corrosion inhibitor was sug-
ing one death, were reported in workers at a facility gested to avoid leaching of tin (Grassino et al., 2010).
producing indium-tin oxide (ITO), consisting of 90% Studies in animals and human subjects demon-
indium oxide (In2O3) and 10% tin oxide (SnO2), for flat strated that the absorption rate of ingested inorganic
panel displays (Cummings et al., 2010). Both work- tin is approximately 5% in mice, rats, dogs, and mon-
ers were exposed to airborne ITO dust and developed keys, but is influenced by dose and the presence of
pulmonary alveolar proteinosis. However, it was sug- other substances. In humans, absorption rate is 3%
gested that the exposure to indium compounds caused or less from a diet supplemented with 50 mg tin per
pulmonary fibrosis, emphysema, and pneumothoraces day (Johnson and Greger, 1982). Similar to symptoms
(Cummings et al., 2012). in humans, gastrointestinal irritation such as vomit-
ing was observed in cats (but not dogs) receiving 5
5.1.1.2 Ingestion or 10 mL/kg b.w. of an orange juice containing over
Inorganic tin compounds are relatively harmless 450 mg/kg tin by stomach tube. Cats fed orange juice
due to their poor absorption, relative insolubility, and containing 540 mg/kg tin, providing a dose of 5.4 mg/
low retention in the tissue (Johnson and Greger, 1982; kg weight, showed vomiting, and the incidence of
Krigman and Silverman, 1984; Winship, 1988). Several vomiting increased with high doses (Benoy et al.,
cases of gastrointestinal manifestations of tin poison- 1971). Ingestion of stannous chloride (SnCl2) for 4-13
ing have been associated with high concentrations of weeks resulted in gastrointestinal irritation, anemia,
tin in drinks or solid foods. Vomiting, diarrhea, fatigue, and reduced bone strength.
and headaches were recorded after the consumption
of canned orange beverages in 1963. The content of 5.1.2  Skin Contact
tin ranged from 75 to 500 mg/kg. Over 100 cases of tin Inorganic tin has been used in cosmetics for many
poisoning were reported in Washington and Oregon in years, in particular as toothpaste abrasive (tin fluo-
1969 after ingestion of canned tomato juice. The toxic ride) and in nail care (tin oxide). Dermal toxicity of
effect of tin depends on the concentration in the food- tin fluoride and tin oxide was not reported. However,
stuff ingested (WHO, 2001, 2006). Analysis of the juice tin tetrachloride or stannic chloride (SnCl4), stannous
consumed showed a tin content of 477 mg/L. Five chloride (SnCl2), and stannous sulfate (SnSO4) can
adult participants of clinical study were given 240 mL cause skin irritation due to their acidic nature. These
orange juice containing 0, 498, 540, or 1370 mg/kg tin. compounds absorb moisture from the air and become
Nausea and diarrhea were recorded in all participants dissolved in it to form an acidic solution. Contact with
after ingestion of the orange juice with highest tin con- these substances may cause severe burns to the skin
centration, whereas lower concentrations equal to a and eyes. Sodium stannate and potassium stannate are
dose range of 1.6-3.6 mg/kg b.w. inorganic tin did not strong alkalis. Tin salts are skin irritants, especially in
induce any symptoms (Benoy et al., 1971). Ingestion of the form of dusts or mists, because of acidic or alkaline
fruit juice containing 342 mg caused symptoms of gas- reactions. Persons with existing skin disorders may
trointestinal irritation (MAFF, 1983); similar juice con- be more susceptible to the effect of these compounds.
taining between 125 and 182 mg had no effect. Peaches However, only occasional cases of allergic dermatitis
containing 350-600 mg/kg tin and orange and grape- in human have been reported. In rabbits, it has been
fruit juices containing 330-400 mg/kg tin caused gastro- shown that a 1% solution of SnCl2 applied to dermal
intestinal effects (Blunden and Wallace, 2003). The level scratches for 18 h generated a reaction with intraepi-
of tin that may cause gastrointestinal irritation, nausea, dermal pustules (Stone and Willis, 1968).
vomiting, and diarrhea varied from 250 to 2000 mg/kg.
Symptoms started within 0.5-3 h after consumption and 5.1.3 Distribution
recovery occurred within 48 h. Dietary products more
likely to be responsible for gastrointestinal irritation are Tin is present in small amounts in animals and
acidic fruits and fruit juices packed in unlacquered, par- humans in nearly all organs, with a total content of
tially unlacquered, or damaged tinplate cans (Schafer approximately 352 mg in adult men (Kehoe et al.,
and Femfert, 1984; Blunden and Wallace, 2003). The 1940). The highest accumulation of tin was observed
presence of tin at levels up to 250 mg/kg in canned food in the bone and thymus, but also occurs in the lungs,
caused no adverse effects in healthy adults (Boogaard liver, kidney, spleen, lymph nodes, and other organs.
et al., 2003). In cans containing tomato puree and potas- Thus, in tissue samples from adults who died in acci-
sium nitrate, dissolution of tin started after 30 days (at dents the concentration of tin was found to be as high
36°C) and 60 days (at 20°C) of storage as a consequence as 4.1 mg/kg in bone ash, whereas in muscle and
56 Tin 1257

brain tin levels were 0.07 and 0.06 mg/kg wet weight, dosage. The tin concentration in urine samples from
respectively (Hamilton and Minski, 1972). In other miners after a work shift in Brazilian mines was shown
studies, tin concentration in the bone was determined to be 0.45 μg/L, compared to 0.13 μg/L before the shift
to be as high as 6.2 mg/kg and as low as 0.47 mg/kg and 0.08 μg/L in the control group, suggesting that the
(Garcia et al., 2001; Llobet et al., 1998; Tipton and Cook, workers have a daily intake of tin (Juliao et al., 2007). In
1963; Teraoka 1981; Chiba et al., 1990). Tin concentra- human and animals, the concentration of tin is higher
tion in lung samples from deceased subjects without in urine than in hepatic bile (Ishihara and Matsushiro,
known occupational tin exposure varied between 0.24 1986).
and 3.4 mg/kg. In blood, tin concentrations of 2-9 μg/L In laboratory animals, gastrointestinal absorption of
were reported. In subjects occupationally exposed inorganic tin is reported to be extremely low (Ishihara
to tin, tin concentrations were elevated in the lungs, and Matsushiro 1986). Rats and cats given orange juice
spleen, liver, and kidney (Teraoka, 1981). supplying tin at 7-20 mg/kg excreted 99% of tin in
The application of tin fluoride solutions leads to tin feces within 48 h (Benoy, Hooper, and Schneider, 1971).
deposition onto the tooth surface and its incorporation Tin chloride administered at a dose of 50 mg/kg in
into carious enamel and dental hard tissue (Ellingsen, water was mainly (90-99%) excreted in the feces within
1986). The administration of highly concentrated solu- 48 h (Fritsch et al., 1977). Traces of tin were found
tions of tin (2100 mg/kg Sn2+) led to its incorporation to in the urine and in the organs. Tin(II) or tin(IV) given at
a depth of 20 μm; a less-concentrated solution (400 mg/ single oral dose of 20 mg/kg as fluoride or citrate was
kg Sn2+) led to small amounts of tin being incorporated mainly excreted in feces (99%), with less than 1% in the
into the outer 10 μm (Schlueter et al., 2009). urine (Hiles, 1974). After a single intravenous dose of
In mice intraperitoneally injected with stannous tin(II) at 2 mg/kg, 35% of the tin was found in the urine
chloride, tin accumulated in all organs except the tes- and 12% in feces, whereas 40% of tin was excreted in
tis (Chiba et al., 1990). However, the most tin was urine and 3% in the feces after single intravenous dose
retained in bone, followed by muscle, liver, and kidney of tin(IV). However, intravenous injection of mice with
­(Furchner and Drake, 1976). Following a single gavage 1% tin citrate did not result in tin excretion in the feces
dose of 20 mg/kg b.w. of tin(II) or tin(IV) as fluoride or or urine. Observed differences in the excretion routes
citrate, the main deposition of tin was observed in the for tin may be due to the differences in the form of tin,
skeleton (1 and 0.24%, respectively), followed by the the dose, and the administration route. For example,
liver and kidney (Hiles, 1974). A single oral administra- the biliary route was shown to be more important for
tion of tin salt at a dose of 20 mg/kg for 4 weeks resulted tin(II) than for tin(IV) compounds.
in the predominant accumulation of tin in the bone.
Tin concentration in bone tissue of healthy unexposed 5.1.5  Biological Half-Life
mice was reported to be 0.69 mg/kg dry weight, which
The half-life of tin in the femur was estimated to be
drastically increased after tin administration. Lifelong
34-40 days (Hiles, 1974). For liver and kidney, the half-
exposure of mice to stannous chloride (SnCl2) in drink-
life of tin(II) has been estimated as 10-20 days. In later
ing water resulted in tin accumulation in the kidney,
studies, the biological half-life of inorganic tin in rats
liver, heart, lungs, spleen, and thyroid (Schroeder et al.,
was reported to be approximately 85 days in the liver
1968). In pregnant rats fed with stannous chloride, tin
and 50 days in the spleen (Marciniak, 1981). In mice, a
has been found in embryos (Chmielnicka et al., 1981).
biological half-life of approximately 30 days was shown
However, in pregnant rats exposed to tin fluoride, no tin
using a whole-body counting method (Brown et al.,
was detected in fetal or placental tissues (Hiles, 1974).
1977). Complete elimination of tin(II) has been observed
after 90-100 days (Furchner and Drake, 1976). Stannous
5.1.4 Excretion and stannic compounds show differences in blood clear-
ance, excretion, and soft tissue uptake (Srivastava et al.,
Inorganic tin is mainly excreted in the feces (about
1985). Thus, Sn (IV) shows higher bone uptake and less
95-99%), with additional slow elimination in the urine
soft tissue accumulation than Sn (II). Such differences in
(Winship, 1988; WHO, 2001). Bile excretion is less than
cation affinity between Sn (II) and Sn (IV) are attributed
15% (WHO, 2003, 1980a,b). The average tin concentra-
to slow oxidation or reduction during their absorption
tion in the urine was approximately 0.023 mg/L, with
and transport in the organism.
a range of 0-0.004 mg/L (Baselt and Cravey, 1989). Tin
concentration in the urine of adult human males receiv-
5.1.6 Biotransformation
ing food containing 0.11 mg or 50 mg tin per day for 20
days was around 30 μg/L and 120 μg/L, respectively Inorganic tin may undergo oxidation-reduction,
(Johnson and Greger, 1982). Fecal excretion accounted ligand exchange, and methylation reactions. In extremely
for 55% at the low dosage of tin and 97% at the high anaerobic conditions inorganic tin may be converted into
1258 Elena A. Ostrakhovitch

stannane (SnH4) by decaying algal material (Donard and headaches and nausea. In the most severe cases of
Weber, 1988). Biomethylation of inorganic tin has been organotin poisoning, bradycardia, hypotension, and
shown in bacterial cultures, sediments, marsh grass, abrupt variations in the sinus rhythm of the heart were
decaying plant materials, soils, and household and indus- observed. Common clinical symptoms of organotin
trial composts (Braman and Tompkins, 1979; Furuhashi poisoning are headache, dizziness, photophobia, nau-
et al., 2008; Donard et al., 1986; Ashby and Craig, 1988a; sea, and vomiting, with some incidences of temporary
Falke and Weber 1994a; Duester et al., 2005). Methylation loss in consciousness, epileptic seizures, dermatitis,
of tin in sediments positively correlated with increasing and asthenia (Manzo et al., 1981; Lyle, 1958). In 1981, six
organic content in sediments (Hadjispyrou et al., 1998). workers were exposed to a mixture of dimethyltin and
Ashby and Craig (1991) showed that tin(II) can be meth- trimethyltin vapor while cleaning a cauldron. One of
ylated by methylcobalamin under environmentally the workers died and two of surviving workers devel-
relevant conditions; this depends on salinity, pH, and oped neurological problems including memory loss
whether conditions are aerobic or anaerobic (Chen (Rey et al., 1984). Symptoms preceding death included
et al., 2007). In solutions with pH higher than 1.0, respiratory depression and coma associated with high
monomethyltin was produced by carbonium trans- levels of tin in the urine. The excretion rate of total tin
fer and further methylated to dimethyltin by addi- in the urine was reported to correlate with the sever-
tional carbanion transfer. The highest concentrations ity of intoxication by dimethyltin and trimethyltin;
of methylated tin were observed in agricultural soils, patients with an excretion level above 1 mg/L showed
garden soils, and flood plain soils (Duester et al., 2005). severe neurological defects (Rey et al., 1984). Exposure
Concentrations of dimethylated species and mono- to a vapor of dimethyltin and trimethyltin for 3 months
methyltin were between < 0.01-7.6 μg/kg dry mass. resulted in mental confusion and epileptic seizures
Soil parameters such as pH, water content, and tem- in two chemists involved in dimethyltin synthesis
perature did not correlate significantly with the degree (Fortemps et al., 1978). Following a spillage in a plant in
of biomethylation observed. However, it was shown the United States in 1978, 22 chemical workers exposed
that the pH of the reaction solution was highly associ- to trimethyltin chloride developed forgetfulness,
ated with photochemical methylation of tin(II) in the fatigue, depression, and attacks of rage (Ross et al.,
presence of acetone under UV irradiation (Chen et al., 1981). A patient who had been exposed to triphenyltin
2012). The highest yield of methyltin was observed at also showed acute urticarial eruption, signs of hepatic
pH 5.0. Furthermore, pH affected the redox potential injury, and electroencephalographic abnormalities
of tin(II) in aqueous solution. Transformation of stan- (Colosio et al., 1991). In one report, a 27-year-old male
nic chloride in landfills was also documented. Inor- studying chemistry was exposed to trimethyltin chlo-
ganic tin is methylated if methyl donors are present. ride when he measured 300 mg (15 cm3) of a powdered
Monomethyltin trichloride at an initial concentration substance (Saary and House, 2002); he developed head-
of 500 mg Sn/L found in landfills promoted methyla- aches, agitation, shortness of breath, chest discomfort,
tion of inorganic tin present in the inoculum under and abdominal pain. Acute poisoning with dimethyltin
methanogenic conditions (Bjorn et al., 2011): the mono- dichloride was reported in worker after cleaning a tank
methyltin concentration increased by a factor of about used in the manufacture of this organotin. Although
five over the 211-day incubation period. Methylation the man wore personal protective clothes and an air
of inorganic tin compounds also contributes to increas- supply mask, he suffered from dizziness, disorienta-
ing the volatility, toxicity, and mobility of tin in the tion, hallucination, irritability, and decreased memory
environment. after 4 days of exposure (Yoo et al., 2007). The neuro-
logical manifestation was similar to trialkyltin enceph-
alopathy. Brain magnetic resonance imaging revealed
5.2  Organotin Compounds extensive symmetric lesions in the subcortical and deep
5.2.1 Absorption white matter of the bilateral cerebral hemispheres, pos-
terior rim of internal capsule, corpus callosum, cerebral
5.2.1.1 Inhalation peduncle, and middle cerebellar peduncle. Analysis
Cases of intoxication due to occupational exposure showed dimethyltin levels to be as high as 47.6 μg/L in
to triphenyltin acetate have been documented. The blood and 779.4 μg/g Sn in urine 13 days after the ces-
main route of absorption is reported to be the respira- sation of exposure, whereas the trimethyltin concentra-
tory tract. In 1951, Zeman and others (1951) reported tion was as high as 327.5 μg/L in blood and 946.3 μg/g
four cases of employee exposure to unknown con- Sn in urine. A trace amount of urinary dimethyltin was
centrations of tetramethyltin and tetraethyltin. Initial detected on day 27 after discharge from the hospital;
symptoms in all four subjects exposed included severe however, trimethyltin was not detected.
56 Tin 1259

A tributyltin aerosol at a concentration of 20 mg/m3 tributyltin and triphenyltin affected male sexual devel-
was lethal to guinea pigs within 1 h of exposure opment, associated with decreased serum testosterone
(­Schweinfurth and Gunzel, 1987). Similarly, lethality and luteinizing hormone.
was recorded in mice following single or repeated The most toxic organotin compounds are trimethyl-
daily exposure to a mixture of tributyltin (81%) and and triethyltin, which are easily absorbed from the
dibutyltin (3.7%) at a total concentration 5.65 mg/m3 gastrointestinal tract (Winship, 1988). During the 1999
Sn (Igarashi, 1959). Inflammatory changes consisting New Year holiday, over 1000 people in southeast China
of hyperemia and bronchitis were observed in the were poisoned by dimethyltin-contaminated cooking
respiratory system of rabbits exposed to 4-6 mg/m3 oil: of these, 100 people were hospitalized and three
(0.30-0.45 mg/kg) tributyltin chloride for 95 days. people died. Urine samples of those poisoned with
Rats exposed to 2 mg tin/m3 (0.41 mg/kg) an aero- dimethyltin contained dimethyltin and trimethyltin
sol mixture of tributyltin bromide (0.39 mg/kg) and at approximately 80 ng/mL (Jiang et al., 2000). The
dibutyltin dibromide (0.02 mg/kg) displayed severe overall no observed adverse effect level (NOAEL) for
bronchitis and both vascular and alveolar edema neuropathological effects of dimethyltin chloride is
(Iwamoto, 1969). considered to be 0.6 mg/kg b.w.

5.2.1.2 Ingestion 5.2.1.3  Skin Contact


Organotin compounds can be digested from the Organotins range in toxicity from nontoxic trioc-
gastrointestinal tract (Kimbrough, 1976). The degree of tyltin acetate to the highly irritant tributyltins. Expo-
tributyltin oxide absorption was shown to be 20-55%. sure to dimethyltin at 80 mg/kg was shown to result
Human exposure to organotins occurs through con- in skin necrosis and scar formation in rats. The mix-
taminated dietary sources (e.g. seafood and shellfish), ture of diethyltin diiodide and linoic acid used to treat
fungicides on food crops, and water systems (Golub furuncles and other skin infections caused 217 poison-
and Doherty, 2004). High levels of tributyltin reaching ings and 111 deaths in 1954 (Barnes and Stoner, 1959).
up to 11 mg/kg were found in bivalves, gastropods, However, the identified toxic component was triethyl-
and fish, suggesting that human diet can contain high tin. Short-chain alkyltin compounds can be absorbed
levels of organotin compounds (WHO, 1990). The total easily, which makes them the most toxic for dermal
tolerable average level of tributyltin consumption in application. Studies on rat dorsal skin demonstrated
Taiwan was calculated to be 243 ng/g wet weight (Lee that tributyltin is more irritating than dibutyltin
et al., 2005). The lowest observed adverse effect level (Middleton and Pratt, 1978). The application of tribu-
(LOAEL) value of 0.025 mg/kg per day for dibutyl- tyltin at 33 nmol/cm2 produced epidermal necrosis
tin for oral exposure was derived from the LOAEL of and dermal inflammation, whereas dibutyltin was
2.5 mg/kg per day for rats in mid-term and long-term less irritating and caused a similar effect at concen-
toxicity tests. The maximum oral exposure value was tration 10 times higher. Contact with butyltin com-
set at around 0.035 μg/kg per day. The half-life for the pounds caused skin lesions in process workers (Lyle,
gastric hydrolysis of dibutyltin ranges between 0.5 and 1958). Another report described irritant foot dermati-
3.5 h. The absorption of dibutyltin and triphenyltin tis caused by tributyltin oxide used to disinfect socks
compounds is approximately 10% in rats, whereas the (Molin and Wahlberg, 1975). Follicular inflammation
absorption of monoethyltin is approximately 8% and and skin lesions induced by butyltins were reported on
of octyltin is less than 0.1% (Sole et al., 1998). Exposure the skin of volunteers’ hands (Lyle, 1958). Workers of
of rats to diets containing 50 mg bis(tributyltin)oxide/ a marine paint plant developed a sharply demarcated
kg for 4.5 months significantly reduced thymus weight skin rash 2 days after skin contact with a chemical
and suppressed natural killer (NK) cell activity in the composed of 50% triphenyltin fluoride and 49% aro-
spleen after 16 months (Vos et al., 1990). Oral admin- matic solvent (Andersen and Petri, 1982). A week after
istration of tributyltin to mice at dose levels of 0.5, 5, exposure, skin biopsy showed follicular and perifollic-
and 50 μg/kg b.w. for 45 days resulted in an increase ular lymphocytic infiltration. Several cases of contact
in body weight gain and hepatic steatosis, accompa- dermatitis caused by triphenyltin have been reported
nied by hyperinsulinemia and hyperleptinemia (Zuo (Goh, 1985; Lewis and Emmett, 1987). Application of
et al., 2011). The plasma levels of insulin, leptin, and 50% triphenyltin fluoride to the forearm skin of four
adiponectin in mice receiving 5 μg/kg tributyltin were volunteers induced an intense burning sensation after
more than 1.5 times higher than in controls. In contrast, 30 min; erythema developed, followed by epidermal
a reduction in body weight gain was observed in rats necrosis. A patient who had been exposed to triphe-
fed diets containing 15 mg/kg tributyltin or 6 mg/kg nyltin showed an acute urticarial eruption (Colosio
triphenyltin (Grote et al., 2004). Dietary exposure to et al., 1991). Exposure to antifouling paints containing
1260 Elena A. Ostrakhovitch

tributyltin oxide resulted in irritation, erythema, and respectively (Harino et al., 2008b). Accumulation of
mild pustule lesions (Lewis and Emmett, 1987). The butyltin compounds was reported in the liver, kidney,
contact-sensitizing potential of tributyltin oxide was and brain tissue of adult southern sea otters found
studied in mice: mice sensitized with 0.25, 0.5, or 1% dead along the coast of California during 1992-1996
tributyltin oxide showed increasing swelling reactions (Kannan et al., 2004). The concentrations of butyl-
24 h after challenge with 1% tributyltin (Stringer et al., tins in liver, kidney, and brain tissues were 0.04-5.3,
1991). A single application of 1% tributyltin oxide to the 0.004-0.265, and 0.0039-0.14 μg/g wet weight butyltin,
ears of mice led to increases in auricular lymph node respectively. The highest concentrations of tributyltin
weight. The contact allergenicity of tributyltin oxide and dibutyltin were found in the liver, whereas mono-
reflects its immunosuppressive effects (Krajnc et al., butyltin was predominant in most tissues and organs,
1984; Vos et al., 1984). Spontaneous dermatitis and except the liver. After single oral administration of
dermatitis induced by sensitization with 2,4,6-trinitro- tributyltin (25 mg/kg b.w.), high levels of tributyltin
chlorobenzene were observed in mice fed a diet con- and its metabolites were found in the liver and kidney.
taining 100 mg/kg tributyltin for 30 days. Diminished The liver predominantly (95%) contained dibutyl- and
interferon gamma (IFN-γ) level and increased interleu- monobutyltin compounds. Preferential accumulation
kin-13 (IL-13) and IL-5 induced by tributyltin aggra- of butyltins in the liver and kidney may be associated
vate atopic dermatitis-like lesions in mice treated with with the presence of increased amounts of metal-­binding
2,4,6-trinitrochlorobenzene (Iwamura et al., 2006). proteins, such as metallothionein-like proteins and glu-
Acute dermal toxicity of 1000-3000 mg/kg triphenyltin tathione, which may play a role in the detoxification
was reported in rabbits. Skin irritation in rabbit is min- of alkyltin agents. Furthermore, liver plays a key role
imal when triphenyltin was used as powder. However, in detoxification. Lipid-soluble toxicants are detoxified
triphenyltin incorporated into paint becomes irritating by cytochrome P450 family proteins. The distribution
to the skin; an inflammatory reaction developed after of tributyltin within organism is rapid. After single
the daily application to the ears of rabbits for 2 days. administration of a 25 mg/kg b.w. dose, high levels
In vitro studies on dioctyltin absorption in rat and were found within 1-3 days. In blood, tributyltin and
human epidermis showed that amount absorbed after its metabolites were detected within 3 h of administra-
24 h was 0.039 μg/cm2 for human skin and 4.14 mg/ tion. In blood, tributyltin and its degradation metabo-
cm2 for rat skin when dioctyltin was applied at a dose lites bind to serum albumin, which may determine the
equivalent to 17,007 mg/cm2 as tin (Ward, 2003). fate, metabolism, and biological function of organotins
(Zhang et al., 2006). The equilibrium-binding constant
[log (Kb)] of tributyltin to the serum albumin complex
5.2.2 Distribution
was calculated to be 7.3 (Sun et al., 2012). The percent-
The accumulation of butyltin compounds such age tributyltin in the serum was shown to be higher
as mono-, di, and tributyltin was reported in the than in tissues (Shim et al., 2002). Dibutyltin is mainly
soft tissues, muscle, and feathers of wild migratory distributed in the kidney, liver, and thymus follow-
birds (Guruge et al., 1996; Senthilkumar et al., 1998; ing dietary administration for 1 week at 100 mg/kg
Senthilkumar et al., 2001). The highest concentrations (Arakawa et al., 1983). Intraperitoneal administration
were found in feathers (ranging from 12 to 290 ng/g), of dibutyltin to rats led to accumulation of the dibu-
whereas concentrations recorded in tail feathers were tyltin metabolite, butyl (3-hydroxybutyltin), in the
73-360 ng/g for monobutyltin, 29-56 ng/g for dibutyl- kidney at a relatively high concentration compared
tin, and 37-67 ng/g for tributyltin. These data indicate with other metabolites; moreover, its concentration
that of the butyltins, monobutyltin is the predominant increased with time (Ishizaka et al., 1989). Dibutyltin
compound retained in birds. It has been shown in fish and its metabolites were found in brain tissue, whereas
and marine mammals that tributyltin predominantly butyl-(3-hydroxybutyltin) was also found in urine.
accumulates in blubber and muscle, whereas dibutyltin Triphenyltin was mainly distributed in the kidney,
levels were shown to be highest in the liver and kidney followed by liver, brain, and heart of rats a few days
(Guruge et al., 1996; Kannan et al., 1996). Distribution after its administration (Eckert et al., 1989; Kellner
of tributyltin is rapid and reaches the maximum plasma and Eckert, 1989). A similar distribution pattern was
level in 1 day. In seven species of dolphin (bottlenose observed after chronic exposure for 104 weeks (Dorn
dolphin, finless porpoise, Indo-Pacific humpbacked and Werner, 1989; Tennekes et al., 1989). Relatively
dolphin, long-beaked common dolphin, pantropi- high concentrations of triphenyltin were detected
cal spotted dolphin, spinner dolphin, and striped in the liver, pancreas, kidney, and brain of hamsters
dolphin), butyltin and phenyltin compounds were within 24 h of administration of 50 mg/kg triphenyltin
found in the range of 16-1152 μg/kg and 1-62 μg/kg, (Ohhira and Matsui, 1996). However, dearylation was
56 Tin 1261

slower in hamsters than in rats. There was a high pan- tetraethyltin at 10 mg/kg accumulated in the kidney at
creatic accumulation of triphenyltin in hamsters. High relatively high concentrations, whereas only tetrabu-
levels of triphenyltin were found in the brain tissue of tyltin was retained in the liver.
both rats and hamsters after a single dose of 50 mg/
kg triphenyltin chloride or 6 mg/kg fungicide contain- 5.2.3 Excretion
ing 60% triphenyltin (Lehotzky et al., 1982; Ohhira
The excretion of organotins chiefly depends on
and Matsui, 1996). Triphenyltin was shown to cross
the type of compound. The major routes of elimina-
the blood-brain barrier due to its high lipophilicity.
tion from the body are urine, feces, and bile. Ethyl-
After percutaneous administration in guinea pigs, tri-
tin is almost exclusively excreted in the urine in rats,
phenyltin (2 mg/kg b.w.) was distributed to the high-
whereas diethyltin is excreted in the feces, urine, and
est extent in the liver, followed by the adrenal glands,
bile (Bridges et al., 1967; Cremer, 1957). After daily
kidney, brain, spinal cord, and pancreas (Nagamatsu
intravenous administration, carboxyethyltin is rapidly
et al., 1978). It was shown that tetraphenyltin accumu-
excreted unchanged in urine without the presence of
lates more rapidly than triphenyltin in the liver of rats
metabolites (Penninks and Seinen, 1985). However,
fed with 55.4 mg/kg of this compound (Ohhira and
carboxyethyltin was excreted in feces and urine after
Matsui, 2003).
daily oral administration at a dose of 15 mg/kg. Tri-
Dioctyltin is rapidly distributed to the liver and kid-
ethyltin can be eliminated in urine and milk, at least
ney, and to a lesser extent to the adrenal, pituitary, and
in lactating sheep. The excretion route for tributyltin
thyroid glands, with poor diffusion into the brain in
is the bile rather than the urine (Evans et al., 1979).
rats dosed orally at 6.3 mg/kg b.w. or intravenously at
Thus, approximately 90-95% of ingested tributyltin is
1.2 mg/kg (Penninks et al., 1987). There was no selec-
excreted in the feces. Similar to butyltin, fecal excretion
tive accumulation of dioctyltin in the thymus, despite
was predominant after oral administration of dodecy-
the fact that thymus atrophy is the most sensitive
ltin, ocyltin, and phenyltin compounds (Eckert et al.,
marker of dioctyltin toxicity in rats. Dodecyltin com-
1989; Penninks et al., 1987; IPCS, 1999). About 10% of
pounds were mainly found in the thymus and liver,
dioctyltin is excreted in the urine as the unchanged
followed by the kidney and bone marrow (Schering,
parental compound. Approximately 32-53% of tri-
1992). The concentration of this organotin in the brain
phenyltin orally and intravenously administered to
was only 1.4% of the amount found in the liver.
Wistar rats at a dose of 2 mg/kg b.w. was excreted in
The high concentrations of triethyltin were found in
the feces. Renal excretion was 12% after oral admin-
the liver of rabbits, with smaller quantities in the kid-
istration of triphenyltin, reaching 24% following
ney, brain, and blood 2 h after intravenous administra-
repeated administration. After intravenous adminis-
tion of triethyltin at 20 mg/kg for 5 days followed by
tration, approximately 30% was excreted renally. Tri-
10 mg/kg for another 2 weeks (Cremer, 1957). Accumu-
phenyltin administered for seven consecutive days
lation of triethyltin was observed in the kidney, blood,
was predominantly excreted fecally (96-100%) within
and lung 1 h after administration, with penetration into
7 days, with less than 1% excreted renally (Eckert et al.,
the brain and adipose tissue 12 h later (Doctor and Fox,
1989). Although some of the administered triphenyl-
1982). Other experiments have shown that exposure
tin was excreted unchanged, a significant portion was
to triethyltin or trimethyltin results in higher tin con-
excreted as diphenyltin and monophenyltin, along
centrations in brain, liver, and kidney tissues, with less
with other polar products (Kimmel et al., 1977).
accumulation in the heart and blood (Cook et al., 1984).
Rates of elimination were shown to be greater in all tis-
5.2.4  Biological Half-Life
sues following triethyltin exposure than following tri-
methyltin exposure. Administration of trimethyltin at Biodegradation of tributyltin differs in various
dosages of 3.0, 6.0, and 9.0 mg/kg led to its accumula- organs. The calculated half-life for elimination of
tion in the brain, reaching 4.4, 8.5, and 12.7 ng tin/mg injected tributyltin is a few days in mice, with initial
protein, respectively, 12 h after exposure. Trimethyltin rapid elimination being followed by slow elimina-
can easily cross the blood-brain barrier and becomes tion in the feces for up to 29 days (Brown et al., 1977).
evenly distributed across the cerebellum, medulla-pons, A half-life of 8.9 days was estimated for dioctyltin
hypothalamus, hippocampus, and striatum. Dimethyl- (Penninks et al., 1987). However, urinary excretion of
tin exposure caused hemorrhages in the digestive tract dioctyltin was continuous over a period of 25 days. For
and severe hepatic and spleen injury in both rats and diethyltin, the half-life was estimated at 3-4 days in the
mice (Tang et al., 2013b). The amount of triethyltin has liver and kidney, with a prolonged half-life in muscles
been shown to increase with time (­Arakawa et al., 1981). and bones after oral administration. The longest bio-
Orally administered tetrapropyltin, tetrabutyltin, and logical half-life was reported in the brain, compared
1262 Elena A. Ostrakhovitch

with other organs (WHO, 1980a). For tricyclohexyl- et al., 1999). Exposure of the wharf roach (Ligia exotica
tin, the half-life ranges from 5 to 40 days, with slow Roux) to a tributyltin-containing diet (0.1 mg/g food)
removal from the brain. For triphenyltin, the biological for 2 days resulted in the accumulation of tributyltin
half-life was calculated to be approximately 3 days in and its metabolite dibutyltin. The highest tributyltin
rat brain, with a much longer half-life in guinea pig concentration (2760 ng/g) was detected 1 day after
brain. The biological half-life of triphenyltin in guinea cessation of exposure, whereas the highest dibutyltin
pigs was estimated to be 9.4 days (Nagamatsu et al., concentration (2960 ng/g) was measured 2 days later
1978). The biological half-life of triphenyltin in short- (Undap et al., 2013b). The concentrations of these com-
necked clams and guppies was estimated to be 30 and pounds gradually decreased during the depuration
48 days, respectively (Tas et al., 1990). The estimated period; the half-life of tributyltin was estimated to be
half-lives of dimethyltin and trimethyltin in the urine about 4 days for the wharf roach. Tributyltin rapidly
of mice were calculated to be 0.3 and 1.3 days, respec- accumulates in the digestive and reproductive organs
tively (Furuhashi et al., 2008). In rats, the half-lives of of the sea snail (Thais clavigera; of the rock shell fam-
dimethyltin and trimethyltin were estimated as 0.35 ily) and its degradation index ranged from 0.4 to 23.3
and 11.6 days, respectively. The half-life of trimethyltin (Wang et al., 2010; Fang et al., 2011). However, biodeg-
in rats was much longer than that in mice. radation of tributyltin was lower in female reproduc-
tive organs. Digestive and reproductive organs have
higher degradation indices than other tissues. The deg-
5.2.5 Biotransformation
radation rate of tributyltin to dibutyltin is higher than
Biotransformation of organotin compounds by ter- that of dibutyltin degradation to monobutyltin, or of
restrial and aquatic organisms proceeds by successive monobutyltin degradation to inorganic tin.
dealkylation and dearylation of tetra-, tri-, and disub- Butyltin and phenyltin compounds undergo deal-
stituted organotin compound, with each successive kylation and dearylation by the microsomal cyto-
breakdown intermediate being less toxic than the last. chrome P450 monooxygenase system (Fish, 1984;
In experiments conducted by Stasinakis et al. (2005) Ueno et al., 1995; Appel, 2004). In the presence of
in an activated sludge reactor, tributyltin and dibu- phenobarbital, which activates the cytochrome P450
tyltin biodegradation took 18 days. Biodegradation system, triorganotin metabolism accelerates (Ohhira
of tributyltin occurs in lower organisms, but with an et al., 1999b). The total yield of tributyltin and triphe-
elimination rate that is much slower than in mammals; nyltin metabolites increased by approximately 1.8
this suggests that organotin bioaccumulation is much and 8.9 times, respectively, in rats, 2.1 and 1.2 times
higher in lower organisms than in mammals. The half- in hamsters, and 1.6 and 1.5 times in human isolated
lives of tributyltin, dibutyltin, monobutyltin, and tri- hepatic microsomes (Ohhira et al., 2003). Tributyltin
phenyltin are 1.4, 3.6, 9.8, and 5 days, respectively. The was metabolized more readily than triphenyltin in
metabolism of organotin compounds determines their all species. Dealkylation of dibutyltin to monobutyl-
environmental fate and retention. tin by the monooxygenase system is slower than that
Dearylation reactions have been reported for triphe- of tributyltin to dibutyltin (Kannan et al., 1996). Four
nyltin (Stasinakis et al., 2005). In sediments, triphen- cytochrome P450 human isoforms (CYP2C9, CYP2C18,
yltin degradation occurs relatively slowly through CYP2C19, and CYP3A4) were shown to mediate deal-
photodegradation (Fent et al., 1991a,b; Kannan and Lee, kylation and dearylation of tributyltin and triphenyl-
1996). Less than 2.5% of triphenyltin was found bio- tin compounds in isolated human hepatic microsomes
transformed in the goldfish Carassius auratus, whereas (Ohhira et al., 2006b). However, the CYP2C18 content
triphenyltin biotransformation was not observed in is low in human hepatic microsomes, whereas the per-
larvae of the minnow Phoxinus phoxinus. Only half of centages of CYP2C9 and CYP2C19 accounts for about
the initial triphenyltin concentration remained; the 53 and 11% of the CYP2C subfamily. Therefore, it
remainder was biotransformed into diphenyltin, mon- was suggested that CYP2C9, CYP2C19, and CYP3A4,
ophenyltin, and inorganic tin. The ecological half-life which is the most abundantly expressed cytochrome
of triphenyltin in gastropods was estimated to be 347 P450 in human liver, are the major isoforms involved
days (Mensink et al., 1996). in the metabolism of organotins in the human liver. In
The bioconcentration factor for tributyltin is highest isolated liver microsomes, cytochrome P450 monooxy-
for organisms that lack efficient degradation systems, genases hydroxylate the principal carbon-hydrogen
but it is easily degraded by fish, birds, and mammals bonds in tributyltin to produce hydroxylated alpha
(Walker et al., 2006). Exposure of Chlorella vulgaris to metabolites, which rapidly split to form the dibutyl
tributyltin resulted in rapid biosorption, with sequen- derivative, followed by further oxidation to 1-butanol
tial degradation to dibutyltin and monobutyltin (Tsang and then to 1-butane and ketones (Kannan et al., 1996).
56 Tin 1263

In rats, CYP2C subfamily members have a key role in including water and sediments, near “hot spots” such
tributyltin and triphenyltin metabolism (Ohhira et al., as boat marinas and shipyards. Concentrations of butyl-
2004, 2006a). CYP2C6 was shown to be responsible tins > 1 μg/g wet weight were observed in the ovary,
for dearylation of triphenyltin. However, triphenyl- digestive gland, kidney, heart, ctenidium, osphradium,
tin may also convert cytochrome P450 to cytochrome stomach, head ganglia, and penis of both imposex-
P420, resulting in inactivation of monooxygenase sys- exhibiting female and male rock shells (T. clavigera) col-
tem (Prough et al., 1981). Dearylation was shown to be lected at Jogashima, Japan, which is considered to be the
slower in hamsters than in rats. most highly organotin-contaminated site (Horiguchi
Dealkylation of diethyltin may occur in the liver, gut, et al., 2012). About 90% of collected rock shells were
and other organs. Alkylated organotin compounds can sterile. Concentrations of tributyltin and triphenyltin
be demethylated by P450 monooxygenases in mam- in the ovaries were 2.46 and 2.28 μg/g wet weight. A
mals. Besides dealkylation, dimethyltin can be also similar accumulation of tributyltin compounds was
methylated to form trimethyltin in mice and rats, sug- observed in Ocenebra erinacea and T. clavigera (Horiguchi
gesting that alkylation and dealkylation reactions can et al., 2006). One-third of the total butyltin concentra-
co-occur (Furuhashi et al., 2008); however, the dealkyl- tion was detected in the digestive glands. The total
ation rate is greater than the alkylation rate. The con- concentration of organotins in different organs of the
version rate of dimethyltin to trimethyltin in rats was cobia (Rachycentron canadum) collected from the waters
estimated to be about 0.8%, resulting in about 2.4% in southern Taiwan ranged between 206 and 859 ng/g
of trimethyltin being present in the blood following wet weight (Liu et al., 2011). Accumulation of tri- and
exposure to dimethyltin (Tang et al., 2013b). However, dibutyltin in the Queen conch (Strombus gigas) was
the mechanism of methylation is not understood. recorded at Road Harbour in the British Virgin Islands
(Titley-O’Neal et al., 2013). The tributyltin concentra-
tion was in the range of 8.3-23.7 ng Sn/g dry weight,
6  LEVELS IN TISSUE AND BIOLOGICAL while the dibutyltin concentration was in the range of
FLUIDS 84.2-88.9 ng Sn/g dry weight. Butyltin concentrations
in mussels collected from the Venetian Lagoon between
The average tin concentration in urine is about 4.2- 1999 and 2003, in particular those from the area around
42.2 nmol/L, and in hair is about 0.42-3.37 nmol/g. the city of Chioggia, were in the range of 187-6666 for
The average concentration of tin in the blood of nor- tributyltin, 80-4187 for dibutyltin, and 31-1538 μg/kg
mal subjects is 0.14 mg/L; it is found mainly in eryth- dry weight for monobutyltin. The detected values did
rocytes (Baselt and Cravey, 1989). Tissue analyses have not significantly differ from those reported in 2000
shown that tin is present in lung, adrenal gland, and (Zanon et al., 2009; Gallina et al., 2000). Butyltin com-
liver tissues at concentrations of 37, 23, and 23 mg/kg, pounds in the tissue of mussels from Egremont were
respectively. The highest amount of tin (in dry ash) has shown to be in the range 0.217-0.508 mg/kg; these
been found in the cecum (130 mg/kg), ileum (79 mg/ values were higher than those of mussels from New
kg), rectum (57 mg/kg), and sigmoid colon (45 mg/ Brighton (0.11-0.201 mg/kg dry weight) (Harino et al.,
kg) (Schroeder et al., 1964). Inorganic tin does not 2005). Since the prohibition of organotin-based anti-
accumulate in soft tissues with increasing age (Baselt fouling paints on ships worldwide after January 2008,
and Cravey, 1989). A dose-dependent increase in the the percentage of Nassarius reticulatus females affected
tin content of the tibia and kidney of weaning rats has with imposex along the northern continental shelf of
been reported after administration of tin in the diet, Portugal decreased from 84% (in 2006) to 20% (in 2010)
ranging from 1 to 2000 μg/g food (Johnson and Greger, in the same surveyed sites, suggesting recovery of the
1985). ecosystem (Barroso et al., 2011). Considerable levels of
The level of organotin compounds in the environ- tributyltin (1.4-190 ng/mL) were reported in the blood
ment have shown declining trends since the early of various species of fish collected at costal sites near
2000s as a result of the ban on tributyltin and triphen- Fukuoka in Japan in 2008 (Miki et al., 2011). Tributyltin
yltin use, but these compounds have still been detected accumulation in ripe testes (over 50 ng/g wet weight)
in a wide variety of organisms, and even in deep-sea was reported for male turbot (Scophthalmus maximus)
organisms. On the one hand, tributyltin, triphenyltin, collected from the Gulf of Gdansk and the Pomeranian
and their degradation products are still being released Bay in the southern Baltic Sea (Gosz et al., 2011). The
from sediments and soils; on the other hand, dibutyl- high concentrations of tributyltin and its metabolites
tin and monobutyltin are still in use in industrial pro- were found in the liver and kidney of adult common
cess in some parts of the world. Some studies reported cormorants (Phalacrocorax carbo) collected from Lake
high levels of organotins in environmental samples, Biwa in Japan. The levels of butyltin compounds
1264 Elena A. Ostrakhovitch

in the liver were lower in 2007 (8 ng/g) than in 1993 The number of studies that have examined organo-
(270 ng/g), but were still detectable (Mizukawa et al., tin compounds in human tissues is limited. It was
2009). Diphenyltin and triphenyl tin were distributed reported that the concentrations of butyltins in the
uniformly among organ and tissues, and their levels liver of ­Japanese subjects ranged from 59 to 96 ng/g
significantly decreased from 130 ng/g in 1993 to 2 ng/g wet weight, whereas monobutyltin and dibutyltin,
in 2007. The level of tributyltin and its metabolites degradation products of tributyltin, were predominant
in the blubber, liver, lung, and muscle of adult killer (Takahashi et al., 1999). Hepatic deposition of butyltin
whales (Orcinus orca) collected from Rausu, Hokkaido, in a group of Danish men was in the range of 1-33 ng/g,
Japan, were in the range of 37-90, 385-676, 15, and while total butyltin concentrations in human liver sam-
26-53 μg/kg wet weight, respectively (Harino et al., ples collected in Poland were in the range of 2.4-11 ng/g
2008a). Phenyltin concentrations were below 4 mg/ (Nielsen and Strand, 2002; Kannan and Falandysz,
kg in all tissues except the liver (58 mg/kg). Tributyl- 1997). Variations in concentrations butyltin among indi-
tin oxide can be transferred across from the placenta viduals from different countries probably reflect differ-
to the fetus. However, total butyltin concentrations in ences in their dietary intake of fish (Nielsen and Strand,
the blubber and liver of a whale calf were lower than 2002). Butyltin concentration in blood collected from
those in adult whales. It was shown that concentra- 32 volunteers during a blood drive organized by the
tions of butyltin and phenyltin compounds were in the American Red Cross in central Michigan in 1998 ranged
range of 16-1152 μg/kg and 1-62 μg/kg in organs and from 0 to 101 ng/mL (Kannan et al., 1999). After remov-
tissues of dolphins (bottlenose dolphin, finless por- ing the highest concentration from the data, the aver-
poise, Indo-Pacific humpbacked dolphin, long-beaked age concentration was about 4.5 ng/mL. The amounts
common dolphin, Pantropical spotted dolphin, spin- of monobutyltin, dibutyltin, and tributyltin measured
ner dolphin, and striped dolphin) from the coasts of in human blood collected from individuals in central
Thailand (Harino et al., 2008b). Michigan were 15.4 ± 5.2, 6.08 ± 3.33, and 10.5 ± 16.7 ng/
The administration of 17.6 μg/mL of tributyltin in mL, respectively (Kannan et al., 1999). Whalen et al.
drinking water leads to accumulation in the kidney at found a maximum concentration of 94 ng/mL dibutyltin
concentration equivalent to 2.1 mg/g wet tissue and in in human blood (Whalen et al., 1999). Analysis of urine
the brain to 0.36 mg/g wet tissue. The total butyl con- samples from four male volunteers aged 25-45 years
tent in the liver of mice exposed to tributyltin is about indicated the presence of tributyltin in two subjects at
1000 ppb and remains unchanged 24 h after adminis- a relatively low level (25 and 49 ng/L) (Zachariadis and
tration. The content of tin in the liver of rats and guinea Rosenberg, 2009a). Monobutyltin was detected in one
pigs is lower at 3 h and higher at 24 h than those of mice sample at concentration of 34 ng/L. The occurrence of
(Ueno et al., 2003). The main form of butyltin in the butyl compounds in human blood suggests widespread
liver of rats treated with tributyltin was dibutyltin. exposure to these compounds from a variety of sources.
After a single oral dose of tributyltin, high levels of tri- When tributyltin in a mixture of 3 mL cherry brandy
butyltin were seen in the frontal and temporal lobes and 7 mL ethanol was given orally to a volunteer, only
and in the cerebellum of rabbits. Oral administration 5.1-5.4% of the dose was excreted in the urine, mainly
of monooctyltin trichloride to rats at 25 mg/kg b.w. as dibutyltin metabolites (Uhl, 1986). In 1999, more than
resulted in an increased concentration of monooctyltin 1000 people were exposed to methyltin in cooking oil
in the blood, reaching 62 ng/mL within 3 h (Penninks in southeast China. The concentration of trimethyltin
et al., 1987). Absorption was estimated at 0.03% of the in blood samples from these people after the incident
dose received. After a single oral dose of 55.4 mg/kg was about 70 ng/g, whereas levels of di- and trimethyl-
tetraphenyltin in oil to Wistar rats, the highest concen- tin in urine were about 80 ng/mL (Jiang et al., 2000). In
tration of diphenyltin was observed in the liver and another study, the concentration of trimethyltin in urine
kidney (Ohhira et al., 2003). Urinary dibutyltin excre- samples from 15 occupationally exposed volunteers
tion rates in mice and rats after intraperitoneal injec- ranged from below the detection limit to 1277 ng/L
tion of dimethyltin dichloride (10 mg/kg b.w.) for (Cui et al., 2011). Concentrations of dimethyltin were
four consecutive days were approximately 43% for in the range of 26-6353 ng/L, whereas monomethyl-
both species (Furuhashi et al., 2008). Dimethyltin was tin was detectable in less than half of the samples, and
detected until day 10, with the highest level of 98 μg ranged from 68 to 261.9 ng/L. Analysis of tin in organs
detected in the urine at day 3 in mice and of 845 μg showed the highest methyltin concentrations in the liver
in the urine at day 4 in rats. Trimethyltin was absent (1.93 μg/g dimethyltin and 1.42 μg/g trimethyltin), kid-
in the urine on day 0 but a time-dependent increase in ney (1.05 μg/g dimethyltin and 0.47 μg/g trimethyltin),
its concentration thereafter was observed. The highest and heart (0.1 μg/g dimethyltin and 1.48 μg/g trimeth-
concentrations were 1.26 μg in mice and 3.49 μg in rats. yltin). In the Korean worker cleaning the tank used in
56 Tin 1265

production of dimethyltin chloride, the circulatory level gastrointestinal disturbances (Nehring, 1972). A total
of trimethyltin was highest on day 12 after the 4-day of 74 out of 78 people who ingested canned peaches
exposure to dimethyltin; it was 208.3 μg/L (Yoo et al., containing 100 mg or more of tin developed digestive
2007). The concentration of dimethyltin was lower than problems. In a randomized, double-blind study, 3 out
the concentration of trimethyltin at all time points; they of 18 healthy volunteers of both sexes who consumed
were approximately 40 μg/L or slightly over from day 250 mL tomato juice containing stannous chloride at
10 to day 15 after exposure. The highest urinary concen- concentrations of 264 mg/kg developed gastrointesti-
trations of dimethyltin and trimethyltin were 779.4 and nal symptoms (Boogaard et al., 2003). Consumption of
946.3 μg/g Sn, respectively. However, monomethyltin tomato juice to which stannous chloride was added at a
was not detected in urine samples. concentration of 529 mg/kg caused mild and moderate
gastrointestinal disturbances in four out of five volun-
teers. In a case-control study conducted in Belgium, an
7  EFFECTS AND DOSE-RESPONSE increased risk of chronic failure was reported for occu-
RELATIONSHIPS pational exposure to tin (Nuyts et al., 1995).
Symptoms of gastrointestinal illness were reported
Tin is not an essential element for humans and no in five volunteers of each sex after drinking orange
specific role for tin has been identified in human health. juice containing tin at concentrations up to 1400 mg/
However, tin has been established to be required for kg (Benoy et al., 1971). However, nine volunteers who
some animals such as rats to grow and develop (Reicks ingested tin at a dose of 120-200 mg/day did not show
and Rader, 1990; Yokoi et al., 1990b). adverse effects (Calloway and McMullen, 1966). Inges-
tion of a tin solution containing a dose of 700 mg/kg
caused nausea and diarrhea in two volunteers, whereas
7.1  Inorganic Tin six others did not experience adverse effects (WHO,
7.1.1  Local Effects and Dose-Response Relationships 2001). Consumption of 36 mg tin with a meal resulted in
decreased zinc absorption in 10 volunteers aged 16-46
7.1.1.1 Humans years (Valberg et al., 1984). A 40-day balance study
Most natural food products contain tin in trace revealed an increase in fecal excretion of selenium by
amounts, but concentration may increase in canned food 30% in healthy volunteers after ingestion of food con-
as a result of dissolution of the tin coating or tin plate in taining tin at a dose of 50 mg (Greger et al., 1982).
damaged or improperly stored cans. There have been Positive allergic reactions for metallic tin and stan-
several reports on outbreaks of food poisoning. A case nous chloride have been described in several case
was reported in which 31 persons experienced nausea, reports (Menne et al., 1987; Fine Olivarius et al., 1993;
abdominal cramps, and vomiting within 1-2 h of drink- Gaddoni et al., 1993). Of 73 patients, six developed
ing fruit punch containing 2000 mg/kg tin (Warburton reactions in patch tests with tin-plated copper disks.
et al., 1962). Eight cases were reported in 1969 after Among 2205 people tested for allergic reaction to 1%
ingestion of tomato juice containing 250 mg/kg tin. stannous chloride in petrolatum, 5 subjects produced
Acute gastrointestinal illness was reported among peo- positive reaction, 15 showed questionable positive
ple who ingested canned tomato juice contaminated responses, while 10 showed irritation to stannous chlo-
with tin in Washington and Oregon in 1969 (Barker and ride. Testing with 10% stannous chloride showed that
Runte, 1972). The most commonly described symptoms 11 out of 19 subjects had irritant skin reaction.
were bloating, abdominal cramps, headaches, fever,
and mouth lesions. Tin levels measured in the cans 7.1.2  Systemic Effects and Dose-Response
implicated in outbreak ranged from 131 to 405 mg/kg. Relationship
The high concentrations of nitrate on the tomatoes used
to prepare the juice and the excessive nitrate fertiliza- 7.1.2.1 Humans
tion of tomato plants were associated with corrosion of There have been many case reports of acute gas-
the can lining. Vomiting, diarrhea, and other digestive trointestinal illness following the intake of canned
problems have been observed in a large number of per- fruits and fruit juices (Blunden and Wallace, 2003). The
sons after the consumption of canned orange juice and most frequently reported symptoms were abdomi-
apple juice containing 250-390 mg/kg tin in Kuwait nal cramps, nausea, vomiting, and diarrhea. Tin(II)
(Benoy, Hooper, and Schneider, 1971). Fifteen out of compounds (except for tin hydride) affect heme
26 persons who consumed orange juice containing metabolism. Gaseous tin hydride is neurotoxic when
about 300 mg/kg tin showed gastric symptoms. Simi- inhaled: similar to arsine gas, tin hydride causes nerve
larly, canned peaches containing 563 mg/kg tin caused damage, but its effects are less profound because it does
1266 Elena A. Ostrakhovitch

not cause hemolysis (Baldwin and Marshall, 1999). Stan- for stannous chloride after 24 h. Cats are more sensitive
nane (SnH4) is an analog of methane and may cause than rats to oral administration of tin. Vomiting and
hemolytic anemia. Exposure to high doses of stannane diarrhea developed only in cats after the oral adminis-
may cause gastrointestinal illness, skin rash, abdominal tration to cats and rats of fruit beverages containing tin
pain, and headaches, whereas low levels of stannane at 472 mg/L or a complex of tin chloride and sodium
result in fatigue, depression, low cardiac output, short- citrate (9 mg/kg b.w.) (JECFA, 1982).
ness of breath, asthma, headaches, and insomnia. Tin at Early reports showed tin-induced toxic pathological
concentrations found in the human diet can decrease changes in the liver and kidney and neurological dam-
copper status, which in turn produces a wide variety age in rats, with an increased incidence of changes in
of anatomical, chemical, and physiological pathologies fatty acids (der Meulen et al., 1974; Conine et al., 1976).
in the cardiovascular system (Klevay and Combs, 2004; The uptake and metabolism of iron, copper, zinc, and
Klevay, 2000; Pekelharing et al., 1994). It was suggested manganese are influenced by dietary tin at concentra-
that uremic patients might be prone to elevated tin levels tions greater than 50 mg/kg. Significant effects of tin
because tin was shown to affect kidney enzyme activity. on liver glutathione peroxidase and superoxide dis-
Increased risk of chronic renal failure was documented mutase activities and on liver iron and total glutathi-
for occupational exposure to tin (Nuyts et al., 1995). one concentrations were observed (Reicks and Rader,
1990). A single intraperitoneal injection of tin tartrate at
7.1.2.2 Animals 20 mg/kg b.w. caused a decrease in glutathione, lead-
Tin is considered an essential nutrient for rats. Rats ing to lipid peroxidation that damaged hepatocytic
fed a tin-deficient diet containing 17 ng/g tin showed membranes (Dwivedi et al., 1983). Exposure to tin pro-
poor growth, lowered response to sound, and alope- duced hematological changes in animals (ATSDR, 2005;
cia, with decreased food absorption efficiency com- Pekelharing et al., 1994). In 4- and 13-week feeding stud-
pared with rats receiving a diet containing 1.99 μg/g ies in rats, oral administration of tin salts (especially the
tin (Yokoi et al., 1990b). Tin deficiency in the diet led more soluble tin salts, i.e. chloride, orthophosphate, and
to increased calcium concentration in the lung and sulfate) and tin oxides at concentrations above 3000 mg/
increased iron concentration in the spleen and kidney, kg caused anemia and extensive damage to the liver
whereas zinc and copper concentrations in the heart and kidney (de Groot et al., 1973; JECFA, 1982). Stan-
were reduced. A tin-deficient diet resulted in poor nous chloride administered at a dose of 10 mg/kg for 3
growth, reduced feeding efficiency, and bilateral (male months caused hemolytic anemia, including decreased
pattern) hair loss. However, chronic administration of hematocrit, erythrocyte number, and hemoglobin levels.
tin chloride at a dose of 10 mg/100 g caused signifi- Intraperitoneal administration of tin for 3 months at a
cant but reversible growth retardation of Wistar-Kyoto dose of 17 mM/kg caused anemia after 2 weeks, whereas
rats (Escalante et al., 1991). Feeding rats a diet supple- anemia was observed 6-10 weeks after oral administra-
mented with tin (10-200 mg/kg) for 28 days did not tion (Chmielnicka et al., 1993). Animal studies into the
affect body weight; however, there was a linear inverse effects of tin revealed that tin interacts with zinc ions
response with food intake (Pekelharing et al., 1994). and blocks the activity of zinc-dependent proteins. Tin
Marked differences in toxicity were shown for stan- was shown to interfere with heme synthesis by replac-
nic and stannous oxides and salts in Wistar rats when ing zinc in δ-aminolevulinic acid dehydratase (ALADH)
administered orally (de Groot et al., 1973). Insoluble and thus decreasing activity of the enzyme. The effect of
tin compounds are relatively harmless if administered tin on heme biosynthesis was shown to depend on the
orally, whereas cationic tin compounds that are soluble concentration of zinc (Chmielnicka et al., 1992). Several
in water or acids can be very toxic because of absorp- studies have reported an effect of tin on bone strength:
tion from the gastrointestinal tract. The reported oral the calcium content and compressive strength of the
median lethal dose (LD50) for stannous chloride is femoral bone were reduced in Wistar rats given 1, 3,
250 mg/kg for mice and 700 mg/kg for rats (Registry 10, and 30 mg/kg b.w. of stannous chloride twice daily
of Toxic Effects of Chemical Substance, 1979), with an for 3, 30, and 90 days (Yamaguchi and Okada, 1980;
oral LD50 value of 775 mg/kg for stannous oxide in ­Yamaguchi et al., 1980, 1982). Stannous chloride admin-
deer mice (Schafer and Bowles, 1985; WHO, 1980a). istered at 1 mg/kg for 30 days produced a significant
Rabbits are less sensitive to stannous chloride, with an decrease in alkaline phosphatase activity of the femo-
oral LD50 of 10000 mg/kg. Sodium pentafluorostan- ral epiphysis. At 0.6 mg/kg b.w. per day, changes in the
nite (NaSn2F5) showed higher toxicity, with an LD50 of calcium content in the femoral epiphysis were not sig-
150 mg/kg in mice and 400 mg/kg in rats. The LD50 in nificant. The results suggested a lowest observed effect
rats after intraperitoneal injection was determined to level of 0.6 mg/kg b.w. per day for orally administered
be 43-50 mg/kg for pentafluorostannite and 136 mg/kg inorganic tin. Decreased bone compressive strength
56 Tin 1267

was also observed in Wistar rats administered with Stannous ions may cause genotoxicity, immuno-
50-600 mg/kg stannous chloride in their drinking water toxicity, neurotoxicity, and oxidative stress. Evidence
(Ogoshi et al., 1981). Calcium content in the tibia was suggests that stannous ions induce DNA damage via
significantly decreased in rats fed 100 mg/kg of stan- the formation of reactive oxygen species, thus linking
nous chloride for 28 days (Johnson and Greger, 1985). tin-induced oxidative stress and genotoxicity (Mclean
The minimal risk level (MRL; defined as an estimate et al., 1983a; Dantas et al., 1996, 1999). Oral adminis-
of daily exposure to substance without appreciable risk tration of SnCl2 led to the generation of free radicals
of adverse noncarcinogenic effects) was calculated to in rabbit liver, testes, kidney, lung, brain, and heart,
be 0.3 mg/kg/day for intermediate-duration oral expo- accompanied by diminished glutathione S-transferase
sure (15-364 days). The intermediate-duration MRL (GST) activity and a decreased level of sulfhydryl (SH-)
was based on a NOAEL of 32 mg/kg for hematologi- groups (El Demerdash et al., 2005). Histopathological
cal effects in Wistar rats fed a diet containing tin in a analysis revealed marked changes in hepatocytes, duct
13-week dietary study (de Groot et al., 1973). A dose of epithelium, and kidney; dilatation and congestion of
95 mg/kg per day is considered the minimal LOAEL. blood vessels; and a mononuclear inflammatory infil-
However, the Agency for Toxic Substances and Disease trate. The volume of the Bowman space was increased,
Registry did not derive a chronic oral MRL for inorganic with infiltration of renal parenchyma and changes in
tin because survival rate was reduced in a 42-month cells lining the proximal and distal convoluted tubules.
drinking water study in rats at the lowest dose (0.7 mg/ Genotoxicity of stannous chloride has been shown in
kg per day as stannous chloride) tested. There were various in vivo and in vitro systems. Genotoxicity was
no adverse effects when Wistar rats were fed stannous demonstrated in the SOS chromotest, calf thymus DNA,
chloride, stannous sulfate, stannous orthophosphate, and isolated nucleotides (Olivier and Marzin, 1987;
stannous oxalate, or stannous tartrate at amounts up de Mattos et al., 2005). Tin(II) ions form a complex with
to 0.1% in the diet for 4 weeks. However, at 0.3% and DNA and generate reactive oxygen species, which
above, these compounds resulted in growth retarda- may be responsible for DNA lesions. Genotoxic and
tion, anemia, and histological changes in the liver. Like- comutagenic effects of stannous chloride were shown
wise, there were no adverse effects in studies in which in V79 Chinese hamster lung fibroblasts, Chinese ham-
stannous chloride was administered to F344 rats and ster ovary cells, and human blood cells (Mclean et al.,
B6C3F1 mice at 1900, 3800, 7500, 15,000 and 30,000 (the 1983a,b; Viau et al., 2009). Exposure of V79 cells to stan-
highest dose in mice only) mg/kg in the diet for 2 weeks nous chloride resulted in a dose-dependent (at concen-
(NTP, 1982). At the highest dose, a loss of weight was tration range of 100-1000 μM) increase in DNA damage.
reported. No significant toxicity was observed in Wistar Although 50 μM SnCl2 did not affect DNA migration in
rats given pentafluorostannite at 20 mg/kg b.w. per day the comet assay, it did inhibit the repair of DNA dam-
for 1 month by stomach tube (Conine et al., 1976). At a aged by methyl methanesulfonate, suggesting that tin
dosage of over 100 mg/kg b.w. per day, degenerative can interfere with steps of the DNA repair systems. Inter-
changes were detected in the kidney in 15-20% of the ference with the DNA repair system may contribute to
animals. Long-term oral administration of tin salts may mutations by shifting the balance from error-free to error-
increase the occurrence of tumors. Mammary adenocar- prone repair processes. Pungartnik et al. (2005) defined
cinoma, uterine sarcoma, and adenocarcinoma near the SnCl2 (stannous chloride) as a moderate mutagen of low
jaw occurred in a group of 13 male and 17 female rats toxicity in Saccharomyces cerevisiae. SnCl2 was reported
fed on diet containing 2% chlorostannate for more than to induce a dose-dependent increase in the frequency of
1 year (Roe et al., 1965). Sixteen percent of B6C3F1 mice chromosomal aberrations and teratogenic effects in mice
fed stannous chloride at concentration 2000 mg/kg (or and zebrafish (El Makawy et al., 2008; Sisman, 2011). No
0.2%) for 105 weeks developed hepatocellular carcino- mutagenic and teratogenic effects were reported for stan-
mas. An increased incidence of histolytic lymphomas nic chloride, stannous oxide, and stannic oxide.
was reported in female mice fed a diet containing stan- It was determined that stannous chloride admin-
nous chlorite. Similarly, injection of metallic tin pow- istered orally up to 50 mg/kg to pregnant mice for 10
der into Lewis rats caused enlargement of the regional days had no discernible effects on maternal or fetal sur-
draining lymph nodes, with epithelioid cell granulo- vival. Similarly, when administered to pregnant rabbits
mas around phagocytosed particles of tin and intense at 41.5 mg/kg for 13 days, maternal or fetal survival was
hyperplasia of plasma cells (Levine and Saltzman, unaffected. Stannous chloride with casein in an aqueous
1996). Injection of metallic tin powder into August rats medium at dose levels of 200, 400, or 800 mg/kg did not
enlarged the lymph nodes, but the enlargement was affect reproductive performance in rats, although ane-
caused by granulomas without a major concomitant mia developed in the offspring before weaning (JECFA,
plasma cell response. 1982). However, administration of stannous chloride at
1268 Elena A. Ostrakhovitch

a high dose of 20 mg/kg b.w. caused a reduction in the tributyltin poisoning were muscle weakness, tremors,
rate of successful pregnancies in female rats, with no numbness in the extremities, eye irritation, respiratory
adverse effects on the implantation number in females distress, chest pain, poor coordination, unsteadiness,
achieving pregnancy (El Makawy et al., 2008). Further- and even seizures. Organic tin compounds are toxic
more, administration of tin decreased fetal body weight because they (1) inhibit the synthesis of heme oxygen-
and was associated with delayed bone ossification. Stan- ase and (2) can also be genotoxic. They cause severe
nous chloride reproductive toxicity was observed in irritation and burning to the skin because they are
rabbits (Yousef, 2005). Teratogenic and genotoxic effects absorbed through this route; this can lead to systemic
of stannous chloride were also observed in zebrafish. toxicity, which is also produced when these compounds
Exposure of zebrafish embryos to 10-250 μM tin for are inhaled or ingested. Major effects may be anemia,
120 h resulted in developmental delay, reduced body as well as renal and hepatocellular damage. Catechol-
weight, morphological malformation, and decreased amine production may also be stimulated, which may
survival rate for embryos and larvae (Sisman, 2011). lead to hyperglycemia, changes in blood pressure, and
Embryos exposed to 100 μM tin displayed tail deforma- damage to the immune system. Alkyl and aromatic tin
tion and larvae exposed to 50 μM tin showed reduced compounds are highly potent neurotoxins. Trialkyltin
body growth, smaller head and eyes, a bent trunk, mild compounds, such as triethyltin, can cause encephalop-
pericardial edema, and a smaller caudal fin. athy and cerebral edema. Excessive industrial exposure
Effects of SnCl2 include stimulation followed by to triethyltin produces symptoms such as headaches,
depression of the central nervous system (CNS) in visual defects, and nausea (Prull and Rompel, 1970).
laboratory animals (Silva et al., 2002). A recent study Triethyltin uncouples oxidative phosphorylation,
demonstrated a neuroprotective effect of stannous which leads to mitochondrial damage, cerebral edema,
chloride in the hippocampus of rats subjected to cere- and nerve damage. Workers exposed to organic tin
bral ischemia (Volti et al., 2011). The administration of compounds (trimethyl and triethyl derivatives) devel-
stannous chloride at 10 mg/100 g b.w. 18 h before the oped psychomotor disturbances, tremor, convulsions,
induction of cerebral ischemia decreased inducible hallucinations, and psychotic behavior (Hu, 1998).
nitric oxide synthase (iNOS) expression in ischemized Occupational exposure to trimethyltin was associated
rats, increased cell survival, and improved short-term with an increased prevalence of nephrolithiasis (kidney
memory recovery. The protective effect of SnCl2 (given stones). Workers exposed to trimethyltin had a higher
12 h before K2Cr2O7) against K2Cr2O7-induced neph- prevalence of kidney stones (18.06%) in comparison
rotoxicity in rats was associated with the induction of with unexposed workers (5.88%) (Tang et al., 2013a).
heme oxygenase 1 (HO-1) activity (Barrera et al., 2003). Patients who inhaled fungicide powder containing
The simultaneous injection of SnCl2 and K2Cr2O7 had 60% triphenyltin or were exposed to triphenyltin while
no protective effect, whereas injection of SnCl2 12 h after spraying it complained of dizziness, nausea, and pho-
K2Cr2O7 exacerbated renal damage. Stannous chloride tophobia (Manzo et al., 1981). No neurobehavioral
is used as a reducing agent in nuclear medicine kits for studies have been done in children whose mothers
the binding between Tc-99m and a drug such as meth- were exposed to organotins during pregnancy (Sokas,
ylenediphosphonate (MDP) for bone scintigraphy and 1998). Skin burns and severe lesions on the hands have
diethylenetriaminepentaacetic acid (DTPA) for kidney been reported among workers occupationally exposed
and brain scintigraphy to obtain the radiopharmaceu- to liquid tri- and dibutyltin chlorides because of leak-
tical needs. It was shown that tin complexes with MDP ing gloves or a failure to wear hand protection (Lyle,
and DTPA to form atoxic compounds (Guedes et al., 1958). The development of severe dermatitis has been
2006). DNA damage was detected in peripheral blood described in workers accidentally exposed to tributyl-
nuclear cells from patients who had received endove- tin and in painters applying tributyltin formulations
nously injected radiopharmaceuticals containing stan- (Baaijens, 1986; Goh, 1985; Lewis and Emmett, 1987).
nous chloride as a reducing agent (Dantas et al., 2002). Dermatitis and irritant and allergic reactions have been
reported among Italian agricultural and ex-agricultural
workers using triphenyltin-containing pesticide for-
7.2  Organotin Compounds mulations (Lisi et al., 1987).
7.2.1  Local Effects and Dose-Response Relationships
7.2.1.2 Animals
7.2.1.1 Humans Acute oral LD50 values for tributyltin in mice and
Several incidents were reported when residents and rats range from 44 to 234 mg/kg b.w., but LD50 val-
workers were exposed to walls and joists treated with ues for intravenous and intraperitoneal injections are
tributyltin oxide wood preservative. The symptoms of much lower (Funahashi et al., 1980; Schweinfurth,
56 Tin 1269

1985; Pelikan and Cerny, 1968; Polster and Halacka, in fish exposed to tributyltin at 20 μg/L for 5 days
1971). For rats, the intraperitoneal LD50 value is decreased from 52.5 to 42.0%. Exposure of young rats
10 mg/kg and for mice the intravenous LD50 is esti- to low doses of tributyltin oxide resulted in functional
mated to be 6 mg/kg (Poitou et al., 1978; Truhaut alterations to the immune system (­Smialowicz et  al.,
et al., 1976). In rats, LD50 values for oral tributyltin 1989). Short-term exposure to tributyltin reduced
administration range from 94 to 224 mg/kg, and LD50 weight and caused morphological changes in lym-
values in mice range from 46 to 230 mg/kg. A single phoid tissues, particularly the thymus, associated
dose of tributyltin at 500 mg/kg caused numerous with a reduction in circulating lymphocyte numbers
hemorrhagic lesions in the digestive tract in mice and total serum immunoglobulin levels (Penninks,
(Pelikan and Cerny, 1968). Signs of toxicity such as 1993). Both triphenyltin and tributyltin have been
anorexia, emesis, tremor, and diarrhea have been shown to induce thymus atrophy and to suppress
observed after a single oral administration of triphe- T-cell-induced immune responses (Seinen et al., 1979;
nyltin. The oral LD50 for triphenyltin in rats ranges Snoeij et al., 1985, 1988). Tributyltin at a dietary level
from 140 to 300 mg/kg b.w., and is 81-93 mg/kg b.w. of 20-320 mg/kg caused thymus weight loss and atro-
for mice, whereas dermal the LD50 is 350 mg/kg phy, leading to a reduction in peripheral lymphocytes
b.w. in mice and over 2000 mg/kg b.w. in rats (WHO, and T cells in the spleen (Krajnc et al., 1984). It has been
1992). For oral ingestion of triphenyltin, the LD50 proposed that thymus atrophy is related to inhibition
for mice, guinea pigs, and rabbits is estimated to be of immature thymocyte proliferation and thymocyte
80-1000, 10-41.2, and 30-140  mg/kg, respectively. apoptosis (Gennari et al., 1997; Grundler et al., 2001;
An acute dose of triphenyltin (50 mg/kg) produces Baken et al., 2007). Additionally, tributyltin represses
marked hyperglycemia in hamsters, but this was not mitochondrial function and immune cell activation.
observed when hamsters were fed diets containing Triphenyltin intraperitoneally injected into mice for
phenyltin concentrations below 60 mg/kg (Matsui 14 days inhibited T-cell-dependent humoral and cel-
et al., 1984; Ohhira and Matsui, 1996; Ohhira et al., lular immune responses (Nishida et al., 1990). It was
1996). The hyperglycemic action of triphenyltin may shown that mice and other species appear to be more
be an effect of its dose-dependent accumulation in resistant than rats to the effects of ingested organo-
the pancreas. In rats, neither tributyltin nor dibutyltin tin compounds on the thymus. Thus, in rats, dietary
caused any abnormalities in the intestinal tract, despite dibutyltin, at concentrations as low as 20 mg/kg,
the fact that tributyltin is a potential inducer of intes- caused thymus atrophy within 2 weeks (Seinen et al.,
tinal heme oxygenase (Krajnc et al., 1984; Rosenberg 1977b). Oral administration of tributyltin chloride at
et al., 1984). However, atrophy of hepatocytes in the 20 mg/kg for 28 days caused weight loss of both the
centrilobular region has been noted in liver at 80 mg/ thymus and spleen accompanied by reduced prolif-
kg and 320 mg/kg (Krajnc et al., 1984). Feeding rats erative activity of T lymphocytes and increased thy-
with dibutyltin caused inflammation and damage to mocyte apoptosis in mice (Chen et al., 2011). Decrease
the bile duct (Gaunt et al., 1968; Krajnc et al., 1984). in weight and cellularity of the thymus has been seen
Repeated exposure to dibutyltin at 50 mg/kg for in rats fed with 50-150 mg/kg dibutyltin dichloride
3 days produced severe damage of the bile duct, char- or 150 mg/kg dioctyltin dichloride (Seinen et al.,
acterized by complete deterioration of the luminary 1977a). The lower molecular weight diethyltin and
epithelium, mainly in the pancreatic regions of the dipropyltin compounds produced less pronounced
bile duct (Barnes and Magee, 1958). The reported oral effects, and both octyltin and dioctadecyltin did not
LD50 for trimethyltin in rats was 14.7 mg/kg b.w. and have a measurable impact on the immune system
13 mg/kg b.w. for rats; the LD50 values for female and (Seinen et al., 1977b). The inter-species differences in
male mice were estimated to be 3.16 and 4.64, respec- susceptibility to these compounds were attributed to
tively (Tang et al., 2013b; Hoch, 2001). Oral LD50 val- differences in their gastrointestinal absorption and
ues for dimethyltin were calculated to be between metabolism (Boyer, 1989). The effects of dialkyltin
147 and 383 mg/kg b.w. for rats and between 216 and compounds on the thymus are reported to be due to
316 mg/kg b.w. for mice (Tang et al., 2013b). their interference with thymocyte proliferation via
Several studies suggest that the immune system is cytostatic rather than cytotoxic mechanism (Penninks
the target organ for organotin toxicity (Snoeij et al., et al., 1985; Seinen et al., 1977b; Boyer, 1989). Tribu-
1985; Vos and Krajnc, 1983). Exposure to tributyltin is tyltins, in contrast to dialkyltins, induced immunode-
reported to decrease the proportion of neutrophils and ficiency through direct action on lymphocytes in the
diminish respiratory burst activity in Japanese floun- thymus (Vos et al., 1984). The reported NOAEL for
ders (Paralichthys olivaceus) (Nakayama et al., 2007). dioctyltin immunotoxicity is 0.23 mg/kg b.w./day,
The neutrophil proportion of the total leukocytes based on the incidence of thymus lymphoma after
1270 Elena A. Ostrakhovitch

exposure for 2 years (Dobson et al., 2006). A 3-month in human and rat neurons. The cytotoxic potency of
exposure period resulted in a NOAEL of 0.87 mg/kg trimethyltin increased 70-fold in human cortical neu-
b.w./day based on decreased thymus weight. Dioc- rons 5 days after exposure. Rat hippocampal neurons
tyltin was reported to cause thymus atrophy and were the most vulnerable to trimethyltin, exhibit-
dysfunction of the T-lymphocyte-mediated immune ing a LC50 value of 1.4 μM; in contrast, a LC50 value
response (Miller et al., 1986; Seinen et al., 1979). of 44.28 μM was calculated for rat cerebellar granule
Oral exposure to dioctyltin chloride for 6 weeks at cells. Behavioral disturbances were observed after
150 mg/kg was shown to increase relative liver and exposure to 10 mg/kg triethyltin (Reiter et al., 1980).
kidney weights (Seinen and Willems, 1976). In rats A diminished startle response to a puff of air or sud-
fed dioctyltin chloride from postnatal day 10, kidney den noise proved to be a sensitive indication of tri-
weight decreased at postnatal day 21, with a further ethyltin exposure (Squibb et al., 1980). Tributyltin
increase of 11 and 5% on postnatal days 21 and 90, given as a single dose at 10-60 mg/kg to rat pups on
whereas relative liver weight showed a tendency to postnatal day 5 caused persistent alterations in motor
increase at postnatal day 21 without further changes activity development and the acoustic startle response
(Tonk et al., 2011c). The developing immune system (Crofton et al., 1989). Alterations in brain weight and
is reported to show greater sensitivity to the immu- brain histology have been found after intraperitoneal
nosuppressive effects of dioctyltin compared with injection of tributyltin to 5-day-old rats (O’Callaghan
the fully developed immune system (Smialowicz and Miller, 1988).
et al., 1988). Oral administration of dioctyltin chlo- Oral administration of 2.5 and 5 mg/kg dibutyltin
ride to rats from postnatal day 10 decreased white in olive oil 3 days per week to pregnant rats increased
blood cell concentrations and lymphocyte counts at the incidence of apoptotic cell death in the neocortex
postnatal days 42 and 70 in dose-dependent manner and hippocampus of offspring at postnatal day 38.
(Tonk et al., 2011a,c). Thymus weight, thymus cellu- The NOAEL for maternal toxicity of dibutyltin was
larity, and relative thymic cell count were decreased reported to be 1 mg/kg b.w. and 5 mg/kg b.w. by dif-
at all time points evaluated, with the largest decrease ferent research groups (ORTEPA, 1994; Ema et al.,
of 74% observed at postnatal day 21. While effects on 1991b). Male rats exposed to 10 mg/kg per day of
lymphocyte subpopulations in the thymus were only tributyltin for 10 days exhibited histological altera-
observed in the 30-mg/kg group at postnatal day 42, tions in seminal vesicles and epididymis, as well as
effects on lymphocyte subpopulations in the spleen reduced sperm counts. Lumen formation in the semi-
were found in the 30-mg/kg group at postnatal niferous tubule was remarkably delayed in response
days 42 and 70 (Tonk et al., 2011b). The delayed-type to a single administration of 25, 50, or 100 mg/kg
hypersensitivity response was seen at a dose of 3 mg/ tributyltin (Kim et al., 2008). Reduced serum testos-
kg. Hematological changes such as anemia, lympho- terone concentration and downregulated expression
penia, and thrombocytosis have been noted in rats of 3-beta-hydroxysteroid dehydrogenase (3β-HSD II)
fed tributyltin at a dose of 50 mg/kg (Wester et al., and estradiol 17-beta-dehydrogenase 1 (17β-HSD 1)
1990). It has also been shown in vitro that organotin were associated with reduced male fertility. Wistar
compounds decrease the survival, proliferation, and females receiving 100 ng/kg tributyltin per day for 16
differentiation of isolated human B lymphocytes (De days by gavage had decreased cycle regularity, dura-
Santiago and Aguilar-Santelises, 1999). In another tion of the reproductive cycle, proestrus and diestrus
study, tributyltin was shown to decrease the percent- phases, and number of epithelial cells in the proestrus
age of the CD19+CD22+ B cell subset in human bone phase (Podratz et al., 2012). Administration of phen-
marrow cell cultures (Carfi et al., 2010). yltins during early pregnancy led to implantation fail-
Triethyltin-induced neurotoxicity in the brain ure and increased resorption, and ­post-implantation
causes the formation of myelin edema and the loss of loss in rats (Noda et al., 1992b; Ema and Miyawaki,
both axons and myelin. Myelin deficits produced by 2002; Adeeko et al., 2003). Oral administration of
early postnatal exposure to triethyltin are permanent triphenyltin chloride at 2 or 6 mg/kg b.w. caused
and cannot be repaired as the brain matures. There- an increase in the number of oocytes at all follicle
fore, triethyltin is more toxic than trimethyltin to the stages at both dose levels, whereas no histological
CNS of rodents. The LC50 values for trimethyltin were changes were observed in the oviduct, uterus, vagina,
estimated to be 335.5 and 609.7 μM for human pri- and mamma (Watermann et al., 2008). Exposure to
mary neurons and astrocytes, respectively, after 24 h 2 mg/kg triphenyltin led to a significant reduction in
of exposure (Cristofol et al., 2004). With the exception the diameter of tertiary follicles, whereas the number
of rat hippocampal neurons, triethyltin was more toxic of atretic follicles was increased in tertiary and pre-
than trimethyltin, showing LC50 values of 3.5-16.9 μM ovulatory follicles.
56 Tin 1271

7.2.2  Systemic Effects and Dose-Response of micrograms per liter. The tendency of organotin
Relationships compounds to accumulate in organisms depends on
its partition between lipid and aqueous phases. The
7.2.2.1 Human octanol-water partition coefficient (Kow) for tributyltin
No human studies are available on chronic low-level indicates a potential for bioaccumulation: the log Kow
exposure to organotin compounds. However, some case values range from 3.2 to 4.1 but can be modified by
reports describe various health effects after accidental salinity (Laughlin et al., 1986). Studies in algae, aquatic
exposure to tributyltin and triphenyltin compounds. invertebrates, and fish have confirmed that tributyl-
Patients who had been exposed mainly through cuta- tin bioaccumulation in these organisms is substantial.
neous absorption developed acute nephropathy and Bioconcentration factor values range up to 10,000 in
disorders of the CNS (Prull and Rompel, 1970; Colosio periwinkles, 50,000 in fish, and 500,000 in clams. Expo-
et al., 1991; Grace et al., 1991; Manzo et al., 1981; Wax sure to organotins such as tributyltin and triphenyltin
and Dockstader, 1995). Exposure to triphenyltin causes results in imposex. The imposition of male sexual char-
spontaneous involuntary movements of the hands, acteristics on female gastropod mollusks was linked to
facial twitching, and crying. Some of the patients expe- the androgen signaling pathway because simultane-
rienced diplopia, drowsiness, giddiness, vertigo, bidi- ous injection of tributyltin with an androgen receptor
rectional nystagmus, impairment of numerical ability, inhibitor, cyproterone acetate, diminished masculin-
and disorientation in time and place (Lin et al., 1998). ization in this species (Stange et al., 2012). Imposex
In addition to CNS abnormalities, delayed peripheral develops experimentally in mud snails (Nassarius
neuropathy, hepatitis, and leucopenia have been moni- obsoletus) after 60 days exposure to tributyltin at con-
tored 6 and 9 days after triphenyltin consumption. centrations of 4.5-5.5 mg/L.
Two case reports of triphenyltin poisoning indicated With regard to dibutyltin toxicity, it was shown that
leucopenia, decreased neutrophil responsiveness, the acute half maximal effective concentration (EC50)
elevated hepatic enzymes, hepatomegaly, and CNS for growth inhibition in the diatom Skeletonema costa-
effects (encephalopathy and delayed neuropathy) tum is 30 μg/L, whereas at 48 h the EC50 was calculated
(Colosio et al., 1991; Lin et al., 1998). Liver damage has to be 17 μg/L for swimming inhibition in the crustacean
been reported in people using triphenyltin acetate as Daphnia magna, with an LC50 of 980 μg/L for fish spe-
a spray (Horacek and Demcik, 1970). More than 100 cies. Evidence of disruption of the endocrine system,
deaths and over 200 cases of illness occurred in France e.g. the induction of imposex, is seen at 0.5 ng Sn/L
in 1954 due to ingestion of a preparation containing in dogwhelks. Some marine benthic invertebrates are
diethyltin diiodide and triethyltin monoiodide. Doses also very sensitive to tributyltin in sediments. Popula-
of diethyltin were estimated to be 45-675 mg in non- tions of benthic invertebrates such as polychaetes and
fatal cases and 380-675 mg in fatal cases (Barnes and amphipods have been shown to be reduced as a result
Stoner, 1959). Severe headaches, vertigo, visual abnor- of exposure to tributyltin in sediments. Exposure to tri-
malities, paralysis, and convulsions were reported butyltin leads to masculinization in some fish species
after only a few days of exposure. Death occurred from (Shimasaki et al., 2003). In male and female mammals,
coma and respiratory or cardiac failure (Gruner, 1958). organotins also affect the reproductive system but do
Pronounced edema of white matter of the brain was not alter sex ratios (Omura et al., 2001; Ogata et al.,
seen in fatal cases. 2001). Exposure of zebrafish embryos to trimethyltin
up to 72 h postfertilization was shown to result in mal-
7.2.2.2 Animals formation with an EC25 of 5.55 μM (Chen et al., 2011).
While the inorganic forms of tin are generally consid- Exposure of zebrafish embryos to various organotin
ered nontoxic, its organic derivatives exhibit a complex compounds during very early development (< 100 h
pattern of toxicity. The biological effects of organotin postfertilization) revealed that dibutyltin dichloride
species are mostly dependent on the number and kind is the most potent (developmental) neurotoxic com-
of organic moieties bound to the tin atom, with trisub- pound (Beker van Woudenberg et al., 2013). Embryos
stituted compounds being the most toxic. Trisubsti- exposed to dibutyltin at a concentration of 8 μM had
tuted organotin compounds are toxic to many aquatic reduced hatching rates; in contrast, dimethyltin
organisms, including fish. In some fish, acute toxic- dichloride showed effects only at high concentrations
ity occurs at a few milligrams per liter, while chronic of 600 μM. Octyl derivatives (monooctyltin trichloride
toxicity can be found at concentrations in the order and dioctyltin dichloride) were not identified as devel-
of micrograms per liter. These compounds are also opmental toxicants due to their low solubility.
highly toxic to mollusks, with chronic toxicity in oys- Tributyltin oxide is known to disrupt the endocrine
ters and clams occurring at concentrations of fractions and reproductive system in rats. Maternal exposure to
1272 Elena A. Ostrakhovitch

organotin compounds such as triphenyltin and dibu- Dibutyltin is neurotoxic and exposure of rats during
tyltin caused embryonic death or suppressed embry- development causes an increased incidence of apop-
onic growth (Tryphonas et al., 2004). Reduction of fetal totic cell death in certain brain tissues (Jenkins et al.,
ossification was observed at doses that are nontoxic to 2004). Furthermore, exposure to trimethyltin dur-
the mother. Intraperitoneal injection of tributyltin to ing development affected the memory and impaired
pregnant C57BL/6 mice, providing a dose of 0.05 or learning capacity (Jenkins and Barone, 2004), as well
0.5 mg/kg b.w., from gestational day 12 to gestational as having toxic effects on testes development in mice
day 18 led to increased lipid accumulation in the pups (Kumasaka et al., 2002).
(Grun and Blumberg, 2006). Organotin compounds Numerous studies have reported teratogenic effects
were shown to cross the placental barrier and accu- of dibutyltins. Dibutyltins have been shown to cause
mulate in large quantities in placental and fetal tissues malformation of rat fetuses after oral administration to
(Cooke et al., 2008; Golub and Doherty, 2004). Accu- pregnant rats on day 8 of gestation (Noda et al., 1992a,
mulation of organotins in the placenta was five times 1993, 1994). Dibutyltin at 7.6 mg/kg b.w. and above
higher than in the dam’s blood, whereas the level in caused implantation failure in rats exposed for up to
pups was half of that found in the placenta. The lev- 3 days (Ema et al., 2003). Dibutyltin affects the normal
els of tributyltin compounds in the liver and brain tis- development of embryos during embryogenesis and
sues of pups declined by days 6 and 12 after birth even its dysmorphogenic potential varies with develop-
though they continue to ingest organotins with milk. mental stage. The NOAEL for fetus malformation was
The toxic effects of tributyltin throughout the repro- defined as 45 mg/kg b.w.
ductive cycle were attributed to a marked decrease in Trimethyltins are the most toxic trialkyltins. These
ovary weight and in serum 17β-estradiol levels. Fur- compounds are neurotoxic, induce pathological lesions
thermore, a significant increase in progesterone level in the mammalian brain, and lead to neurological and
and an accumulation of apoptotic cells in the corpus behavioral changes associated with hypothermia and
luteum and granulosa cell layer contributed to the toxic inhibition of brain protein synthesis (Geloso et al.,
effect. Tributyltin significantly increased the baseline 2011; Chang, 1986a, 1986b; Costa and Sulaiman, 1986).
coronary perfusion pressure and impaired vasodila- Neurotoxicity of tributyltin compounds has been
tion via suppression of 17β-estradiol accompanied by a shown in various systems. Intraperitoneal adminis-
significant rise in serum progesterone after 100 ng/kg tration of tributyltin at 2, 3, and 4 mg/kg on postna-
tributyltin was orally administered daily to rats for 15 tal day 5 caused dose-dependent decreases in brain
days (Dos Santos et al., 2012). Furthermore, tributyltin weight (O’Callaghan and Miller, 1988). A single acute
promoted collagen deposition in the heart interstitium exposure to nonlethal doses of tributyltin chloride
and induced endothelium denudation and platelet (50 mg/kg) led to significant changes in rat behavior
accumulation. Treatment with triphenyltin chloride at (Ema et al., 1991a). Dietary administration of tributyl-
6 mg/kg b.w. caused a high mortality rate in dams and tin chloride (125 mg/kg) for 30 days inhibited ligand
decreased the survival rate of offspring (Grote et al., binding to the N-methyl-d-aspartate (NMDA) receptor
2007). A considerable number of pups from surviving in mice, leading to depressed NMDA receptor func-
dams died up to postnatal day 4. In utero treatment tion associated with an array of negative symptoms
with 2 mg/kg triphenyltin also caused early postna- (Konno et al., 2001). Symptoms of triethyl poisoning
tal mortality up to postnatal day 4 (Grote et al., 2009). in rodents include weakness of hindlimbs, dyspnea,
The surviving offspring developed sex-dependent peripheral vasodilation, and transient edema of central
differences in response to developmental exposure and peripheral nervous systems (Reuhl and ­Cranmer,
to triphenyltin: the body weight of female offspring 1984). These compounds also cause hemolysis of
was unaffected, whereas male offspring exhibited erythrocytes (Ali et al., 1987). Alkyl radicals have been
a decrease in body weight after the end of lactation. shown to damage the erythrocyte membrane. A single
Similarly, a decrease in body weight gain was reported trimethyltin injection resulted in neuronal death in
after tributyltin administration at similar dose levels the hippocampus (Balaban et al., 1988; Chang, 1986a;
(Kimura et al., 2005; Makita and Omura, 2006). At high O’Callaghan et al., 1989). Toxic doses of trimethyltin
doses of triphenyltin (6 mg/kg and 10 mg/kg b.w.), were higher for rats than for mice, hamsters, or mar-
delayed development of the vaginal opening was mosets. Both male and female rats developed seizures
observed in female offspring (Grote et al., 2006). Water- with symptoms including convulsion, head tremors,
mann et al. reported that triphenyltin chloride treat- and salivation (Tang et al., 2013b). In mice, trimeth-
ment of adult or peripubertal female rats at doses of yltin severely affects dentate gyrus granule cells. In
2 mg/kg or higher increased the incidence of follicular rats, effects of trimethyltin are restricted to pyrami-
atresia (Watermann et al., 2008). dal neurons in cornu ammonis (CA) areas CA1-CA3.
56 Tin 1273

Trymethyltin (7  mg/kg, intraperitoneally) reduced dibutyltin dichloride-induced thymus atrophy in rats,
neurotransmission at the CA3-CA1 synapse and resulting from the depletion of small CD4+CD8+ thy-
decreased cell numbers and the width of the CA1 pyra- mocytes (Pieters et al., 1994b). In addition, organotin
midal cell layer in the brain of Wistar rats (Gasparova compounds have been reported to induce genotoxicity
et al., 2012). Neurodegenerative effects of trimethyltin and immunosuppression in aquatic organisms (Hagger
were shown to be associated with the production of et al., 2005). Tributyltin may exert endocrine, bio-
inflammatory cytokines and oxidative stress (Harry transformation, and lipid peroxidative effects through
et al., 2008; Maier et al., 1995; Shuto et al., 2009; Corvino modulation of cyclic AMP/protein kinase A second
et al., 2011). Triethyltins also affect protein phosphory- messenger signaling, with overt physiological conse-
lation in the rat brain. Neuronal death was shown to be quences (Pavlikova et al., 2010). Tributyltin was shown
delayed for 2 days after a single intraperitoneal injec- to modulate multiple hepatic responses in salmon,
tion of trimethyltin but then developed progressively including effects on peroxisome proliferator-activated
over the course of 3 weeks (Whittington et al., 1989). receptors (PPARs), CYP19 isoforms, CYP3A, estrogen
Neonatal exposure to trimethyltin delayed learning in receptor alpha (ERα), and nuclear receptor subfamily 1
preweanling rats; this was associated with a reduction group I member 2/pregnane X receptor (PXR) at both
in neuronal viability and a significant decrease in the the transcriptional and translational levels.
expression of the dopamine receptors in the hippocam- Organotin compounds, especially tributyltin and
pal area (Mignini et al., 2012). Tributyltins were shown triphenyltin, were shown to be endocrine disrupt-
to be neurotoxic, causing impaired learning ability and ers (Bryan et al., 1989; Oberdorster and McClellan-
memory loss in rats (Elsabbagh et al., 2002). Compari- Green, 2002; Golub and Doherty, 2004; Horiguchi,
son of the toxicity of monomethyltin, dimethyltin, and 2009). Tributyltins are obesogens: they were shown
dibutyltin compounds to the known neurotoxicant to promote adipogenesis in murine 3T3-L1 preadipo-
trimethyltin in an in vitro model of neuronal devel- cyte cells and to induce the differentiation of multipo-
opment in PC12 cells showed that dimethyltin is less tent mesenchymal stem cells into adipocytes (Inadera
potent than trimethyltin and that monomethyltin is and Shimomura, 2005; Grun et al., 2006; Kirchner
not toxic at any concentration examined (Jenkins et al., et al., 2010; Carfi et al., 2008). Tributyltin compounds
2004). The toxicity of organotins correlated with their were shown to activate PPARs/retinoic acid recep-
lipophilicity. Dibutyltin has the highest octanol-water tors (RXRs), a transcriptional complex that plays a
partition coefficient (log Kow = 1.9), followed by tri- critical role in energy balance, including effects on tri-
methyltin (log Kow = 0.3), dimethyltin (log Kow = −1.1) glyceride metabolism and both fatty acid and glucose
and monomethyltin (log Kow = −1.29). Tributyltin and homeostasis (Grun and Blumberg, 2006; Kanayama
dibutyltin, but not monobutyltin, were shown to sup- et al., 2005). Tributyltin induced activation of adipo-
press acetylcholine synthesis in slices of the cortex and genesis and adipogenic commitment in mesenchymal
to inhibit the activity of choline acetyltransferase, thus stem cells is via activation of PPAR and suppression
affecting both high-affinity and low-affinity uptake of of RXR function. Furthermore, tributyltin has a high-
choline into synaptosomes (Kobayashi et al., 1996). All binding affinity to RXR via bond formation between
three butyltins at concentrations from 10 μM to 100 μM butyltin and the Cys-432 residue of RXR (Le Maire
had no effect on the activity of acetylcholinesterase. et al., 2009). Activation of RXR has been implicated in
Organotin compounds exert various toxic effects on the development of imposex in gastropods (Sternberg
the thymus, liver, and immune system (Vos et al., 1990; et al., 2008; Urushitani et al., 2011). It was suggested
Pieters et al., 1994a; Grinwis et al., 2009; O’Callaghan that RXR activation by organotins is induced in the
and Miller, 1988; Yoshizuka et al., 1992; Tsunoda et al., penis-forming area behind the right tentacle of female
2004). In short-term feeding studies in rats, tributyltin T. clavigera (Horiguchi et al., 2012). Gene enrichment
ingestion caused thymus atrophy, decreased numbers analysis revealed that transcripts involved in biologi-
of lymphocytes in spleen and lymph nodes, increased cal processes including general metabolism, immune
serum immunoglobulin M (IgM) levels, and decreased response, lipid metabolism, and stress response are
serum IgG levels (Krajnc et al., 1984; Snoeij et al., responsible for the development of imposex in the
1987). In mice, tributyltins reduced spleen weight and Queen conch (S. gigas) (Titley-O’Neal et al., 2013) Tri-
decreased the number of leukocytes (Ishaaya et al., butyltin induces adipogenesis, triglyceride storage,
1976). Mice receiving a 100-mg/kg tributyltin diet for and the expression of adipogenic marker genes in 3T3-
30 days had a decreased number of lymph node cells. L1 cells in a PPAR-dependent manner (Li et al., 2011).
The production of IFN-γ, IL-3, IL-4, and IL-5 was also Lipid plasma triacylglycerols, cholesterol, and lipases
reduced, indicating immunosuppressive effects of tri- exhibited monotonic increases with enhanced body
butyltin on mice (Iwamura et al., 2006). Exposure to weight in response to tributyltin (Meador et al., 2011).
1274 Elena A. Ostrakhovitch

Acute tributyltin and triphenyltin toxicity induces cellular effects, including disorganization of the cyto-
hyperglycemia without abnormalities in islet tissue, skeleton, suppression of enzyme activity, and altered
indicating that organotins have a direct effect on adi- expression of proteins involved in protein synthesis,
pose tissue (Inadera and Shimomura, 2005). Acute cellular trafficking, and proliferation (Osman et al.,
triphenyltin toxicity induces hypertriglyceridemia in 2009). Thus, tributyltin oxide was shown to suppress
the hamster (Ohhira et al., 1999a). Dialkyltins, such transcription factors involved in energy homeostasis,
as dibutyltin dichloride, induce acute intestinal pan- including PPARα/β (Colliar et al., 2011). Organotins
creatitis in rats at approximately half the lethal dos- disrupt mitochondrial energy metabolism and block
age of 6 mg/kg b.w. (Merkord et al., 1998). Dibutyltin mitosis by affecting spindle formation (Penninks et al.,
induces the development of chronic pancreatitis in ani- 1983; Raffray and Cohen, 1991; Jensen et al., 1991).
mal models, which imitates progression from the acute However, the mechanism through which organotin
to the chronic stage. A single intravenous application compounds affect so many cellular processes remains
of dibutyltin at a dose of 8 mg/kg resulted in a high unclear.
concentration in the bile, followed by necrosis of bilio- The toxic effects of organotins have been attributed
pancreatic duct epithelial cells, chronic inflammation, to changes in calcium homeostasis (Corsini et al., 1998;
and acute interstitial pancreatitis (Merkord et al., 1997, Gennari et al., 2000). Trisubstituted organotin com-
1998). In rats, dibutyltin dichloride-induced chronic pounds, such as tributyltin, triphenyltin, trimethyltin,
pancreatitis is used as an experimental model for the and triethyltin, increase calcium flux in mitochondria,
observation of pancreatic edema, inflammatory cell thus altering ATPase activity and ATP synthesis (Miura
infiltration, and fibrosis (Stange et al., 2003; Yamashita et al., 1997; Viviani et al., 1995; Gasso et al., 2000; Nesci
et al., 2007). Abnormal biliopancreatic duct fibrosis, et al., 2011a,b). Tributyltin compounds were shown to
acinar degeneration, and inflammatory cell infiltra- increase intracellular calcium levels in rat thymocytes,
tion were observed in tributyltin-treated rats 28 days Jurkat (a human T lymphocyte cell line) cells, B lym-
after administration (Zhang et al., 2010; Glawe et al., phocytes, and NK cells (Grundler et al., 2001; Lane
2005). Apoptosis of single acinar cells was observed et al., 2009; Bissonnette et al., 2010). The function of
in dibutyltin-induced pancreatitis. Fibrosis in dibutyl- ATPase activity and voltage-gated calcium channels is
tin-treated rats peaked after 1 week and was limited affected by tributyltin, resulting in suppressed calcium
to areas around the pancreatic ducts after 2 weeks; flow from the cytoplasm to the endoplasmic reticulum
the fibrosis was composed of both type I and type III (ER). Perturbation in calcium homeostasis was shown
collagen (Miyauchi et al., 2007). Overexpression of to destabilize the interaction between calmodulin
transforming growth factor-beta receptor-associated (CaM) and its interaction partners, including matrin-3,
protein 1 (TGF-A1/TRAP1) was shown to contribute which is a substrate of caspase-3 and caspase-8
to the pancreatic fibrogenesis in rats. (Osman and van Loveren, 2012). Downregulation of
ribosomal protein S6 kinase alpha-1 (S6K-α1), a down-
stream effector of the serine/threonine-protein kinase
7.3  Mechanism of Action
mTOR/mammalian target of rapamycin pathway and
Organotin compounds are highly lipophilic, and target of AMP-activated protein kinase (AMPK), was
can therefore permeate across biological membranes; implicated in inhibiting protein synthesis in the pres-
they target the plasma and mitochondrial membranes. ence of tributyltin oxide (Osman et al., 2009; Osman
It was shown that organotins affect the lipid bilayer and van Loveren, 2012). A dose- and time-dependent
by altering membrane fluidity and decreasing plasma interaction of tributyltin with CaM was also reported
membrane potential, which leads to breakdown of (Cima et al., 2002). This interaction was hydrophobic
the plasma membrane (Boyer, 1989). Trialkyltin com- in nature, mediated by the aliphatic chains of tribu-
pounds inhibit oxidative phosphorylation in mito- tyltin and the hydrophobic regions of Ca2+-activated
chondria in vitro (Matsui et al., 1983). ATP levels are CaM. Both di- and monobutyltin were less capable
severely depleted in response to tributyltin oxide of inducing conformational changes in CaM, with no
and tributyltin 3-hydroxyflavone exposure (Usta and significant differences between the two compounds.
Griffiths, 1992). Organotin compounds inhibit both Triethyltin at 50 μM rapidly increases the intracellular
ATP hydrolysis and ATP synthesis catalyzed by mem- calcium level in human osteoblasts via stimulation of
brane-bound and isolated complex V (F0F1 complex). extracellular calcium influx and intracellular calcium
Organotin compounds react noncovalently with ATP release (Lu et al., 2003). However, the triethyltin- and
synthase, and the inhibitory effect of these compounds trimethyltin-induced calcium influx was not sensi-
is reversed by dithiothreitol or mercaptoethanol. In tive to voltage-gated calcium channel blockers. More-
addition, organotin compounds exert several other over, triethyltin was shown to be more effective than
56 Tin 1275

trimethyltin in increasing intracellular calcium con- 2008). Tributyltin can be neurotoxic through the pro-
centration in human and rat neurons (Cristofol et al., duction of peptide hormones in the brain (Santos et al.,
2004). 2005). Activation of M-calpain, together with induc-
A prolonged calcium increase affects diverse cellu- tion of ER-mediated apoptosis, has been described in
lar processes such as protein activation, gene expres- rat hepatocytes and PC12 cells exposed to tributyltin
sion, ion transport, and transmitter secretion and may (Nakatsu et al., 2006; Grondin et al., 2007). Tributyltin
lead to cell death. Increased calcium efflux from the at concentrations ranging from 30 to 100 nM reduced
ER to the cytoplasm and activation of calpain-2 cata- the frequency of GABAergic miniature postsynaptic
lytic subunit (M-calpain), together with induction of currents in immature neurons and decreased synapse
nuclear factor-kappa-B (NF-κB/p65) and activation numbers via disruption of Cl homeostasis in embry-
of nuclear factor of activated T cells (NFAT) and its onic cortical neurons (Yamada et al., 2010). However,
relocation to the nucleus, were reported to result in there were no changes recorded in gamma-aminobu-
T-cell activation and apoptosis in response to tribu- tyric acid (GABAA) receptor function in response to
tyltin oxide exposure (Osman and van Loveren, 2012; tributyltin. Trimethyltin and triethyltin, but not tri-
van Kol et al., 2012). Low concentrations of tributyl- butyltin, were shown to induce activation of primary
tin (0.5 μM) caused the release of cytochrome c from microglial cells purified from neonatal rat brains in the
isolated liver mitochondria, which is indicative of the presence of astrocytes (Roehl et al., 2009). Triethyltin-
induction of mitochondrial permeability transition induced calcium overload was implicated in medi-
and apoptosis (Gogvadze et al., 2002). Both tributyltin ating apoptosis in PC12 cells exposed to triethyltin
and triphenyltin suppressed DNA and protein synthe- (Viviani et al., 1995). Diorganotin compounds were
sis in choriocarcinoma cells (Nakanishi et al., 2002). also shown to induce apoptosis in PC12 cells, with a
At concentrations above 1 μM, tributyltin compounds displayed potency of cytotoxicity in the order: dibu-
inhibited activities of 3β-HSD II and 17β-HSD 1 (Ohno tyltin → diphenyltin → dimethyltin (Liu et al., 2013).
et al., 2005). Components of the cellular cytoskeleton Triethyltin was cytotoxic for oligodendrocytes, the
are also targeted by organotins (Cima et al., 1998). Tri- myelin-forming cells, and led to apoptotic cell death,
butyltin and triphenyltin compounds inhibit microtu- as indicated by DNA fragmentation along with con-
bule assembly through direct interaction with tubulin densed and fragmented nuclei (Stahnke and Richter-
(Jensen et al., 1991). The toxic effects of triphenyltin Landsberg, 2004). Upregulation of HO-1, an indicator
were linked to depolymerization of filamentous actin of oxidative stress, as well as activation of extracellular
in thymocyte (Chow and Orrenius, 1994). The effect of signal-regulated kinases (ERKs), was observed in oli-
tributyltin and dibutyltin on chondrogenesis varied godendrocytes treated with 0.2-1.5 μM triethyltin for
depending on the concentration of organotin applied. 24 h. Trimethyltin toxicity was linked to secretion of
Organotins at a low concentration of 0.016 ppb slightly Cl− (Yu et al., 2010). Exposure to trimethyltin increased
increased human chondrocyte proliferation and dif- Cl− secretion in a dose-dependent manner in rat colonic
ferentiation, while these compounds demonstrated mucosa. Cl− secretion was essentially regulated by
strong inhibition of chondrogenesis at concentrations basolateral calcium-sensitive K+ channels in response
of 0.16 and 7.5 ppb (Banu et al., 2006). to trimethyltin. The LC50 for tetramethyltin in CaCo
Organotin compounds were shown to affect the cells was 170.7 μmol/L. In CHO-9 and HepG2 cells,
CNS, disrupt components of glutamate homeostasis tetramethyltin at concentration of 429.4 μmol/L and
in astrocytes, induce noradrenaline (norepinephrine) 161.7 μmol/L did not reduce cell viability below 50%
release from rat hippocampal slices and norepineph- (Dopp et al., 2011).
rine release in PC12 chromaffin cells, and induce apop- Tributyltin oxide inhibits cell proliferation and
tosis in neuronal cells (Mizuhashi et al., 2000; Yamada cell cycle progression, and induces apoptosis. Tri-
et al., 2010). Trialkyltin compounds are the most neu- butyltin treatment was shown to cause apoptosis in
rotoxic. While trimethyltin showed localized neuronal human amnion cells (Zhu et al., 2007). Suppression of
damage, triethyltin chloride showed a specific effect matrin-3, mitogen-activated protein kinase (MAPK)
on myelin formation. Exposure to trimethyltin dimin- signaling, and ribonucleotide reductase (all implicated
ished neuronal viability, accompanied by a significant in cell proliferation) was demonstrated in mouse thy-
decrease in the expression of dopamine receptors (D1 moma cells treated with tributyltin oxide (Osman and
and D2) and dopamine transporters (Mignini et al., van Loveren, 2012). Exposure to tributyltin resulted
2012). Several finding suggest that trimethyltin dam- in upregulation of genes involved in ER stress, NF-κB
ages mitochondria and induces apoptosis or necrosis and tumor necrosis factor (TNFα) pathways, and genes
in cultured cerebellar granule cell, depending on the involved in DNA damage, p53 signaling, and apopto-
concentration used (Misiti et al., 2008; Morita et al., sis in primary mouse thymocytes (van Kol et al., 2012).
1276 Elena A. Ostrakhovitch

Both tributyltin and triphenyltin at 1 μM induced phos- assay (Oshiro, 1991). These data indicate that organo-
phorylation and MAPK activation (Yu et al., 2000). tin compounds are potential mutagens.
Tributyltin organotins are known to induce immu- Despite intensive studies into the toxicity of organo-
nosuppression. They inhibit lymphocyte proliferation tin compounds, the molecular mechanisms underlying
and induce apoptosis in rat primary thymocytes, Jur- their toxicity and the molecular targets of organotins
kat cells, and platelets (Raffray et al., 1993; Grundler have not yet been fully elucidated.
et al., 2001; Berg et al., 2003). The percentage of early
apoptotic cells was increased by tributyltin oxide at
Acknowledgment
0.5 and 1.0 μM after 3 h of exposure; genes expressed
after 3 h were largely related to the apoptotic process I wish to dedicate this chapter to my teacher Dr M. George Che-
in primary rat thymocytes (Baken et al., 2007). It was rian and to the late Dr Magnus Piscator, who wrote the Tin chapter
in the earlier editions of this book.
suggested that tributyltin oxide exerts its toxic effects
on the thymus primarily by affecting apoptotic pro-
cesses. Exposure of Jurkat cells to 0.5 μM tributyltin References
oxide for 12 and 24 h resulted in reduced viability of
the cells to below 80% (Katika et al., 2011). Tributyltin ACGIH (American Conference of Industrial Hygienists), 2003. Tin.
In: Threshold limit values for chemical substances and physical
at concentrations of 0.2 μM or 119 ng/mL, which corre- agents and biological exposure indices. American Conference of
spond better to the butyltin concentration measured in Governmental Industrial Hygienists, Cincinnati, OH. 56.
human tissues, did not dramatically affect Jurkat cell Adams, W.A., Xu, Y., Little, J.C., et al., 2011. Environ. Sci. Technol.
viability. However, tributyltin at a low concentration 45, 6902–6907.
of 0.2 μM induced T-cell activation accompanied by ER Adcock, L.H., Hope, W.G., 1970. Analyst 95, 868–874.
Adeeko, A., Li, D.M., Forsyth, D.S., et al., 2003. Toxicol. Sci. 74,
and oxidative stress (Katika et al., 2012). Exposure of 407–415.
primary cultures of rat thymocytes to tributyltin oxide Afonso, D.D., Baytak, S., Arslan, Z., 2010. J. Anal. At. Spectrom. 25,
at a concentration of 1 μM for 3 and 6 h led to reduced 726–729.
cell viability (Baken et al., 2007). The major mode of tri- Aghaie, H., Giahi, A., Monajjemi, M., et al., 2005. Sensors Actuat.
butyltin action is suggested to be via targeting the ER. B-Chem. 107, 756–761.
Airaksinen, R., Rantakokko, P., Turunen, A.W., et al., 2010. Environ.
Organotin compounds were shown to be cytotoxic Res. 110, 544–547.
and genotoxic in several test systems (Hamasaki et al., Ali, A.A., Upreti, R.K., Kidwai, A.M., 1987. Toxicol. Lett. 38, 13–18.
1992; Florea et al., 2004; Florea et al., 2004). Dimethyltin Allner, B., der Goenna, S., Griebeler, E.M., et al., 2010. Environ. Sci.
was found to be the most toxic in hamster fibroblasts Pollut. Res. 17, 505–518.
(CHO-9 cells), followed by trimethyltin, tetramethyl- Almeida, A.C., Wagener, A.D.R., Maia, C.B., et al., 2004. Appl.
Organomet. Chem. 18, 694–704.
tin, and monomethyltin. Only dibutyltin, at the Alzieu, C., 2000. Sci. Total. Environ. 258, 99–102.
relatively high concentration of 1 mM, induced a sig- Alzieu, C., 2006. In: The Energy and Resource Institute. Teri Press,
nificant increase in chromosome aberrations and sister New Delhi, pp. 444–457.
chromatid exchange after a 1-h exposure. Trimethyl- Alzieu, C., Sanjuan, J., Deltreil, J.P., et al., 1986. Mar. Pollut. Bull. 17,
tin did not exert a genotoxic effect. All butyltins (tri, 494–498.
Alzieu, C., Sanjuan, J., Michel, P., et al., 1989. Mar. Pollut. Bull. 20,
di, and mono) were found to be genotoxic; moreover, 22–26.
dibutyltin was the most genotoxic, followed by tri-, Andersen, K.E., Petri, M., 1982. Contact Dermatitis 8, 173–177.
tetra-, and monobutyltin in the SOS chromotest (Kub- Antes, F.G., Krupp, E., Flores, E.M., et al., 2011. Environ. Sci. Technol.
alla et al., 1995). The greatest mutagenicity was shown 45, 10524–10530.
for tributyltin in the SOS chromotest with Escherichia Antizar-Ladislao, B., 2008. Environ. Int. 34, 292–308.
APHA (American Public Health Association), 1998a. Metals—­
coli PQ37, whereas there was no evidence of mutagen- electrothermal absorption spectroscopy, 3113B. American P ­ ublic
icity for dibutyltin and dimethyltin (Hamasaki et al., Health Association, Washington, DC. American Water Works
1992). However, dibutyltin dichloride, tributyltin chlo- ­Association, and Water Environment Federation.
ride, tributyltin oxide, dimethyltin dichloride, and APHA, 1998b. Metals—flame atomic absorption spectroscopy, 3111.
trimethyltin chloride were demonstrated to be as American Water Works Association, and Water Environment
Federation. American Public Health Association, Washington,
genotoxic in the “rec assay.” The genotoxicity of tribu- DC.
tyltin and triphenyltin was demonstrated in zebrafish APHA, 1998c. Standard methods for the examination of water and
circulating erythrocytes by the erythrocytic nuclear wastewater. Washington, DC.
abnormalities assay (Micael et al., 2007). Triphenyltin Appel, K.E., 2004. Drug Metab. Rev. 36, 763–786.
compounds induced micronuclei formation and sister Arakawa, Y., Wada, O., Yu, T.H., 1981. Toxicol. Appl. Pharmacol. 60,
1–7.
chromatid exchange in Chinese hamster ovary cells Arakawa, Y., Wada, O., Manabe, M., 1983. Anal. Chem. 55, 1901–1904.
(Chao et al., 1999). Triphenyltins were positive in the Ashby, J.R., Craig, P.J., 1988a. Sci. Total Environ. 73, 127–133.
Ames test and the mouse lymphoma TK+/mutation Ashby, J.R., Craig, P.J., 1988b. Sci. Total Environ. 73, 127–133.
56 Tin 1277

Ashby, J.R., Craig, P.J., 1991. Sci. Total Environ. 100, 337–346. Braman, R.S., Tompkins, M.A., 1979. Anal. Chem. 51, 12–19.
ATSDR (Agency for Toxic Substances and Disease Registry), 2005. Bridges, J.W., Davies, D.S., Williams, R.T., 1967. Biochem. J. 105,
Toxicological profile for Tin, Agency for Toxic Substances and 1261–1266.
Disease Registry. Atlanta, GA. Brown, R.A., Nazario, C.M., de Tirado, R.S., et al., 1977. Environ. Res.
Aung, N., Yoshinaga, J., Takahashi, J., 2006. Food Addit. Contam. 23, 13, 56–61.
883–894. Browning, E., 1969. Toxicity of industrial metals, second ed. London
Azenha, M., Vasconcelos, M.T., 2002. Anal. Chim. Acta. 458, 231–239. Butterworths, pp. 323–330.
Baaijens, P.A., 1986. Proceedings of an ORTEPA workshop, Bryan, G.W., Gibbs, P.E., Huggett, R.J., et al., 1989. Mar. Pollut. Bull.
­Berlin. ORTEP Association, Vlissingen-Oost, The Netherlands, 20, 458–462.
pp. 191–208. Bulten EJ, M.H., 1991. Metals and their compounds in the environ-
Baken, K.A., Arkusz, J., Pennings, J.L., et al., 2007. Toxicology 237, ment: Occurrence, analysis, and biological relevance. In: Merian,
35–48. E., VCH (Eds.) Weinheim, pp. 1243–1259.
Balaban, C.D., Ocallaghan, J.P., Billingsley, M.L., 1988. Neuroscience Burton, E.D., Phillips, I.R., Hawker, D.W., 2004. Environ. Sci. Tech-
26, 337–361. nol. 38, 6694–6700.
Baldwin, D.R., Marshall, W.J., 1999. Ann. Clin. Biochem. 36, 267–300. Cai, Y., Bayona, J.M., 1995. J. Chromatogr. Sci. 33, 89–97.
BAN (Basel Action Network), 2004. Mobile Toxic Waste: Recent Calloway, D.H., McMullen, J.J., 1966. Am. J. Clin. Nutr. 18, 1–6.
Findings on the Toxicity of End-of-Life Cell Phones. A Report by Carfi, M., Croera, C., Ferrario, D., et al., 2008. Toxicology 249,
Basel Action Network (BAN). http://ban.org/library/mobileph 11–18.
onetoxicityrep.pdf (accessed 5.12.13). Carfi, M., Bowe, G., Pieters, R., Gribaldo, L., 2010. Toxicology 276,
Bancon-Montigny, C., Maxwell, P., Yang, L., et al., 2002. Anal. Chem. 33–40.
74, 5606–5613. Carlin Jr. J.F., 2004. U.S. Geological Survey, Mineral Commodity
Bancon-Montigny, C., Seidel, J.L., Brissaud, F., Elbaz-Poulichet, F., Summaries.
2008. J. Environ. Monitor. 10, 638–647. Carvalho, P.N., Pinto, L.F., Basto, M., Vasconcelos, A., 2007. Micro-
Banu, N., Tsuchiya, T., Sawada, R., 2006. Animal Cell Technology: Ba- chem. J. 87, 147–153.
sic and Applied Aspects: Proceedings of the 19th Annual Meeting Centineo, G., Gonzalez, E.B., Sanz-Medel, A., 2004. J. Chromat. A
of the Japanese Association for Animal Cell Technology (JAACT). 1034, 191–197.
Kyoto, Japan, pp. 181–187. September 25–28 2006 Springer. Chalupa, J., Hanlir, K., Padelkova, Z., et al., 2008. Appl. Organomet.
Barker Jr., W.H., Runte, V., 1972. Am. J. Epidemiol. 96, 219–226. Chem. 22, 308–313.
Barnes, J.M., Magee, P.N., 1958. J. Pathol. Bacteriol. 75, 267–279. Chang, L.W., 1986a. Fund. Appl. Toxicol. 6, 217–232.
Barnes, J.M., Stoner, H.B., 1959. Pharmacol. Rev. 11, 211–231. Chang, L.W., 1986b. Neurotoxicol 7, 355.
Barrera, D., Maldonado, P.D., Medina-Camposa, O.N., et al., 2003. Chao, J.S., Wei, L.Y., Huang, M.C., et al., 1999. Mutat. Res. 444,
Life Sci. 73, 3027–3041. 167–174.
Barroso, C.M., Rato, M., Veríssimo, A., et al., 2011. J. Environ. Monit. Chen, B., Zhou, Q., Liu, J., et al., 2007. Chemosphere 68, 414–419.
13, 3018–3025. Chen, B., Thanh, W., Yin, Y., et al., 2012. Anal. Methods 4, 2109–2114.
Baselt, R.C., Cravey, R.H., 1989. Deposition of toxic drugs and chemi- Chen, B.W., Jiang, G.B., Yang, R.Q., Liu, J.Y., 2006. Appl. Organomet.
cals in man, third ed. Year Book Medical Publ., Inc., Chicago IL, Chem. 20, 161–167.
USA. Chen, J., Huang, C., Zheng, L., et al., 2011. Neurotoxicol. Teratol. 33,
Baul, T.S.B., Mizar, A., Chandra, A.K., et al., 2008. J. Inorg. Biochem. 721–726.
102, 1719–1730. Chen, J.Q., Mclaughlin, J.K., Zhang, J.Y., et al., 1992. J. Occup. Envi-
Beker van Woudenberg, A., Wolterbeek, A., Te Brake, L., et al., 2013. ron. Med. 34, 311–316.
Reprod. Toxicol. 41, 35–44. Chen, Q., Zhang, Z., Zhang, R., et al., 2011. Int. J. Hyg. Envir. Heal.
Benoy, C.J., Hooper, P.A., Schneider, R., 1971. Food Cosmet. Toxicol. 214, 145–150.
9, 645–656. Chen, W., Hnizdo, E., Chen, J.Q., et al., 2005. Am. J. Ind. Med. 48, 1–9.
Berg, C.P., Rothbart, A., Lauber, K., et al., 2003. Oncogene 22, 775–780. Chen, W.H., Yang, J., Chen, J.Q., Bruch, J., 2006. Am. J. Ind. Med. 49,
Berge, J.A., Brevik, E.M., Bjorge, A., et al., 2004. J. Environ. Monitor. 67–76.
6, 108–112. Chiba, M., Kikuchi, M., Tohyama, C., Nishikawa, M., 1990. Biol.
Bergmann, K., Neidhart, B., 2001. J. Sep. Sci. 24, 221–225. Trace Elem. Res. 25, 137–147.
Bi, X., Simoneit, B.R., Wang, Z., et al., 2010. Atmos. Environ. 44, Chmielnicka, J., Szymanska, J.A., Sniec, J., 1981. Arch. Toxicol. 47,
4440–4445. 263–268.
Bissonnette, S.L., Haas, A., Mann, K.K., et al., 2010. Toxicol. Sci. 118, Chmielnicka, J., Zareba, G., Grabowska, U., 1992. Ecotox. Environ.
108–118. Safe 24, 266–274.
Bjorn, A., Horsing, M., Ejlertsson, J., et al., 2011. Waste Manag. Res. Chmielnicka, J., Zareba, G., Polkowskakulesza, E., et al., 1993. Biol.
29, 1327–1336. Trace Elem. Res. 36, 73–87.
Blunden, S., Wallace, T., 2003. Food Chem. Toxicol. 41, 1651–1662. Choi, M., Moon, H.B., Choi, H.G., 2012. Arch. Environ. Contam.
Boa Morte, E.S., Korn, M.G., Saraiva, M., et al., 2009. Talanta 79, Toxicol. 62, 333–340.
1100–1103. Chow, S.C., Orrenius, S., 1994. Toxicol. Appl. Pharmacol. 127, 19–26.
Bocio, A., Nadal, M., Domingo, J.L., 2005. Biol. Trace Elem. Res. 104, Cima, F., Ballarin, L., Bressa, G., Burighel, P., 1998. Ecotoxicol. Envi-
193–201. ron. Saf. 40, 160–165.
Boogaard, P.J., Boisset, M., Blunden, S., et al., 2003. Food Chem. Toxi- Cima, F., Dominici, D., Mammi, S., Ballarin, L., 2002. Appl.
col. 41, 1663–1670. Organomet. Chem. 16, 182–186.
Booth, M.D., Fleet, B., 1970. Anal. Chem. 42, 825. Cobbing, M., 2008. Full Report, Greenpeace International. Report
Borghi, V., Porte, C., 2002. Environ. Sci. Technol. 36, 4224–4228. Greenpeace International, The Netherlands. http://www.green
Boutakhrit, K., Crisci, M., Bolle, F., Van Loco, J., 2011. Food Addit. peace.org.
Contam. A 28, 173–179. Coelho, M.R., Bebianno, M.J., Langston, W.J., 2002. Mar. Environ.
Boyer, I.J., 1989. Toxicology 55, 253–298. Res. 54, 179–192.
1278 Elena A. Ostrakhovitch

Colliar, L., Sturm, A., Leaver, M.J., 2011. Comp. Biochem. Physiol. Donard, O.F.X., Rapsomanikis, S., Weber, J.H., 1986. Anal. Chem. 58,
C Toxicol. Pharmacol. 153, 168–173. 772–777.
Colosio, C., Tomasini, M., Cairoli, S., et al., 1991. Br. J. Ind. Med. 48, Dopp, E., von Recklinghausen, U., Hippler, J., et al., 2011. J. Toxicol.
136–139. 2011, 503576.
Conine, D.L., Yum, M., Martz, R.C., et al., 1976. Toxicol. Appl. Phar- Dorn, E., Werner, H.J., 1989. Unpublished report CR077/88 (A41974)
macol. 35, 21–28. of Analytisches Laboratorium Hoechst AG. Submitted to WHO
Cook, L.L., Stine, K.E., Reiter, L.W., 1984. Toxicol. Appl. Pharmacol. by Hoechst AG, Frankfurt-am-Main.
76, 344–348. Dos Santos, R.L., Podratz, P.L., Sena, G.C., et al., 2012. J. Toxicol. Env.
Cooke, G.M., Forsyth, D.S., Bondy, G.S., et al., 2008. J. Toxicol. Env. Heal. A. 75, 948–959.
Heal. A 71, 384–395. Duester, L., Diaz-Bone, R.A., Kosters, J., et al., 2005. J. Environ. Moni-
Corbin, H.B., 1973. Anal. Chem. 45, 534–537. tor. 7, 1186–1193.
Corsini, E., Viviani, B., Marinovich, M., Galli, C.L., 1998. Toxicol. Dwivedi, R.S., Kaur, G., Srivestava, R.C., et al., 1983. Chemosphere
Vitro 12, 551–556. 12, 333–340.
Corvino, V., Marchese, E., Zarkovic, N., et al., 2011. Neurochem. Res. Eckert, H.G., Kellner, H.M., Beurkle, W.L., 1989. Unpublished Work.
36, 1490–1500. EFSA (European Food Safety Authority), 2004a. European Food
Costa, L.G., Sulaiman, R., 1986. Toxicol. Appl. Pharmacol. 86, Safety Authority (EFSa) 102, 1–114.
189–196. EFSA, 2004b. Opinion of the Scientic Panel on Contaminants in the
COT (Committee on Toxicity of Chemicals in Food), 2008. COT Food Chain on a request from the Commission to assess the health
statement on the 2006 UK Total Diet Study of metals and other risks to consumers associated with exposure to organotins in food-
­elements. Annual Report. stuffs.
Craig, P.J., Rapsomanikis, S., 1985. Environ. Sci. Technol. 19, 726–730. El Demerdash, F.M., Yousef, M.I., Zoheir, M.A., 2005. Food Chem.
CRC Handbook of Chemistry and Physics, CRC Press, 2011. Toxicol. 43, 1743–1752.
Cremer, J.E., 1957. Biochem. J. 67, 87–96. El Makawy, A.I., Girgis, S.M., Khalil, W.K., 2008. Mutat. Res. Gen.
Cristofol, R.M., Gasso, S., Vilchez, D., et al., 2004. Toxicol. Lett. 152, Tox. Environ. Mutagen 657, 105–110.
35–46. Elekes, C.C., Dumitriu, I., Busuioc, G., et al., 2010. Environ. Sci.
Crofton, K.M., Dean, K.F., Boncek, V.M., et al., 1989. Toxicol. Appl. ­Pollut. R 17, 1230–1236.
Pharmacol. 97, 113–123. Ellingsen, J.E., 1986. Scand. J. Dent. Res. 94, 299–305.
Cui, Z., Zhang, K., Zhou, Q., et al., 2011. Talanta 85, 1028–1033. Elsabbagh, H.S., Moussa, S.Z., El Tawil, O.S., 2002. Pharmacol. Res.
Cui, Z., Zhang, K., Zhou, Q., et al., 2012. Sci. China Chem. 55, 45, 201–206.
323–328. Ema, M., Miyawaki, E., 2002. Reprod. Toxicol. 16, 309–317.
Cummings, K.J., Donat, W.E., Ettensohn, D.B., et al., 2010. Am. J. Ema, M., Itami, T., Kawasaki, H., 1991a. Neurotoxicol. Teratol. 13,
Respir. Crit. Care Med. 181, 458–464. 489–493.
Cummings, K.J., Nakano, M., Omae, K., et al., 2012. Chest 141, Ema, M., Itami, T., Kawasaki, H., 1991b. Toxicol. Lett. 58, 347–356.
1512–1521. Ema, M., Harazono, A., Hirose, A., et al., 2003. Toxicol. Lett. 143,
Dabek-Zlotorzynska, E., Lai, E.P.C., Timerbaev, A.R., 1998. Anal. 233–238.
Chim. Acta 359, 1–26. Engberg, A., 1973. Analyst 98, 137–145.
Dantas, F.J.S., Moraes, M.O., Carvalho, E.F., et al., 1996. Toxicology Errecalde, O., Gommy, C., Maury, G., et al., 1995. Appl. Organomet.
34, 959–962. Chem. 9, 525–529.
Dantas, F.J.S., Moraes, M.O., de Mattos, J.C.P., et al., 1999. Toxicol. Escalante, B., Sacerdoti, D., Davidian, M.M., et al., 1991. Hyperten-
Lett. 110, 129–136. sion 17, 776–779.
Dantas, F.J.S., de Mattos, J.C.P., Moraes, M.O., et al., 2002. Cell. Mol. European Commission, 2004. European Commission Regula-
Biol. 48, 789–791. tion 242/2004. http://eur-lex.europa.eu/LexUriServ/LexU
de Groot, A.P., Feron, V.J., Til, H.P., 1973. Food Cosmet. Toxicol. 11, riServ.do?uri=OJ: L:2004:042:0003:0004:EN: PDF (accessed
19–30. 29.11.13).
de Mattos, J.C.P., Lage, C., Dantas, F.J.S., et al., 2005. Mol. Cell. Bio- European Commission, 2006a. Commission Regulation (EC)
chem. 280, 173–179. No 1881/2006. Official Journal of European Union l 364.
De Santiago, A., Aguilar-Santelises, M., 1999. Hum. Exp. Toxicol. 18, http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:
619–624. L:2006:364:0005:0024:EN: PDF (accessed 29.11.13).
de Smaele, T., Moens, L., Dams, R., et al., 1998. J. Chromat. A 793, European Commission, 2006b. Commission of the European Com-
99–106. munities COM No 398. http://eur-lex.europa.eu/LexUriServ/
Delucchi, F., Tombesi, N.B., Freije, R.H., et al., 2007. Environ. Monit. LexUriServ.do?uri=COM:2007:0398:FIN: EN:PDF (accessed
Assess. 132, 445–451. 5.12.13).
der Meulen, H.C., Feron, V.J., Til, H.P., 1974. Pathol. Eur. 9, 185–192. European Commission, 2007. Commission Regulation (EC)No
Detcheva, A., Grobecker, K., 2006. Spec. Acta B. 61, 454–459. 33/2007. http://eur-lex.europa.eu/LexUriServ/LexUriServ.
Devos, C., Vliegen, M., Willaert, B., et al., 2005. J. Chromat. A 1079, do?uri=OJ: L:2007:010:0007:0008:EN: PDF (accessed 29.11.13).
408–414. European Commission, 2009. EU Commission Regulation (EC) No.
Dietz, C., Sanz, J., Sanz, E., et al., 2007. J. Chromat. A 1153, 114–129. 124/2009. http://eur-lex.europa.eu/LexUriServ/LexUriServ.do
Dizikes, L.J., Ridley, W.P., Wood, J.M., 1978. J. Am. Chem. Soc. 100, ?uri=OJ: L:2009:040:0007:0011:en:PDF (accessed 29.11.13).
1010–1012. European Commission, 2010. EU Commission Regulation No
Dobson, S., Howe, P.D., Floyd, P., 2006. Concise International Chemi- 276/2010. http://eur-lex.europa.eu/LexUriServ/LexUriServ.do
cal Assessment Document 73. ?uri=OJ: L:2010:086:0007:0012:en:PDF (accessed 29.11.13).
Doctor, S.V., Fox, D.A., 1982. J. Toxicol. Environ. Health 10, 43–52. European Commission, 2011. European Commission r­egulation
Domingo, J.L., Perello, G., Gine Bordonaba, J., 2012. Biol. Trace Elem. No 836/2011 amending European Commission regulation No
Res. 146, 420–425. 333/2007. http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?
Donard, O.F.X., Weber, J.H., 1988. Nature 332, 339–341. uri=OJ: L:2011:215:0009:0016:EN: PDF (accessed 29.11.13).
56 Tin 1279

European Council of Vinyl Manufacturers (n.d.) Science—PVC, Geloso, M.C., Corvino, V., Michetti, F., 2011. Neurochem. Int. 58,
Stabilisers and Plasticisers, paper produced by the European 729–738.
Council of Vinyl Manufacturers (ECVM) in conjunction with the Gennari, A., Potters, M., Seinen, W., et al., 1997. Toxicol. Appl. Phar-
European Stabilisers Producers Association (ESPA), E ­ uropean macol. 147, 259–266.
Council of Vinyl Manufacturers. http://www.pvc.org/en Gennari, A., Viviani, B., Galli, C.L., et al., 2000. Toxicol. Appl. Phar-
(accessed 10.12.13). macol. 169, 185–190.
Evans, O., Kauffman, P., Vonderheide, et al., 2009. Microchem. J. 92, Gipperth, L., 2009. J. Environ. Manage. 90, S86–S95.
155–164. Glawe, C., Emmrich, J., Sparmann, G., et al., 2005. Lab. Invest. 85,
Evans, W.H., Cardarelli, N.F., Smith, D.J., 1979. J. Toxicol. Environ. 193–204.
Health 5, 871–877. Gogvadze, V., Stridh, H., Orrenius, S., et al., 2002. Biochem. Biophys.
Falke, A.M., Weber, J.H., 1994a. Appl. Organomet. Chem. 8, 351–359. Res. Commun. 292, 904–908.
Falke, A.M., Weber, J.H., 1994b. Abstr. Papers Amer. Chem. Soc. 207. Goh, C.L., 1985. Contact Dermatitis 12, 161–163.
75–GEOC. Golub, M., Doherty, J., 2004. J. Toxicol. Environ. Health B Crit. Rev.
Fang, C., Wang, X., Wang, W., et al., 2011. Asian J. Ecotoxicol. 6, 54–59. 7, 281–295.
Fent, K., Hunn, J., Sturm, M., 1991a. Naturwissenschaften 78, Gonzalez-Toledo, E., Compano, R., et al., 2003. Trac-Trends Anal.
219–221. Chem. 22, 26–33.
Fent, K., Lovas, R., Hunn, J., 1991b. Naturwissenschaften 78, 125–127. Gosz, E., Horbowy, J., Ruczynska, W., 2011. Mar. Pollut. Bull. 62,
Fent, K., 1996. Crit. Rev. Toxicol. 26, 3–117. 2563–2567.
Fernandez, M.A., Wagener, A.D.R., Limaverde, A.M., et al., 2005. Grace, C.T., Ng, S.K., Cheong, L.L., 1991. Contact Dermatitis 25,
Mar. Environ. Res. 59, 435–452. 250–251.
Fine Olivarius, F., Balslev, E., Menne, T., 1993. Contact Dermatitis Granmo, A., Ekelund, R., Sneli, J.A., et al., 2002. Mar. Pollut. Bull. 44,
29, 110–111. 1142–1148.
Fish, R.H., 1984. Neurotoxicol 5, 159–161. Grassino, A.N., Grabaric, Z., Pezzani, A., et al., 2010. J. Sci. Food.
Florea, A.M., Dopp, E., Obe, G., et al., 2004. Organic Metal and Agric. 90, 2419–2426.
Metalloid Species in the Environment: Analysis, Distribution, Greger, J.L., Smith, S.A., Johnson, M.A., et al., 1982. Biol. Trace Elem.
Processes and Toxicological Evaluation, 205–219. Res. 4, 269–278.
Florence, T.M., Farrar, Y.J., 1974. J. Electroanal. Chem. 51, 191–200. Grinwis, G., Wester, P., Vethaak, A., 2009. Environ. Pollut. 157,
FSA (Food Standards Agency), 2002. Report No 2002-08-22. 2587–2593.
FSA., 2003. Report of the Expert Group on Vitamins and Minerals. Grondin, M., Marion, M., Denizeau, F., et al., 2007. Toxicol. Appl.
Food Standards Agency, UK. Pharmacol. 222, 57–68.
FSA., 2009a. Survey on measurement of the concentrations of metals Grote, K., Stahlschmidt, B., Talsness, C.E., et al., 2004. Toxicology 202,
and other elements from the 2006 UK total diet study. Food Sur- 145–158.
veillance Information Sheet No. 01/09. London (UK): FSA, Com- Grote, K., Andrade, A.J.M., Grande, S.W., et al., 2006. Toxicology 222,
mittee on Toxicity of Chemicals in Food, Consumer Products and 17–24.
Environment (COT). Grote, K., Hobler, C., Andrade, A.J., et al., 2007. Toxicology 238,
FSA, 2009b. The Contaminants in Food (England) Regulations 2009 177–185.
(Statutory Instrument 2009 No. 1223). Grote, K., Hobler, C., Andrade, A.J., et al., 2009. Toxicology 260,
Fortemps, E., Amand, G., Bomboir, A., et al., 1978. Int. Arch. Occup. 53–59.
Environ. Health 41, 1–6. Grun, F., Blumberg, B., 2006. Endocrinology 147, S50–S55.
Frena, M., Campestrini, I., de Braga, O.C., et al., 2011. Electrochim. Grun, F., Watanabe, H., Zamanian, Z., et al., 2006. Mol. Endocrinol.
Acta. 56, 4678–4684. 20, 2141–2155.
Fritsch, P., de Saint, B.G., Derache, R., 1977. Toxicology 8, 165–175. Grundler, W., Dirscherl, P., Beisker, W., et al., 2001. Cytometry 44, 45–56.
Frouin, H., Lebeuf, M., Saint-Louis, R., et al., 2008. Aquat. Toxicol. Gruner, J.E., 1958. Rev. Neurol. 98, 109–116.
90, 243–251. Guard, H.E., Cobet, A.B., Coleman, W.M., 1981. Science 213, 770–771.
Funahashi, N., Iwasaki, I., Ide, G., 1980. Acta Pathol. Jpn. 30, 955–966. Guedes, A.P., Cardoso, V.N., De Mattos, J.C., et al., 2006. Nucl. Med.
Furchner, J.E., Drake, G.A., 1976. Health Phys. 31, 219–224. Biol. 33, 915–921.
Furuhashi, K., Ogawa, M., Suzuki, Y., et al., 2008. Chem. Res. Toxicol. Guerin, T., Sirot, V., Volatier, J., et al., 2007. Sci. Total Environ. 388,
21, 467–471. 66–77.
Gadd, G.M., 2000. Sci. Total Environ. 258, 119–127. Guruge, K.S., Tanabe, S., Iwata, H., et al., 1996. Arch. Environ. Contam.
Gaddoni, G., Baldassari, L., Francesconi, E., et al., 1993. Contact Toxicol. 31, 210–217.
­Dermatitis 28, 127–128. Hadjispyrou, S.A., Anagnostopoulos, A., Nicholson, K., et al., 1998.
Gallego-Gallegos, M., Liva, M., Olivas, R.M., et al., 2006. J. Chromat. Environ. Geochem. Hlth. 20, 19–27.
A 1114, 82–88. Hagger, J.A., Depledge, M.H., Galloway, T.S., 2005. Mar. Pollut. Bull.
Gallina, A., Magno, F., Tallandini, L., et al., 2000. Rapid. Commun. 51, 811–816.
Mass Spec. 14, 373–378. Hallas, L.E., Means, J.C., Cooney, J.J., 1982. Science 215, 1505–1507.
Garcia, F., Ortega, A., Domingo, J.L., et al., 2001. J. Environ. Sci. Hamasaki, T., Sato, T., Nagase, H., et al., 1992. Mut. Res. 280, 195–203.
Health A 36, 1767–1786. Hamilton, E.I., Minski, M.J., 1972. Sci. Total Environ. 1, 375–394.
Garg, A., Meena, R.M., Bhosle, N.B., 2010. Environ. Monit. Assess. Harino, H., Fukushima, M., Kawai, S., 2000. Arch. Environ. Contam.
165, 643–651. Toxicol. 39, 13–19.
Gasparova, Z., Janega, P., Stara, V., et al., 2012. Neuro. Endocrinol. Harino, H., O’Hara, S.C.M., Burt, G.R., et al., 2005. Chemosphere 58,
Lett. 33, 689–696. 877–881.
Gasso, S., Sanfeliu, C., Sunol, C., et al., 2000. Toxicol. Appl. Pharma- Harino, H., Ohji, M., Brownell, R.L., et al., 2008a. Arch. Environ.
col. 162, 189–196. Contam. Toxicol. 55, 137–142.
Gaunt, I.F., Colley, J., Grasso, P., et al., 1968. Food Cosmet. Toxicol. Harino, H., Ohji, M., Wattayakorn, G., et al., 2008b. Arch. Environ.
6, 599–608. Contam. Toxicol. 54, 145–153.
1280 Elena A. Ostrakhovitch

Harino, H., Arifin, Z., Rumengan, F.M.J., et al., 2012. Arch. Environ. ISO, 2003b. Report No 17294-2:2003. Water quality: Application of
Contam. Toxicol. 63, 13–21. inductively coupled plasma mass spectrometry (ICP-MS). Part2:
Harry, G., Funk, J.A., d’Hellencourt, C.L., et al., 2008. Brain Res. 1194, Determination of 62 elements. International Organization for
8–20. Standardization, Geneva.
Health Council of the Netherlands, 2005. Dutch Expert Committee on ISO, 2004. Report No 17240:2004. Fruit and vegetable products: De-
Occupational Standards. Tin and inorganic tin compounds; Health- termination of tin content. Method using flame atomic absorp-
based recommended occupational exposure limit. Health Council tion spectrometry. International Organization for Standardiza-
of the Netherlands, 2005; publication no. 2005/06OSH, The Hague. tion, Geneva.
Henderson, W., Taylor, M.J., 1996. Polyhedron 15, 1957–1964. I.T.R.L, Ltd, 2011. Tin industry review 2011. news release, Frogmore,
Henderson, W., McIndoe, J.S., 2005. Mass Spectrometry of inorganic, United Kingdom.
coordination and organometallic compounds: Tools-Techniques- Iwamoto, I., 1969. J. Tokyo. Med. Coll. 18, 1351–1376.
Tips. John Wiley &Sons, LTD, Chichester. Iwamura, C., Aihara, M., Nakazawa, M., et al., 2006. Int. Arch.
Heroult, J., Zuliani, T., Bueno, M., et al., 2008. Talanta 75, 486–493. ­Allergy Immunol. 141, 337–345.
Hiles, R.A., 1974. Toxicol. Appl. Pharmacol. 27, 366–379. JECFA, 1978. WHO Technical Report Series, No. 631 Report No 22.
Hirano, Y., Yamamura, K., Oguma, K., et al., 2001. Anal. Sci. (Suppl. 17), (Twenty-second report of the Joint FAO/WHO Expert Commit-
il295–il298. tee on Food Additives).
Hoch, M., 2001. Appl. Geochem. 16, 719–743. JECFA, 1982. Joint FAO/WHO Expert Committee on Food Additives
Hoch, M., Schwesig, D., 2004. Appl. Geochem. 19, 323–334. (WHO Food Additives Series No. 17). Cambridge University
Hoch, M., Alonso-Azcarate, J., Lischick, M., 2002. Environ. Toxicol. Press, Cambridge, pp. 297–319.
Chem. 21, 1390–1397. JECFA, 1989. Thirty-third Report of the Joint FAO/WHO Expert
Hoch, M., Alonso-Azcarate, J., Lischick, M., 2003. Environ. Pollut. Committee on Food Additives. WHO Technical Report Series No.
123, 217–227. 776. WHO, Geneva.
Hodgson, J.T., Jones, R.D., 1990. Brit. J. Ind. Med. 47, 665–676. JECFA, 2001. Codex Alimentarius Comission-Foods and Agriculture
Horacek, V., Demcik, K., 1970. Prac.Lék. 22, 61–66. Organization of the UN/World Health Organization (Thirty-
Horiguchi, T., 2009. Ecotoxicol. Antifouling Biocides, pp. 111–124. third report of the Joint FAO/WHO Expert Committee on Food
Horiguchi, T., Cho, H., Shiraishi, H., et al., 2001a. RIKEN Review: Additives). Report No 33.
Focused on New Trends in Bio-Trace Elements Research 35, 9–11. JECFA, 2006. Sixty-fourth report of the Joint FAO/WHO Expert
Horiguchi, T., Cho, H.S., Shiraishi, H., et al., 2001b. Otsuchi Mar. Sci. Committee on Food Additives. WHO Technical Report Series,
26, 61–63. No. 930, Report No 64.
Horiguchi, T., Kojima, M., Hamada, F., et al., 2006. Environ. Health JECFA, 2011. Food standards programme codex committee on contami-
Perspect. 114, 13–19. nants in foods. Joint Food and Agriculture Organization (FAO) of
Horiguchi, T., Lee, J.H., Park, J.C., et al., 2012. Mar. Environ. Res. 76, United Nations/World Health Organization (WHO). Fifth session.
56–62. Jenkins, S.M., Barone, S., 2004. Toxicol. Lett. 147, 63–72.
Hu, H., 1998. Harrison’s principles of Internal medicine. In: Fauci, Jenkins, S.M., Ehman, K., Barone, S., 2004. Dev. Brain Res. 151, 1–12.
A.S., Braunwald, E., Isselbacher, K.J., Wilson, J.D., Martin, J.B. Jensen, K.G., Onfelt, A., Wallin, M., et al., 1991. Mutagenesis 6,
(Eds.). McGraw-Hill, New York, pp. 2564–2569. 409–416.
Huang, J.H., Matzner, E., 2004a. Eur. J. Soil. Sci. 55, 693–698. Jiang, G., Zhou, Q., He, B., 2000. Bull. Environ. Contam. Toxicol. 65,
Huang, J.H., Matzner, E., 2004b. J. Plant Nutr. Soil Sci. 167, 33–38. 277–284.
Hutton, E.A., Ogorevc, B., Hocevar, S.B., Smyth, M.R., 2006. Anal. Jimenez, M.S., Gomez, M.T., Castillo, J.R., 2007. Talanta 72,
Chim. Acta. 557, 57–63. 1141–1148.
Igarashi, I., 1959. J. Tokyo Med. Coll. 17, 1603–1632. Jing, H.W., Amirav, A., 1998. J. Chromatogr. A 805, 177–215.
Impellitteri, C.A., Evans, O., Ravel, B., 2007. J. Environ. Monitor. 9, Johnson, M.A., Greger, J.L., 1982. Am. J. Clin. Nutr. 35, 655–660.
358–365. Johnson, M.A., Greger, J.L., 1985. J. Nutr. 115, 615–624.
Inadera, H., Shimomura, A., 2005. Toxicol. Lett. 159, 226–234. Jones-Lepp, T.L., Varner, K.E., Heggem, D., 2004. Arch. Environ.
Inagaki, K., Takatsu, A., Watanabe, T., et al., 2007. Anal. Bioanal. Contam. Toxicol. 46, 90–95.
Chem. 387, 2325–2334. Juliao, L.M., Melo, D.R., Sousa, W.O., et al., 2007. Radiat. Prot. D
­ osim.
IGES (Institute for Global Environmental Strategies). (2009). 125, 513–515.
Tsydenova O., Bengtsson M. Environmental and Human health Kanayama, T., Kobayashi, N., Mamiya, S., et al., 2005. Mol. Pharma-
risks ­associated with the End-of life Treatment of electrical and col. 67, 766–774.
electronic equipment. http://pub.iges.or.jp/modules/envirolib/ Kannan, K., Falandysz, J., 1997. Mar. Pollut. Bull. 34, 203–207.
view.php?docid=2519 (accessed 29. 11. 13). Kannan, K., Lee, R.F., 1996. Environ. Toxicol. Chem. 15, 1492–1499.
IPCS (Programme on Chemical Safety), 1999. International Pro- Kannan, K., Corsolini, S., Focardi, S., et al., 1996. Arch. Environ. Con-
gramme on Chemical Safety, World Health Organization (WHO). tam. Toxicol. 31, 19–23.
Document 13. Kannan, K., Senthilkumar, K., Giesy, J.P., 1999. Environ. Sci. Technol.
Isensee, J., Watermann, B., Berger, H.D., 1994. Germ. J. Hydrogr. 46, 33, 1776–1779.
355–365. Kannan, K., Kajiwara, N., Watanabe, M., et al., 2004. Environ. Toxi-
Ishaaya, I., Engel, J.L., Casida, J.E., 1976. Pestic. Biochem. Physiol. col. Chem. 23, 49–56.
6, 270–279. Kannan, K., Takahashi, S., Fujiwara, N., et al., 2010. Arch. Environ.
Ishihara, N., Matsushiro, T., 1986. Arch. Environ. Health 41, 324–330. Contam. Toxicol. 58, 901–907.
Ishizaka, T., Suzuki, T., Saito, Y., 1989. J. Agric. Food. Chem. 37, Kapsimali, D., Rosenberg, E., Zachariadis, G., 2012. Anal. Lett. 45,
1096–1101. 642–650.
ISO (International Organization for Standardization), 2003a. Report Katika, M.R., Hendriksen, P.J., van Loveren, H., et al., 2011. Toxicol.
No 9941:2003. Milk and canned evaporated milk. Determination Appl. Pharmacol. 254, 311–322.
of tin content: Spectrometric method. International Organization Katika, M.R., Hendriksen, P.J., de Ruijter, N.C., et al., 2012. Toxicol.
for Standardization, Geneva. Lett. 212, 126–136.
56 Tin 1281

Kaur, V., Malik, A.K., Verma, N., 2006. J. Sep. Sci. 29, 333–345. Lin, T.J., Hung, D.Z., Kao, C.H., et al., 1998. Hum. Exp. Toxicol. 17,
Kawaguchi, M., Inoue, K., Yoshimura, M., et al., 2004. J. Chromat. B: 403–405.
Biomed. Sci. Appl. 805, 41–48. Liscio, C., Di Carro, M., Magi, E., 2009. Comptes. Rendus Chimie 12,
Kehoe, R.A., Cholak, J., Story, R.V., 1940. J. Nutr. 20, 98. 831–840.
Kellner, H.M., Eckert, H.G., 1989. Unpublished Report 01-L42- Lisi, P., Caraffini, S., Assalve, D., 1987. Contact Dermatitis 17,
582-90 (A43434) of Pharma Forschung GB-L Radiochemisches 212–218.
Laboratorium Hoechst, Submitted to WHO by Hoechst AG., Liu, E., Du, X., Ge, R., et al., 2013. Food Chem. Toxicol. 60, 302–
­Frankfurt-am-Main. 308.
Kim, S.K., Kim, J.H., Han, J.H., et al., 2008. Int. J. Toxicol. 27, 175–182. Liu, L.L., Wang, J.T., Chung, K.N., et al., 2011. Mar. Pollut. Bull. 63,
Kimbrough, R.D., 1976. Environ. Health Perspect. 14, 51–56. 535–540.
Kimmel, E.C., Fish, R.H., Casida, J.E., 1977. J. Agric. Food Chem. 25, Llobet, J.M., Granero, S., Schuhmacher, M., et al., 1998. Trace Elem.
1–8. Electroly. 15, 44–49.
Kimura, K., Kobayashi, K., Naito, H., et al., 2005. Biosci. Biotechnol. Lopez de Alda, M.J., Barcelo, D., 2001. J. Chromat. A 938, 145–153.
Biochem. 69, 1104–1110. Lu, T.C., Whang, C.W., 1995. J. Chin. Chem. Soc. 42, 515–520.
Kirchner, S., Kieu, T., Chow, C., et al., 2010. Mol. Endocrinol. 24, Lu, Y.C., Kuo, S.Y., Jiann, B.P., et al., 2003. Environ. Toxicol. Pharma-
526–539. col. 14, 1–7.
Kirk-Othmer Encyclopedia of Chemical Technology, 2000. Tin com- Lyle, W.H., 1958. Br. J. Ind. Med. 15, 193–196.
pounds. Wiley, New York. MAFF (Ministry of Agriculture, Fisheries and Food), 1983. Food Ad-
Kizlink, J., 1996. Chemicke Listy 90, 147–154. ditives and Contaminants Committee Report. Report on the re-
Klevay, L.M., 2000. Clinical Nutrition of the Essential Trace Elements view of metals in canned foods. (FAC/REP/38).
and Minerals: The Guide for Health Professionals. In: Bogden, MAFF, 1998. Lead, Arsenic and other metals in Food. Food Surveil-
J.D., Klevay, L.M. (Eds.). Humana Press Inc., Totowa, N.J, pp. lance Paper No. 52.
251–271. Maier, W.E., Brown, H.W., Tilson, H.A., et al., 1995. J. Neuroimmu-
Klevay, L.M., Combs, G.F., 2004. For World Health Organization, nol. 59, 65–75.
­Geneva. http://www.who.int/water_sanitation_health/dwq/ Makita, Y., Omura, M., 2006. Basic Clin. Pharmacol. Toxicol. 99,
nutrientsindw.pdf (accessed 29.11.13). 128–132.
Kloke, A., Sauerbeck, D.R., Vetter, H., 1984. Changing metal cycles Manzo, L., Richelmi, P., Sabbioni, E., et al., 1981. Clin. Toxicol. 18,
and human health. In: Nriagu, J.O. (Ed.). Springer-Verlag, Berlin, 1343–1353.
pp. 113–114. Manzoori, J.L., Amjadi, M., Abolhasani, D., 2006. J. Hazard. Mater.
Kobayashi, H., Suzuki, T., Kasashima, Y., et al., 1996. Jpn. J. Pharma- 137, 1631–1635.
col. 72, 317–324. Marciniak, M., 1981. Acta Physiol. Pol. 32, 193–204.
Konno, N., Tsunoda, M., Nakano, K., et al., 2001. Arch. Toxicol. 75, Marti-Cid, R., Perello, G., Domingo, J.L., 2009. Biol. Trace Elem. Res.
549–554. 131, 245–254.
Krajnc, E.I., Wester, P.W., Loeber, J.G., et al., 1984. Toxicol. Appl. Matsui, H., Wada, O., Ushijima, Y., et al., 1983. Arch. Toxicol. 54,
Pharmacol. 75, 363–386. 227–233.
Krigman, M.R., Silverman, A.P., 1984. Neurotoxicology 5, 129–139. Matsui, H., Wada, O., Manabe, S., et al., 1984. Experientia 40,
Kuballa, J., Wilken, R.D., Jantzen, E., et al., 1995. Analyst 120, 377–378.
667–673. Matthias, C.L., Bushong, S.J., Hall, L.W., et al., 1988. Appl. Organomet.
Kumasaka, K., Miyazawa, M., Fujimaki, T., et al., 2002. J. Reprod. Chem. 2, 547–552.
Dev. 48, 591–597. McLaughlin, R.L.J., Brindle, I.D., 2002. J. Anal. At. Spectrom. 17,
Landmeyer, J.E., Tanner, T.L., Watt, B.E., 2004. Environ. Sci. Technol. 1540–1548.
38, 4106–4112. McLaughlin, R., Cheese, P., Ding, M., et al., 2006. Am. Lab. 38. 13-+.
Lane, R., Ghazi, S.O., Whalen, M.M., 2009. Arch. Environ. Contam. McLean, J.R., Birnboim, H.C., Pontefact, R., et al., 1983a. Chem. Biol.
Toxicol. 57, 816–825. Interact. 46, 189–200.
Langston, W.J., Burt, G.R., Zhou, M.J., 1987. Mar. Pollut. Bull. 18, Mclean, J.R., Blakey, D.H., Douglas, G.R., et al., 1983b. Mut. Res. 119,
634–639. 195–201.
Laughlin, R.B., Linden, O., 1985. Ambio 14, 88–94. McSheehy, S., Hamester, M., Lindemann, T., et al., 2012. Application
Laughlin, R.B., Guard, H.E., Coleman, W.M., 1986. Environ. Sci. note: 30153, Thermo Fisher Scientific, Bremen, Germany.
Technol. 20, 201–204. Meador, J.P., Sommers, F.C., Cooper, K.A., et al., 2011. Environ. Res.
Le Gac, M., Lespes, G., Potin-Gautier, M., 2003. J. Chromat. A 999, 111, 50–56.
123–134. Menne, T., Andersen, K.E., Kaaber, K., et al., 1987. Contact Derma-
Le Maire, A., Grimaldi, M., Roecklin, D., et al., 2009. EMBO Rep 10, titis 16, 9–10.
367–373. Mensink, B.P., HallersTjabbes, C.C., Kralt, J., et al., 1996. Mar. Envi-
Lee, C.C., Wang, T., Hsieh, C.Y., et al., 2005. Environ. Pollut. 137, ron. Res. 41, 315–325.
198–208. Merkord, J., Jonas, L., Weber, H., et al., 1997. Pancreas 15, 392–401.
Lehotzky, K., Szeberenyi, J.M., Gonda, Z., et al., 1982. Neurobehav. Merkord, J., Weber, H., Sparmann, G., et al., 1998. Gastroenterology
Toxicol. Teratol. 4, 247–250. 114, A482–A483.
Levine, S., Saltzman, A., 1996. Biol. Trace Elem. Res. 52, 303–308. Micael, J., Reis-Henriques, M., Carvalho, A., et al., 2007. Environ. Int.
Lewis, P.G., Emmett, E.A., 1987. Contact Dermatitis 17, 129–132. 33, 1035–1039.
Li, Q., Lu, G.H., Wu, H., et al., 1999. Food Chem. 64, 129–132. Middleton, M.C., Pratt, I., 1978. J. Invest. Dermatol. 71, 305–310.
Li, X., Ycaza, J., Blumberg, B., 2011. J. Steroid Biochem. 127, 9–15. Mignini, F., Nasuti, C., Artico, M., et al., 2012. Int. J. Immunopathol.
Lilley, T., Ruokolainen, L., Vesterinen, E., et al., 2012a. Ecotoxicol 21, Pharmacol. 25, 1107–1119.
1333–1344. Miki, S., Ikeda, K., Oba, Y., et al., 2011. Mar. Pollut. Bull. 62,
Lilley, T.M., Meierjohann, A., Ruokolainen, L., et al., 2012b. Environ. 2533–2536.
Toxicol. Chem. 31, 1781–1787. Miller, K., Maisey, J., Nicklin, S., 1986. Environ. Res. 39, 434–441.
1282 Elena A. Ostrakhovitch

Millour, S., Noel, L., Kadar, A., et al., 2011. J. Food Comp. Anal. 24, NTP, 1982. National Institutes of Health, National ToxicologyPro-
111–120. gram (TR-231; NIH Publication No. 82-1787) Bethesda, MD.
Misiti, F., Orsini, F., Clementi, A., et al., 2008. Neurochem. Int. 52, Nuyts, G.D., Van Vlem, E., Thys, J., et al., 1995. Lancet 346, 7–11.
1092–1099. Oberdorster, E., McClellan-Green, P., 2002. Mar. Environ. Res. 54,
Miura, Y., Kato, M., Ogino, K., et al., 1997. Endocrinology 138, 715–718.
2769–2775. O’Callaghan, J.P., Miller, D.B., 1988. J. Pharmacol. Exp. Ther. 246,
Miyauchi, M., Suda, K., Kuwayama, C., et al., 2007. Histol. Histo- 394–402.
pathol. 22, 1119–1127. O’Callaghan, J.P., Niedzwiecki, D.M., Means, J.C., 1989. Brain Res.
Mizuhashi, S., Ikegaya, Y., Matsuki, N., 2000. Neurosci. Res. 38, Bull. 22, 637–642.
35–42. Ogata, R., Omura, M., Shimasaki, Y., et al., 2001. J. Toxicol. Environ.
Mizukawa, H., Takahashi, S., Nakayama, K., et al., 2009. Report, Health A 63, 127–144.
153–161. Ogoshi, K., Kurumatani, N., Aoki, Y., et al., 1981. Toxicol. Appl. Phar-
Mol, S., 2011. Food Chem. Toxicol. 49, 348–351. macol. 58, 331–332.
Molin, L., Wahlberg, J.E., 1975. Berufs-Dermatosen 23, 138–142. Ohhira, S., Matsui, H., 1996. Toxicol. Lett. 85, 3–8.
Monperrus, M., Martin-Doimeadios, R.C.R., Scancar, J., et al., 2003. Ohhira, S., Matsui, H., 2003. J. Appl. Toxicol. 23, 31–35.
Anal. Chem. 75, 4095–4102. Ohhira, S., Matsui, H., Nitta, K., 1996. Vet. Hum. Toxicol. 38, 206–209.
Morcillo, Y., Cai, Y., Bayona, J.M., 1995. J. High Resol. Chromat. 18, Ohhira, S., Matsui, H., Watanabe, K., 1999a. Toxicology 137, 151–159.
767–770. Ohhira, S., Matsui, H., Watanabe, K., 1999b. Toxicology 137, 151–159.
Morita, M., Imai, H., Liu, Y., et al., 2008. Neuroscience 153, 1135– Ohhira, S., Watanabe, M., Matsui, H., 2003. Arch. Toxicol. 77,­
1145. 138–144.
Motelica-Heino, M., Le Coustumer, P., Thomassin, J.H., et al., 1998. Ohhira, S., Watanabe, M., Matsui, H., 2004. Toxicol Lett. 148. 141-+.
Talanta 46, 407–422. Ohhira, S., Enomoto, M., Matsui, H., 2006a. Toxicol. Appl. Pharma-
Moynier, F., Fujii, T., Telouk, P., 2009. Anal. Chim. Acta 632, 234–239. col. 210, 32–38.
Nagamatsu, K., Kido, Y., Urakubo, G., et al., 1978. Jap. J. Hyg. 33, Ohhira, S., Enomoto, M., Matsui, H., 2006b. Toxicology 228, 171–177.
486–496. Ohji, M., Arai, T., Miyazaki, N., 2007. Estuar. Coast Shelf S. 72,
Nakanishi, T., Kohroki, J., Suzuki, S., et al., 2002. J. Clin. Endocrinol. 721–731.
Metab. 87, 2830–2837. Ohno, S., Nakajima, Y., Ohno, S., 2005. Steroids 70, 645–651.
Nakatsu, Y., Kotake, Y., Ohta, S., 2006. Neurotoxicology 27, 587–593. Okoro, H.K., Fatoki, O.S., Adekola, F.A., et al., 2011. Rev. Environ.
Nakayama, A., Kurokawa, Y., Harino, H., et al., 2007. Aquat. Toxicol. Contam. Toxicol. 213, 27–54. 213.
83, 126–133. Olivier, P., Marzin, D., 1987. Mut. Res. 189, 263–269.
Nehring, P., 1972. Ind. Obst. Gemusseverwert. 57, 489–492. Omura, M., Ogata, R., Kubo, K., et al., 2001. Toxicol. Sci. 64, 224–232.
NEMI (National Environmental Methods Index). Standard Method: ORTEPA (Organotin Environmental Programme Association), 1994.
3113B. https://www.nemi.gov/methods/method_summary/ For the organotin Environmental Programme Association (HD
4698/ (accessed 29.11.13). Project No 380-211).
NEMI. Standard Method: 3125. https://www.nemi.gov/methods/ OSHA (Occupational Safety and Health Administration), 2013. U.S.
method_summary/4796/ (accessed 29.11.13). Department of Labor. Tin, organic compounds (as Sn). https://
Nesci, S., Ventrella, V., Trombetti, F., et al., 2011a. Toxicol. in Vitro 25, www.osha.gov/dts/chemicalsampling/data/CH_271900.html
117–124. (accessed 29.11.13).
Nesci, S., Ventrella, V., Trombetti, F., et al., 2011b. Biochimie 93, Oshiro, Y., Piper, C.E., Balwierz, P.S., Soelter, S.G., (1991). J Appl
1157–1164. Toxicol. 11, 167–177.
Nielsen, J.B., Strand, J., 2002. Environ. Res. 88, 129–133. Osman, A.M., van Loveren, H., 2012. Toxicol. Sci. 126, 84–100.
Nincevic Grassino, A., Grabaric, Z., Pezzani, A., et al., 2009. Food Osman, A.M., van Kol, S., Peijnenburg, A., et al., 2009.
Aaddit. Contam. A 26, 1488–1494. J. ­Immunotoxicol. 6, 174–183.
NIOSH (National Institute for Occupational Safety and Health), Ozcan, M., Akman, S., 2000. Spec. Acta B 55, 509–515.
1994. NIOSH manual of analytical methods, Method No.7300 Pavlikova, N., Kortner, T.M., Arukwe, A., 2010. Aquat. Toxicol. 99,
Department of Health andHuman Services, National Institute 176–185.
for Occupational Safety andHealth (DHHS (NIOSH) Publication Pekelharing, H.L., Lemmens, A.G., Beynen, A.C., 1994. Br. J. Nutr.
94-113. Cincinnati, OH, US. 71, 103–109.
NIOSH, 2013. NIOSH Pocket Guide to Chemical Hazards. US Pelikan, Z., Cerny, E., 1968. Arch. Toxikol. 23, 283–292.
­Department of Health and Human Services. National Institute Penalver, R., Campillo, N., Hernandez-Cordoba, M., 2011. Talanta
for Occupational Safety and Health, (Publication no 97–140). 87, 268–275.
http://www.cdc.gov/niosh/npg/ (accessed 29.11.13). Penninks, A.H., 1993. Food Addit. Contam. 10, 351–361.
Nishida, H., Matsui, H., Sugiura, H., et al., 1990. J. Pharmacobiodyn. Penninks, A.H., Seinen, W., 1985. Toxicology 37, 285–295.
13, 543–548. Penninks, A.H., Verschuren, P.M., Seinen, W., 1983. Toxicol. Appl.
Noda, T., Nakamura, T., Shimizu, M., et al., 1992a. B Environ. Con- Pharmacol. 70, 115–120.
tam. Tox. 49, 715–722. Penninks, A., Kuper, F., Spit, B.J., Seinen, W., 1985. Immunopharma-
Noda, T., Yamano, T., Shimizu, M., et al., 1992b. Arch. Environ. Con- col 10, 1–10.
tam. Toxicol. 23, 216–222. Penninks, A.H., Hilgers, L., Seinen, W., 1987. Toxicology 44,
Noda, T., Morita, S., Baba, A., 1993. Toxicology 85, 149–160. 107–120.
Noda, T., Morita, S., Baba, A., 1994. Food Chem. Toxicol. 32, 321–327. Perring, L., Basic-Dvorzak, M., 2002. Anal. Bioanal. Chem. 374,
Noel, L., Leblanc, J.C., Guerin, T., 2003. Food Addit. Contam. 20, 235–243.
44–56. Pettine, M., Millero, F.J., Macchi, G., 1981. Anal. Chem. 53, 1039–1043.
Nsengimana, H., Cukrowska, E.M., Dinsmore, A., et al., 2009. J. Sep. Pieters, R.H.H., Bol, M., Penninks, A.H., 1994a. Toxicology 91,
Sci. 32, 2426–2433. 189–202.
56 Tin 1283

Pieters, R.H.H., Bol, M., Seinen, W., et al., 1994b. Hum. Exp. Toxicol. Sanderson, J.T., Boerma, J., Lansbergen, G.W., et al., 2002. Toxicol.
13, 876–879. Appl. Pharmacol. 182, 44–54.
Pinel-Raffaitin, P., Rodriguez-Gonzalez, P., Ponthieu, M., et al., 2007. Sano, T., Takagi, H., Nagano, K., et al., 2010. J. Chromat. A 1217,
J. Anal. At. Spectrom. 22, 258–266. 4344–4346.
Plumbing Manufacturers International, 2011. Reduction in lead bill Santos, M., Enes, P., Reis-Henriques, M., et al., 2009. Chemosphere
is signed into law by President Obama: Plumbing Manufacturers 75, 661–666.
International press release, January 6, p. 2. htttp://www.pmiho Santos, M.M., Castro, L.F.C., Vieira, M.N., et al., 2005. Comp.
me.org/i4a/pages/index.cfm?pageID=3504 (accessed 03.02.11). ­Biochem. Physiol.–C: Pharmacol. Toxicol. 141, 101–109.
Podratz, P.L., Filho, V.S.D., Lopes, P.F.I., et al., 2012. J. Toxicol. Env. Sarradin, P.M., Lapaquellerie, Y., Astruc, A., et al., 1995. Sci. Total
Heal. A 75, 1035–1046. ­Environ. 170, 59–70.
Poitou, P., Marignac, B., Certin, C., et al., 1978. Ann. Pharm. Fr. 36, Sayer, C.D., Hoare, D.J., Simpson, G.L., et al., 2006. Environ. Sci.
569–572. Technol. 40, 5269–5275.
Polster, M., Halacka, K., 1971. Ernährungsforschung 16, 527–535. Schafer Jr., E.W., Bowles Jr, W.A., 1985. Arch. Environ. Contam. Toxi-
Prior, C., 2010. Electroanalysis 22, 1446–1454. col. 14, 111–129.
Prough, R.A., Stalmach, M.A., Wiebkin, P., Bridges, J.W., 1981. Schafer, S.G., Femfert, U., 1984. Regul. Toxicol. Pharmacol. 4, 57–69.
­Biochem. J. 196, 763–770. Schering, A.G., 1992. Pharma Research Report No: IC-Kl 28. 19.8.
Prull, G., Rompel, K., 1970. Clin. Neurophysiol. 29, 215. Schlueter, N., Hardt, M., Lussi, A., et al., 2009. Eur. J. Oral Sci. 117,
Pungartnik, C., Viau, C., Picada, J., et al., 2005. Mutagenesis 583, 427–434.
146–157. Schlummer, M., Gruber, L., Maeurer, A., et al., 2007. Chemosphere
Puri, B.K., Munoz-Olivas, R., Camara, C., 2004. Spec. Acta B 59, 67, 1866–1876.
209–214. Schreck, E., Foucault, Y., Sarret, G., et al., 2012. Sci. Total Environ.
Quevauviller, P., Donard, O.F.X., 1990. Appl. Organomet. Chem. 4, 427, 253–262.
353–367. Schroeder, H.A., Balassa, J.J., Tipton, I.H., 1964. J. Chronic. Dis. 17,
Quevauviller, P., Lavigne, R., Pinel, R., et al., 1989. Environ. Pollut. 483–502.
57, 149–166. Schroeder, H.A., Kanisawa, M., Frost, D.V., et al., 1968. J. Nutr. 96,
Quevauviller, P., Martin, F., Belin, C., et al., 1993. Appl. Organomet. 37–45.
Chem. 7, 149–157. Schweinfurth, H., 1985. Tin Uses. Royal Society of Chemistry,
Qunfang, Z., Guibin, J., Zhongyang, L., et al., 2004. Food Addit. Con- London, 143, 9–12.
tam. 21, 1162–1167. Schweinfurth, H.A., Gunzel, P., 1987. Proceedings of the Oceans ‘87
Raffray, M., Cohen, G.M., 1991. Arch. Toxicol. 65, 135–139. Conference, Halifax, Nova Scotia.
Raffray, M., Mccarthy, D., Snowden, R.T., et al., 1993. Toxicol. Appl. Seinen, W., Willems, M.I., 1976. Toxicol. Appl. Pharmacol. 35, 63–75.
Pharmacol. 119, 122–130. Seinen, W., Vos, J.G., van Krieken, R., et al., 1977a. Toxicol. Appl.
Rahmi, D., Takasaki, Y., Zhu, Y., et al., 2010. Talanta 81, 1438–1445. Pharmacol. 42, 213–224.
Raj, A., Mayberry, J.F., Podas, T., 2003. Postgrad. Med. J. 79, 252– Seinen, W., Vos, J.G., van Spanje, I., et al., 1977b. Toxicol. Appl. Phar-
258. macol. 42, 197–212.
Rajendran, R.B., Tao, H., Nakazato, T., et al., 2000. Analyst 125, Seinen, W., Vos, J.G., Brands, R., et al., 1979. Immunopharmacol 1,
1757–1763. 343–355.
Rantakokko, P., Kuningas, T., Saastamoinen, K., et al., 2006. Food Senthilkumar, K., Kannan, K., Tanabe, S., et al., 1998. Fresen. Envi-
Addit. Contam. 23, 749–756. ron. Bull. 7, 561–571.
Rastkari, N., Mesdaghinia, A., Yunesian, M., et al., 2012. Bull. Senthilkumar, K., Kannan, K., Subramanian, A., et al., 2001. Environ.
­Environ. Contam. Toxicol. 88, 74–77. Sci. Poll. Res. 8, 35–47.
Reicks, M., Rader, J.I., 1990. Proc. Soc. Exp. Biol. Med. 195, 123–128. Shim, W.J., Jeon, J.K., Oh, J.R., Kim, N.S., Lee, S.H., 2002. Environ.
Reiter, L., Kidd, K., Heavner, G., et al., 1980. Neurotoxicology 2, Toxicol. Chem. 21, 1451–1455.
97–112. Shimasaki, Y., Kitano, T., Oshima, Y., et al., 2003. Environ. Toxicol.
Reuhl, K.R., Cranmer, J.M., 1984. Neurotoxicology 5, 187–204. Chem. 22, 141–144.
Rey, C., Reinecke, H.J., Besser, R., 1984. Vet. Hum. Toxicol. 26, Shimbo, S., Matsuda-Inoguchi, N., Watanabe, T., et al., 2007. Food
121–122. Add. Contam. 24, 535–545.
Rinehart, R.D., Yanagisawa, Y., 1993. Am. Ind. Hyg. Assoc. J. 54, Shuto, M., Higuchi, K., Sugiyama, C., et al., 2009. J. Pharm. Sci. 110,
593–599. 424–436.
Ritsema, R., de Smaele, T., Moens, L., et al., 1998. Environ. Pollut. Sieniawska, C.E., Jung, L.C., Olufadi, R., et al., 2012. Ann. Clin. Bio-
99, 271–277. chem. 49, 341–351.
Robertson, A.J., Rivers, D., Nagelschmidt, G., et al., 1961. Lancet 1, Silva, C.R., Oliveira, M.B.N., Melo, S.F., et al., 2002. Brain Res. Bull.
1089–1093. 59, 213–216.
Roe, F.J., Boyland, E., Millican, K., 1965. Food Cosmet. Toxicol. 3, Sisman, T., 2011. Environ. Toxicol. 26, 240–249.
277–280. Sluis-Cremer, G.K., Thomas, R.G., Goldstein, B., et al., 1989. S. Afr.
Roehl, C., Grell, M., Maser, E., 2009. Toxicol. in Vitro 23, 1541–1547. Med. J. 75, 124–126.
Rose, M., Baxter, M., Brereton, N., et al., 2010. Food Addit. Contam. Smialowicz, R.J., Riddle, M.M., Rogers, R.R., et al., 1988. J. Toxicol.
A 27, 1380–1404. Environ. Health 25, 403–422.
Rosenberg, D.W., Anderson, K.E., Kappas, A., 1984. Biochem. Bio- Smialowicz, R.J., Riddle, M.M., Rogers, R.R., et al., 1989. Toxicology
phys. Res. Commun. 119, 1022–1027. 57, 97–111.
Ross, W.D., Emmett, E.A., Steiner, J., et al., 1981. Am. J. Psychiat. 138, Smith, R., Bolam, S., Rees, H., et al., 2008. Environ. Monit. Assess.
1092–1095. 136, 245–256.
Saary, M.J., House, R.A., 2002. Occup. Med. (Lond) 52, 227–230. Snoeij, N.J., van Iersel, A.A., Penninks, A.H., et al., 1985. Toxicol.
Sadiki, A.I., Williams, D.T., 1999. Chemosphere 38, 1541–1548. Appl. Pharmacol. 81, 274–286.
1284 Elena A. Ostrakhovitch

Snoeij, N.J., Penninks, A.H., Seinen, W., 1987. Environ. Res. 44, Tsang, C.K., Lau, P.S., Tam, N.F.Y., et al., 1999. Environ. Pollut. 105,
335–353. 289–297.
Snoeij, N.J., Penninks, A.H., Seinen, W., 1988. Int. J. Immunopharma- Tsuda, T., Inoue, T., Kojima, M., et al., 1995. J. Aoac Int. 78, 941–
col. 10, 891–899. 943.
Sokas, R.K., 1998. Reproductive hazards of the workplace. John Wiley Tsunoda, M., Konno, N., Nakano, K., et al., 2004. Environ. Sci. 11,
and sons, Inc., New York. 154–156. 209–219.
Sole, M., Morcillo, Y., Porte, C., 1998. Environ. Pollut. 99, 241–246. Tzollas, N.M., Zachariadis, G.A., 2010. J. Sep. Sci. 33, 1610–1616.
Squibb, R.E., Carmichael, N.G., Tilson, H.A., 1980. Toxicol. Appl. Ueno, S., Susa, N., Furukawa, Y., et al., 1995. Arch. Toxicol. 69,
Pharmacol. 55, 188–197. 655–658.
Srivastava, S.C., Meinken, G.E., Richards, P., et al., 1985. Int. J. Nucl. Ueno, S., Kashimoto, T., Susa, N., et al., 2003. Arch. Toxicol. 77,
Med. Biol. 12 (3), 167–174. 173–181.
Stahnke, T., Richter-Landsberg, C., 2004. Glia 46, 334–344. Uhl, S., 1986. ETH Thesis No. 8014. Zurich, Maus Offsetdruck
Stange, B.J., Glanemann, M., Nuessler, N.C., et al., 2003. Liver Trans- Konstanz.
plant. 9, 612–620. Undap, S.L., Nirmala, K., Miki, S., et al., 2013a. J. Fac. Agric. Kyushu
Stange, D., Sieratowicz, A., Oehlmann, J., 2012. Aquat. Toxicol, Univ. 58, 131–135.
106–107, 20–4. Undap, S.L., Matsunaga, S., Honda, M., et al., 2013b. Ecotoxicol.
Stasinakis, A.S., Thomaidis, N.S., Nikolaou, A., et al., 2005. Environ. ­Environ. Saf. 96, 75–79.
Pollut. 134, 431–438. UNEP (The United Nations Environment Programme), 2006. Global
Stehrer, T., Heitz, J., Pedarnig, J., et al., 2010. Anal. Bioanal. Chem. Programme of Action for the Protection of the Marine Environ-
398, 415–424. ment from Land-based Activities (GPA).
Sternberg, R.M., Hotchkiss, A.K., Leblanc, G.A., 2008. Environ. Sci. Urushitani, H., Katsu, Y., Ohta, Y., et al., 2011. Aquat. Toxicol. 103,
Technol. 42, 1345–1351. 101–111.
Stewart, C., Demora, S.J., 1992. Appl. Organomet. Chem. 6, 507–512. USEPA (U.S. Environmental Protection Agency), 1994a.
Stewart, C., Thompson, J.A.J., 1997. Environ. Techn. 18, 1195–1202. EPA/600/R-94/111 Method 200.15, USEPA Office of Research and
Stone, O.J., Willis, C.J., 1968. Toxicol. Appl. Pharmacol. 13, 332–338. Development. http://water.epa.gov/scitech/methods/cwa/
Stringer, C.P., Hicks, R., Botham, P.A., 1991. Contact Dermatitis 24, bioindicators/upload/2007_07_10_methods_method_200_15.p
210–215. df (accessed 29.11.13).
Summer, K.H., Klein, D., Grem, H., 2003. National Chemicals Inspec- USEPA, 1994b. EPA/600/R-94/111 Method 200.2 USEPA Office of
torate (report No 600). Stockholm. Research and Development.
Sun, J., He, B., Yin, Y., et al., 2010. Anal. Methods 2, 2025–2031. USEPA, 1997. EPA—U.S. Environmental Protection Agency. Stan-
Sun, J., He, B., Liu, Q., et al., 2012. Talanta 93, 239–244. dard Methods for the Examination of Water and Wastewater,
Suzuki, Y., Endo, Y., Ogawa, M., et al., 2008. J. Chromat. B, Analyt. nineteenth ed.
Technol. Biomed. Life Sci. 868, 116–119. USGS (U.S. Geological Survey), 2010. Commodity Statistics and
Swennen, C., Ruttanadakul, N., Ardseungnern, S., et al., 1997. Envi- ­Information. American Metal Market 118, 9.
ron. Technol. 18, 1245–1254. Usta, J., Griffiths, D.E., 1992. Biochem. Biophys. Res. Commun. 188,
Takahashi, S., Mukai, H., Tanabe, S., et al., 1999. Environ. Pollut. 106, 365–371.
213–218. Uveges, M., Abranko, L., Fodor, P., 2007. Talanta 73, 490–497.
Tang, X., Li, N., Kang, L., et al., 2013a. Occup. Environ. Med. 70 (8), Valberg, L.S., Flanagan, P.R., Chamberlain, M.J., 1984. Am. J. Clin.
561–567. Nutr. 40, 536–541.
Tang, X., Wu, X., Dubois, A.M., et al., 2013b. Bull. Environ. Contam. van Kol, S.W., Hendriksen, P.J., van Loveren, H., et al., 2012. Toxicol-
Toxicol. 90 (5), 626–633. ogy 296, 37–47.
Tas, J.W., Opperhuizen, A., Seinen, W., 1990. Toxicol. Environ. Chem. Viau, C., Guecheva, T.N., Sousa, F., et al., 2009. Arch. Toxicol. 83,
28, 129–141. 769–775.
Tennekes, H., Horst, K., Luetkemeier, H., et al., 1989. Unpublished Viviani, B., Rossi, A.D., Chow, S.C., et al., 1995. Neurotoxicology 16,
report 046980 (A40468) of RCC Research and Consulting Com- 19–25.
pany AG, Itingen. Submitted to WHO by Hoechst AG, Frankfurt- Volti, G.L., Zappala, A., Leggio, G.M., et al., 2011. Neurosci. Lett. 492,
am-Main. 33–38.
Teraoka, H., 1981. Arch. Environ. Health 36, 155–165. Vos, J.G., Krajnc, E.I., 1983. Dev. Toxicol. Environ. Sci. 11, 229–240.
Tipton, I.H., Cook, M., 1963. Health Phys. 9, 103–145. Vos, J.G., de Klerk, A., Krajnc, E.I., et al., 1984. Toxicol. Appl. Phar-
Titley-O’Neal, C..P., Spade, D.J., Zhang, Y., et al., 2013. Sci. Total macol. 75, 387–408.
­Environ. 449, 52–62. Vos, J.G., Deklerk, A., Krajnc, E.I., et al., 1990. Toxicol. Appl. Pharma-
Toniolo, R., Pizzariello, A., Tubaro, F., et al., 2009. J. Appl. Electro- col. 105, 144–155.
chem. 39, 979–988. Wahlen, R., Catterick, T., 2003. J. Chromat. B, Analyt. Technol.
Tonk, E., Verhoef, A., De la Fonteyne, L., et al., 2011a. Birth Defects. Biomed. Life Sci. 783, 221–229.
Res. A 91, 343. Walker, C.H., Hopkin, S.P., Sibly, R.M., et al., 2006. Principles of
Tonk, E.C., de Groot, D.M., Penninks, A.H., et al., 2011b. Toxicol. ­Ecotoxicology. Taylor & Francis Group, Boca Raton, USA.
Lett. 204, 156–163. Wang, X., Jin, H., Ding, L., et al., 2008. Talanta 75, 556–563.
Tonk, E.C., Verhoef, A., de la Fonteyne, L.J., et al., 2011c. Reprod. Wang, X., Fang, C., Hong, H., et al., 2010. Aquat. Toxicol. 99, 413–422.
Toxicol. 32, 341–348. Warburton, S., Udler, W., Ewert, R.M., et al., 1962. Public Health Rep.
Toyoda, M., Sakai, H., Kobayashi, Y., et al., 2000. J. Food Hygienic 77, 798–800.
Soc. Japan 41, 280–286. Ward, R.J., 2003. Central Toxicology Laboratory, January (report No.
Truhaut, R., Chauvel, Y., Anger, J.P., et al., 1976. Eur. J. Toxicol. Envi- CTL/JV 1700) Report No CTL/JV 1700.
ron. Hyg. 9, 31–40. Watermann, B., Grote, K., Gnass, K., et al., 2008. Exp. Toxicol. Pathol.
Tryphonas, H., Cooke, G., Caldwell, D., et al., 2004. Food Chem. 60, 313–321.
Toxicol. 42, 221–235. Wax, P.M., Dockstader, L., 1995. J. Toxicol. Clin. Toxicol. 33, 239–241.
56 Tin 1285

Weidenhaupt, A., Arnold, C., Muller, S.R., et al., 1997. Environ. Sci. Yamaguchi, M., Sugii, K., Okada, S., 1982. Toxicol. Lett. 10, 7–10.
Technol. 31, 2603–2609. Yamashita, Y., Taketomi, A., Fukuzawa, K., et al., 2007. Am. J. Surg.
Wester, P.W., Krajnc, E.I., van Leeuwen, F.X., et al., 1990. Food Chem. 193, 454–459.
Toxicol. 28, 179–196. Yang, G., Wang, Y., Qi, F., 2012. Microchimica Acta 177, 365–372.
Whalen, M.M., Loganathan, B.G., Kannan, K., 1999. Environ. Res. Yilmaz, A., Gocmen Ocal, S., Doruk, S., et al., 2009. Tuberkuloz ve
81, 108–116. toraks 57, 422–426.
Whittington, D.L., Woodruff, M.L., Baisden, R.H., 1989. Neurotoxi- Yokoi, K., Kimura, M., Itokawa, Y., 1990a. Anal. Biochem. 190,
col. Teratol. 11, 21–33. 71–77.
WHO (World Health Organization), 1980a. World health organiza- Yokoi, K., Kimura, M., Itokawa, Y., 1990b. Biol. Trace Elem. Res. 24,
tion: International program on Chemical Safety (WHO/IIPCS). 223–231.
Environ. Health criteria 15. Yoo, C.I., Kim, Y., Jeong, K.S., et al., 2007. J. Occup. Health 49,
WHO, 1980b. World Health Organization. International Programme 305–310.
on Chemical Safety (WHO/IPCS). Tin and organotin com- Yoshizuka, M., Hara, K., Haramaki, N., et al., 1992. Arch. Toxicol.
pounds. A preliminary review. 66, 182–187.
WHO, 1990. World Health Organization. International Programme Yousef, M.I., 2005. Toxicology 207, 81–89.
on Chemical Safety. Environ. Health Criteria 116. Yu, H., Chen, S., Yang, Z., et al., 2010. Cell. Biol. Int. 34, 99–108.
WHO, 1992. World Health Organization. FAO Panel of Experts on Yu, Z.H., Sun, J.Q., Jing, M., et al., 2010. Food Chem. 119, 364–367.
Pesticide Residues in Food and the Environment and the WHO Yu, Z.P., Matsuoka, M., Wispriyono, B., et al., 2000. Toxicol. Appl.
Expert Group on Pesticide Residues. Pharmacol. 168, 200–207.
WHO, 1999. World Health Organization. Concise international Zachariadis, G., Rosenberg, E., 2009a. Talanta 78, 570–576.
chemical assessment document 14. Zachariadis, G., Rosenberg, E., 2009b. J. Chromat. B, Analyt. Technol.
WHO, 2001. World Health Organization. Food additives series 46. Biomed. Life Sci. 877, 1140–1144.
WHO, 2003. World Health Organization. Committee on Updat- Zanon, F., Rado, N., Centanni, E., Zharova, N., et al., 2009. Environ.
ing of Occupational Exposure Limits. Health-based Reassess- Monit. Assess. 152, 35–45.
ment of Administrative Occupational Exposure Limits. No. Zeman, W., Gadermann, E., Hardebeck, K., 1951. Deutsches Archiv
2000/15OSH/091, The Hague. fur Klinische Medizin 198, 713–721.
WHO, 2006. World Health Organization. Report Series 930. Zhai, G., Liu, J., Li, L., et al., 2009. Talanta 77, 1273–1278.
Whyte, A.L., Hook, G., Greening, G.E., et al., 2009. Sci. Total Environ. Zhang, S.K., Tsui, N.C., Li, D.H., et al., 2010. Pancreas 39, 252–253.
407, 4348–4355. Zhang, Z.H., Dong, B.B., Zang, S.L., et al., 2006. Acta Chim. Sin. 64,
Winship, K.A., 1988. Adverse Drug React. Acute. Poisoning. Rev. 7, 953–958.
19–38. Zhu, X., Xing, M., Lou, J., et al., 2007. Toxicology 230, 45–52.
Yamada, J., Inoue, K., Furukawa, T., et al., 2010. Toxicol. Lett. 198, Zuliani, T., Milacic, R., Scancar, J., 2012. Anal. Bioanal. Chem. 403,
282–288. 857–865.
Yamaguchi, M., Okada, S., 1980. Toxicol. Lett. 6, 177–180. Zuo, Z., Chen, S., Wu, T., et al., 2011. Environ. Toxicol. 26, 79–85.
Yamaguchi, M., Saito, R., Okada, S., 1980. Toxicology 16, 267–273.

Potrebbero piacerti anche